1
|
Jo D, Choi SY, Ahn SY, Song J. IGF1 enhances memory function in obese mice and stabilizes the neural structure under insulin resistance via AKT-GSK3β-BDNF signaling. Biomed Pharmacother 2025; 183:117846. [PMID: 39805192 DOI: 10.1016/j.biopha.2025.117846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025] Open
Abstract
Obesity is a prevalent metabolic disorder linked to insulin resistance, hyperglycemia, increased adiposity, chronic inflammation, and cognitive dysfunction. Recent research has focused on developing therapeutic strategies to mitigate cognitive impairment associated with obesity. Insulin growth factor-1 (IGF1) deficiency is linked to insulin resistance, glucose intolerance, and the progression of obesity-related central nervous system (CNS) disorders. In this study, we investigated the neuroprotective effects of IGF1 in two obesity models: diet-induced obesity (high-fat diet mice) and genetic obesity (ob/ob mice which is genetically deficient in leptin), and in vitro Neuro2A neuronal cells and primary cortical neurons under insulin resistance conditions. We performed RNA sequencing analysis using the cortex of high-fat diet mice injected with IGF1. Also, we detected cytokine levels in blood of high-fat diet mice injected with IGF1. In addition, we conducted the Barnes maze test as a spatial memory function test and open field test as an anxiety behavior test in ob/ob mice. We measured the levels of proteins and mRNAs related to insulin signaling, including synaptic density proteins in brain cortex of ob/ob mice. Our results showed that IGF1 injection enhanced spatial memory function and synaptic plasticity in obese mice. Furthermore, in vitro data demonstrated that IGF1 treated neurons revealed enhanced neural complexity and improved neurite outgrowth under insulin resistance condition through the AKT-GSK3β-BDNF pathway related to antidepressant, cognitive function and anti-apoptotic mechanisms. Therefore, our results provided that IGF1 have potential to alleviate cognitive impairment by promoting synaptic plasticity and neural complexity in the obese brain.
Collapse
Affiliation(s)
- Danbi Jo
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea.
| | - Seo Yoon Choi
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Republic of Korea.
| | - Seo Yeon Ahn
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Republic of Korea.
| |
Collapse
|
2
|
Kaur N, Aran KR. Uncovering the intricacies of IGF-1 in Alzheimer's disease: new insights from regulation to therapeutic targeting. Inflammopharmacology 2025:10.1007/s10787-025-01641-0. [PMID: 39883327 DOI: 10.1007/s10787-025-01641-0] [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: 12/29/2024] [Accepted: 01/12/2025] [Indexed: 01/31/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of amyloid-β plaques and tau tangles, leading to cognitive decline and dementia. Insulin-like Growth Factor-1 (IGF-1) is similar in structure to insulin and is crucial for cell growth, differentiation, and regulating oxidative stress, synaptic plasticity, and mitochondrial function. IGF-1 exerts its physiological effects by binding to the IGF-1 receptor (IGF-1R) and activating PI3K/Akt pathway. In addition to the physiological activities in the brain, numerous studies point to a potential protective role of the IGF-1 pathway in the pathogenesis of neurodegenerative diseases, such as AD. Interestingly, patients with AD often exhibit altered insulin and IGF-1 levels, along with an inadequate insulin response. Dysregulation of IGF-1 signaling contributes to hyperphosphorylation of tau, NFT accumulation, increased β- and γ-secretase activity, elevated Aβ production, and impaired Aβ clearance, highlighting the need to explore the role of this signaling for potential therapeutic targets of AD. This review explores the role of IGF signaling in AD pathology, highlighting IGF-1 as a promising therapeutic target due to its significant involvement in disease mechanisms. Modulating IGF-1 activity could help mitigate neurodegeneration and preserve cognitive function in AD. A comprehensive understanding of the mechanisms underlying IGF-1 dysregulation is crucial for developing targeted therapeutic strategies to address the complex and multifaceted nature of AD.
Collapse
Affiliation(s)
- Navpreet Kaur
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
3
|
Estrada JA, Hori A, Fukazawa A, Ishizawa R, Hotta N, Kim HK, Smith SA, Mizuno M. Abnormal cardiovascular control during exercise: Role of insulin resistance in the brain. Auton Neurosci 2025; 258:103239. [PMID: 39874739 DOI: 10.1016/j.autneu.2025.103239] [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: 11/01/2024] [Revised: 12/19/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025]
Abstract
During exercise circulatory adjustments to meet oxygen demands are mediated by multiple autonomic mechanisms, the skeletal muscle exercise pressor reflex (EPR), the baroreflex (BR), and by feedforward signals from central command neurons in higher brain centers. Insulin resistance in peripheral tissues includes sensitization of skeletal muscle afferents by hyperinsulinemia which is in part responsible for the abnormally heightened EPR function observed in diabetic animal models and patients. However, the role of insulin signaling within the central nervous system (CNS) is receiving increased attention as a potential therapeutic intervention in diseases with underlying insulin resistance. This review will highlight recent advances in our understanding of how insulin resistance induces changes in central signaling. The alterations in central insulin signaling produce aberrant cardiovascular responses to exercise. In particular, we will discuss the role of insulin signaling within the medullary cardiovascular control nuclei. The nucleus tractus solitarius (NTS) and rostral ventrolateral medulla (RVLM) are key nuclei where insulin has been demonstrated to modulate cardiovascular reflexes. The first locus of integration for the EPR, BR and central command is the NTS which is high in neurons expressing insulin receptors (IRs). The IRs on these neurons are well positioned to modulate cardiovascular responses to exercise. Additionally, the differences in IR density and presence of receptor isoforms enable specificity and diversity of insulin actions within the CNS. Therefore, non-invasive delivery of insulin into the CNS may be an effective means of normalizing cardiovascular responses to exercise in patients with insulin resistance.
Collapse
Affiliation(s)
- Juan A Estrada
- Departments of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Amane Hori
- Departments of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Japan Society for the Promotion of Science, Tokyo 102-0083, Japan; College of Life and Health Sciences, Chubu University, Kasugai 487-8501, Japan
| | - Ayumi Fukazawa
- Departments of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Japan Society for the Promotion of Science, Tokyo 102-0083, Japan
| | - Rie Ishizawa
- Faculty of Sports and Life Science, National Institute of Fitness and Sports in KANOYA, Kagoshima 891-2393, Japan
| | - Norio Hotta
- College of Life and Health Sciences, Chubu University, Kasugai 487-8501, Japan
| | - Han-Kyul Kim
- Departments of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Scott A Smith
- Departments of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Masaki Mizuno
- Departments of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
4
|
Cleary JA, Kumar A, Craft S, Deep G. Neuron-derived extracellular vesicles as a liquid biopsy for brain insulin dysregulation in Alzheimer's disease and related disorders. Alzheimers Dement 2025:e14497. [PMID: 39822132 DOI: 10.1002/alz.14497] [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: 08/20/2024] [Revised: 11/24/2024] [Accepted: 12/01/2024] [Indexed: 01/19/2025]
Abstract
Extracellular vesicles (EVs) have emerged as novel blood-based biomarkers for various pathologies. The development of methods to enrich cell-specific EVs from biofluids has enabled us to monitor difficult-to-access organs, such as the brain, in real time without disrupting their function, thus serving as liquid biopsy. Burgeoning evidence indicates that the contents of neuron-derived EVs (NDEs) in blood reveal dynamic alterations that occur during neurodegenerative pathogenesis, including Alzheimer's disease (AD), reflecting a disease-specific molecular signature. Among these AD-specific molecular changes is brain insulin-signaling dysregulation, which cannot be assessed clinically in a living patient and remains an unexplained co-occurrence during AD pathogenesis. This review is focused on delineating how NDEs in the blood may begin to close the gap between identifying molecular changes associated with brain insulin dysregulation reliably in living patients and its connection to AD. This approach could lead to the identification of novel early and less-invasive diagnostic molecular biomarkers for AD. HIGHLIGHTS: Neuron-derived extracellular vesicles (NDEs) could be isolated from peripheral blood. NDEs in blood reflect the molecular signature of Alzheimer's disease (AD). Brain insulin-signaling dysregulation plays a critical role in AD. NDEs in blood could predict brain insulin-signaling dysregulation. NDEs offer novel early and less-invasive diagnostic biomarkers for AD.
Collapse
Affiliation(s)
- Jacob Alexander Cleary
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Ashish Kumar
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Suzanne Craft
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Gagan Deep
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
5
|
Ni X, Zhang Y, Zhang W, Wen Y, Wang Y, Wang M. Association between the triglyceride glucose-body mass index and memory-related diseases: A nationwide cohort study. Public Health 2025; 239:136-141. [PMID: 39826497 DOI: 10.1016/j.puhe.2024.12.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/10/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
OBJECTIVES This study aimed to examine the longitudinal association between the triglyceride glucose-body mass index (TyG-BMI) index and memory-related diseases (MDs) among Chinese adults. STUDY DESIGN Nationwide cohort study. METHODS Data were obtained from the China Health and Retirement Longitudinal Study (CHARLS), which is an ongoing nationally representative prospective cohort study. The exposure was the participants' TyG-BMI index at baseline from 2011 to 2012. MDs were acquired by self-reporting questions in 2018. Cox proportional hazards regression models were conducted to assess the relationship between the TyG-BMI index and MDs. RESULTS During a median follow-up of 7.0 years, a total of 197 incident MDs events occurred. After multivariate adjustments for established MD-related risk factors, participants with the highest tertile of the TyG-BMI index had a higher risk of MDs (HR: 1.65; 95 % CI: 1.02-2.09; P-trend <0.001) relative to individuals with the lowest tertile. Furthermore, the subgroup analyses of the association remained consistent. CONCLUSION A high TyG-BMI index was associated with an increased risk of MDs. Our study suggests that monitoring long-term changes in the TyG-BMI index could assist with the early identification and prevention of MDs among individuals at high risk.
Collapse
Affiliation(s)
- Xiaoyan Ni
- Department of Pediatrics, The People's Hospital of Chongqing Liang Jiang New Area, Chongqing, 401121, China
| | - Yiwei Zhang
- Department of Pediatrics, The People's Hospital of Chongqing Liang Jiang New Area, Chongqing, 401121, China
| | - Weixian Zhang
- Department of Pediatrics, The People's Hospital of Chongqing Liang Jiang New Area, Chongqing, 401121, China
| | - Yi Wen
- Department of Pediatrics, The People's Hospital of Chongqing Liang Jiang New Area, Chongqing, 401121, China
| | - Yujing Wang
- Department of Pediatrics, The People's Hospital of Chongqing Liang Jiang New Area, Chongqing, 401121, China
| | - Mingmin Wang
- Department of Pediatrics, The People's Hospital of Chongqing Liang Jiang New Area, Chongqing, 401121, China.
| |
Collapse
|
6
|
Jamadar SD, Behler A, Deery H, Breakspear M. The metabolic costs of cognition. Trends Cogn Sci 2025:S1364-6613(24)00319-X. [PMID: 39809687 DOI: 10.1016/j.tics.2024.11.010] [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: 07/22/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 01/16/2025]
Abstract
Cognition and behavior are emergent properties of brain systems that seek to maximize complex and adaptive behaviors while minimizing energy utilization. Different species reconcile this trade-off in different ways, but in humans the outcome is biased towards complex behaviors and hence relatively high energy use. However, even in energy-intensive brains, numerous parsimonious processes operate to optimize energy use. We review how this balance manifests in both homeostatic processes and task-associated cognition. We also consider the perturbations and disruptions of metabolism in neurocognitive diseases.
Collapse
Affiliation(s)
- Sharna D Jamadar
- School of Psychological Sciences, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Victoria, Australia; Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia.
| | - Anna Behler
- School of Psychological Sciences, College of Engineering, Science, and the Environment, University of Newcastle, Newcastle, New South Wales, Australia
| | - Hamish Deery
- School of Psychological Sciences, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Victoria, Australia; Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| | - Michael Breakspear
- School of Psychological Sciences, College of Engineering, Science, and the Environment, University of Newcastle, Newcastle, New South Wales, Australia; School of Public Health and Medicine, College of Medicine, Health and Wellbeing, University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
7
|
Ding C, Lu R, Kong Z, Huang R. Exploring the triglyceride-glucose index's role in depression and cognitive dysfunction: Evidence from NHANES with machine learning support. J Affect Disord 2025; 374:282-289. [PMID: 39805501 DOI: 10.1016/j.jad.2025.01.051] [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: 03/22/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
BACKGROUND Depression and cognitive impairments are prevalent among older adults, with evidence suggesting potential links to obesity and lipid metabolism disturbances. This study investigates the relationships between the triglyceride-glucose (TyG) index, body mass index (BMI), depression, and cognitive dysfunction in older adults, leveraging data from the NHANES survey and employing machine learning techniques. METHODS We analysed 1352 participants aged 60-79 from the 2011-2014 NHANES dataset, who underwent cognitive function testing, depression assessments, and TyG index measurements. Multivariate linear regression and subgroup analyses were conducted to examine associations between the TyG index and depression/cognitive impairment. Machine learning models evaluated the importance of predictive factors for depression, while Mendelian randomization (MR) was employed to explore the causal relationship between BMI and depression/cognitive function. RESULTS The TyG index was negatively associated with cognitive function scores and positively associated with depression scores in adjusted models (p < 0.001). In fully adjusted subgroup analyses, among obese individuals (BMI ≥ 28), a 100-unit increase in the TyG index was linked to a 3.79-point decrease in depression scores. Machine learning models (Xgboost, AUC = 0.960) identified BMI, TyG-BMI, gender, and comorbidities (e.g., asthma, stroke, emphysema) as key determinants of depression. MR analyses revealed a negative association between BMI and depression risk [OR: 0.9934; 95 % CI (0.9901-0.9968), p = 0.0001] and cognitive dysfunction risk [OR: 0.8514; 95 % CI (0.7929-0.9143), p < 0.05]. No evidence of heterogeneity or pleiotropy was detected. LIMITATIONS Depression and cognitive impairments were self-reported, potentially introducing bias. The observed associations may be influenced by unmeasured confounders, necessitating further research into the underlying mechanisms. CONCLUSIONS Our findings reveal associations between the TyG index and psychocognitive health in older adults. While these results highlight lipid metabolism as a potential factor in depression and cognitive dysfunction, further studies are needed to validate these findings and explore underlying mechanisms.
Collapse
Affiliation(s)
- Chao Ding
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Renjie Lu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhiyu Kong
- South China University of Technology, Guangzhou, China
| | - Rong Huang
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
8
|
Wang P, Wang M, Xie Z, Zhi Z, Wang Y, Liu F, Liu Y, Zhao L. Association Between Four Non-Insulin-Based Insulin Resistance Indices and the Risk of Post-Stroke Depression. Clin Interv Aging 2025; 20:19-31. [PMID: 39817260 PMCID: PMC11733171 DOI: 10.2147/cia.s501569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/30/2024] [Indexed: 01/18/2025] Open
Abstract
Purpose Research suggests that insulin resistance (IR) is associated with acute ischemic stroke (AIS) and depression. The use of insulin-based IR assessments is complicated. Therefore, we explored the relationship between four non-insulin-based IR indices and post-stroke depression (PSD). Patients and Methods A total of 638 consecutive AIS patients were enrolled in this prospective cohort study. Clinical data were collected to compute indices such as the triglyceride glucose (TyG) index, triglyceride glucose-body mass index (TyG-BMI), insulin resistance metabolic score (METS-IR), and triglyceride/high-density lipoprotein cholesterol ratio (TG/HDL-C). One month post-stroke, neuropsychological assessments were conducted using the 17-item Hamilton Depression Scale. Binary logistic regression analysis was performed to explore the relationship between the four non-insulin-based IR indices and PSD. Results Ultimately, 381 patients completed the 1-month follow-up, including 112 (29.4%) with PSD. The PSD group exhibited significantly higher levels of the four IR indices compared to the non-PSD group. Logistic regression analysis demonstrated that these indicators were independently associated with PSD occurrence, both before and after adjusting for potential confounders (all P < 0.001). Tertile analyses indicated that the highest tertile group had a greater risk of PSD occurrence than the lowest tertile group for four IR indicators, even after adjusting for potential confounders (all P < 0.05). Restricted cubic spline analysis revealed a linear dose-response relationship between the four IR indices and PSD. In the subgroup analysis, only the TyG index showed a significant interaction with diabetes (P for interaction = 0.014). The area under curve values for the TyG index, TyG-BMI, METS-IR, and TG/HDL-C were 0.700, 0.721, 0.711, and 0.690, respectively. Conclusion High TyG index, TyG-BMI, METS-IR, and TG/HDL-C at baseline were independent risk factors for PSD in AIS. Each of these indicators exhibits predictive value for PSD occurrence, aiding in the early identification of high-risk groups.
Collapse
Affiliation(s)
- Ping Wang
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Mengchao Wang
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Zhe Xie
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Zhongwen Zhi
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Yuqian Wang
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Fan Liu
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Yufeng Liu
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Liandong Zhao
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| |
Collapse
|
9
|
Martins Dos Santos K, Saunders SE, Antunes VR, Boychuk CR. Insulin activates parasympathetic hepatic-related neurons of the paraventricular nucleus of the hypothalamus through mTOR signaling. J Neurophysiol 2025; 133:320-332. [PMID: 39665212 DOI: 10.1152/jn.00284.2024] [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: 07/01/2024] [Revised: 11/05/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024] Open
Abstract
Integration of autonomic and metabolic regulation, including hepatic function, is a critical role played by the brain's hypothalamic region. Specifically, the paraventricular nucleus of the hypothalamus (PVN) regulates autonomic functions related to metabolism, such as hepatic glucose production. Although insulin can act directly on hepatic tissue to inhibit hepatic glucose production, recent evidence implicates that central actions of insulin within PVN also regulate glucose metabolism. However, specific central circuits responsible for insulin signaling with relation to hepatic regulation are poorly understood. As a heterogeneous nucleus essential to controlling parasympathetic motor output with notable expression of insulin receptors, PVN is an appealing target for insulin-dependent modulation of parasympathetic activity. Here, we tested the hypothesis that insulin activates hepatic-related PVN (PVNhepatic) neurons through a parasympathetic pathway. Using transsynaptic retrograde tracing, labeling within PVN was first identified 24 h after its expression in the dorsal motor nucleus of the vagus (DMV) and 72 h after hepatic injection. Critically, nearly all labeling in medial PVN was abolished after a left vagotomy, indicating that PVNhepatic neurons in this region are part of a central circuit innervating parasympathetic motor neurons. Insulin also significantly increased the firing frequency of PVNhepatic neurons in this subregion. Mechanistically, rapamycin pretreatment inhibited insulin-dependent activation of PVNhepatic neurons. Therefore, central insulin signaling can activate a subset of PVNhepatic neurons that are part of a unique parasympathetic network in control of hepatic function. Taken together, PVNhepatic neurons related to parasympathetic output regulation could serve as a key central network in insulin's ability to control hepatic functions.NEW & NOTEWORTHY Increased peripheral insulin concentrations are known to decrease hepatic glucose production through both direct actions on hepatocytes and central autonomic networks. Despite this understanding, how (and in which brain regions) insulin exerts its action is still obscure. Here, we demonstrate that insulin activates parasympathetic hepatic-related PVN neurons (PVNhepatic) and that this effect relies on mammalian target of rapamycin (mTOR) signaling, suggesting that insulin modulates hepatic function through autonomic pathways involving insulin receptor intracellular signaling cascades.
Collapse
Affiliation(s)
- Karoline Martins Dos Santos
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, United States
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sandy E Saunders
- Dalton Cardiovascular Research Center, Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
| | - Vagner R Antunes
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carie R Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, United States
- Dalton Cardiovascular Research Center, Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
10
|
Snyder JR, Ahmed M, Bhave S, Hotta R, Koppes RA, Goldstein AM, Koppes AN. Enteroendocrine Cells Sense Sucrose and Alter Enteric Neuron Excitability via Insulin Signaling. Adv Biol (Weinh) 2024:e2300566. [PMID: 39703141 DOI: 10.1002/adbi.202300566] [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: 07/19/2024] [Revised: 11/25/2024] [Indexed: 12/21/2024]
Abstract
Neurosensory circuits of the gastrointestinal tract sense microbial and nutrient changes in the gut; however, studying these circuits in vivo is hindered by invasive techniques and ethical concerns. Here, an in vitro model of enteroendocrine cells (EECs) and calcium reporting enteric neurons (ENs) is established and validated for functional signaling. Both mechanical and sucrose stimulation of co-cultures increased the percentage of neurons undergoing a calcium flux, indicating an action potential. Neuronal activation is blocked with either a piezo or insulin receptor blocker. At baseline, a flow only stimulus elicited 51.9% of neurons to activate in co-culture, which is decreased to 15.1% with a piezo blocker. Piezo blocked and sucrose stimulated EECs increased neuronal activation to 43.9%, and an insulin blocker reduced response to 12.4%. Since a cell line is used to model the EEC in the previous experiments, primary rat duodenal epithelium enriched for EECs are also stimulated and found to produced measurable insulin. This work shows the ability of EECs to produce insulin and for ENs to sense insulin. These results inspire further work on how insulin production outside the pancreas effects diabetes, insulin as a neurotransmitter, and exploration of additional nutritional and microbiotic stimuli on enteroendocrine-to-neuronal signaling.
Collapse
Affiliation(s)
- Jessica R Snyder
- Department of Bioengineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Minhal Ahmed
- Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Sukhada Bhave
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, 185 Cambridge St, CPZN 6-215, Boston, MA, 02114, USA
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, 185 Cambridge St, CPZN 6-215, Boston, MA, 02114, USA
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, 185 Cambridge St, CPZN 6-215, Boston, MA, 02114, USA
| | - Abigail N Koppes
- Department of Bioengineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
- Department of Biology, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| |
Collapse
|
11
|
Singh R, Jain S, Paliwal V, Verma K, Paliwal S, Sharma S. Does Metabolic Manager Show Encouraging Outcomes in Alzheimer's?: Challenges and Opportunity for Protein Tyrosine Phosphatase 1b Inhibitors. Drug Dev Res 2024; 85:e70026. [PMID: 39655712 DOI: 10.1002/ddr.70026] [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: 05/20/2024] [Revised: 10/22/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024]
Abstract
Protein tyrosine phosphatase 1b (PTP1b) is a member of the protein tyrosine phosphatase (PTP) enzyme group and encoded as PTP1N gene. Studies have evidenced an overexpression of the PTP1b enzyme in metabolic syndrome, anxiety, schizophrenia, neurodegeneration, and neuroinflammation. PTP1b inhibitor negatively regulates insulin and leptin pathways and has been explored as an antidiabetic agent in various clinical trials. Notably, the preclinical studies have shown that recuperating metabolic dysfunction and dyshomeostasis can reverse cognition and could be a possible approach to mitigate multifaceted Alzheimer's disease (AD). PTP1b inhibitor thus has attracted attention in neuroscience, though the development is limited to the preclinical stage, and its exploration in large clinical trials is warranted. This review provides an insight on the development of PTP1b inhibitors from different sources in diabesity. The crosstalk between metabolic dysfunction and insulin insensitivity in AD and type-2 diabetes has also been highlighted. Furthermore, this review presents the significance of PTP1b inhibition in AD based on pathophysiological facets, and recent evidences from preclinical and clinical studies.
Collapse
Affiliation(s)
- Ritu Singh
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Smita Jain
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Vartika Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Kanika Verma
- Department of Internal Medicine, Division of Cardiology, LSU Health Sciences Center Shreveport, Louisiana, USA
| | - Sarvesh Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| |
Collapse
|
12
|
Ulke J, Chopra S, Kadiri OL, Geserick P, Stein V, Cheshmeh S, Kleinridders A, Kappert K. PTPRJ is a negative regulator of insulin signaling in neuronal cells, impacting protein biosynthesis, and neurite outgrowth. J Neuroendocrinol 2024; 36:e13446. [PMID: 39253900 PMCID: PMC11646663 DOI: 10.1111/jne.13446] [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: 03/21/2024] [Revised: 07/29/2024] [Accepted: 08/28/2024] [Indexed: 09/11/2024]
Abstract
Central insulin resistance has been linked to the development of neurodegenerative diseases and mood disorders. Various proteins belonging to the enzyme family of protein tyrosine phosphatases (PTPs) act as inhibitors of insulin signaling. Protein tyrosine phosphatase receptor type J (PTPRJ) has been identified as a negative regulator in insulin signaling in the periphery. However, the impact of PTPRJ on insulin signaling and its functional role in neuronal cells is largely unknown. Therefore, we generated a Ptprj knockout (KO) cell model in the murine neuroblast cell line Neuro2a by CRISPR-Cas9 gene editing. Ptprj KO cells displayed enhanced insulin signaling, as shown by increased phosphorylation of the insulin receptor (INSR), IRS-1, AKT, and ERK1/2. Further, proximity ligation assays (PLA) revealed both direct interaction of PTPRJ with the INSR and recruitment of this phosphatase to the receptor upon insulin stimulation. By RNA sequencing gene expression analysis, we identified multiple gene clusters responsible for glucose uptake and metabolism, and genes involved in the synthesis of various lipids being mainly upregulated under PTPRJ deficiency. Furthermore, multiple Ca2+ transporters were differentially expressed along with decreased protein biosynthesis. This was accompanied by an increase in endoplasmic reticulum (ER) stress markers. On a functional level, PTPRJ deficiency compromised cell differentiation and neurite outgrowth, suggesting a role in nervous system development. Taken together, PTPRJ emerges as a negative regulator of central insulin signaling, impacting neuronal metabolism and neurite outgrowth.
Collapse
Affiliation(s)
- Jannis Ulke
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and PathobiochemistryBerlinGermany
- Max Rubner Center (MRC) for Cardiovascular Metabolic Renal ResearchCharité—Universitätsmedizin BerlinBerlinGermany
| | - Simran Chopra
- Department of Molecular and Experimental Nutritional Medicine, Institute of Nutritional ScienceUniversity of PotsdamNuthetalGermany
| | - Otsuware Linda‐Josephine Kadiri
- Department of Molecular and Experimental Nutritional Medicine, Institute of Nutritional ScienceUniversity of PotsdamNuthetalGermany
| | - Peter Geserick
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and PathobiochemistryBerlinGermany
- Max Rubner Center (MRC) for Cardiovascular Metabolic Renal ResearchCharité—Universitätsmedizin BerlinBerlinGermany
| | - Vanessa Stein
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and PathobiochemistryBerlinGermany
- Max Rubner Center (MRC) for Cardiovascular Metabolic Renal ResearchCharité—Universitätsmedizin BerlinBerlinGermany
| | - Sahar Cheshmeh
- Department of Molecular and Experimental Nutritional Medicine, Institute of Nutritional ScienceUniversity of PotsdamNuthetalGermany
| | - André Kleinridders
- Department of Molecular and Experimental Nutritional Medicine, Institute of Nutritional ScienceUniversity of PotsdamNuthetalGermany
| | - Kai Kappert
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and PathobiochemistryBerlinGermany
- Max Rubner Center (MRC) for Cardiovascular Metabolic Renal ResearchCharité—Universitätsmedizin BerlinBerlinGermany
| |
Collapse
|
13
|
Haas SS, Abbasi F, Watson K, Robakis T, Myoraku A, Frangou S, Rasgon N. Metabolic Status Modulates Global and Local Brain Age Estimates in Overweight and Obese Adults. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024:S2451-9022(24)00354-9. [PMID: 39615789 DOI: 10.1016/j.bpsc.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND As people live longer, maintaining brain health becomes essential for extending health span and preserving independence. Brain degeneration and cognitive decline are major contributors to disability. In this study, we investigated how metabolic health influences the brain age gap estimate (brainAGE), which measures the difference between neuroimaging-predicted brain age and chronological age. METHODS K-means clustering was applied to fasting metabolic markers including insulin, glucose, leptin, cortisol, triglycerides, high-density lipoprotein cholesterol and low-density lipoprotein cholesterol, steady-state plasma glucose, and body mass index of 114 physically and cognitively healthy adults. The homeostatic model assessment for insulin resistance served as a reference. T1-weighted brain magnetic resonance imaging was used to calculate voxel-level and global brainAGE. Longitudinal data were available for 53 participants over a 3-year interval. RESULTS K-means clustering divided the sample into 2 groups, those with favorable (n = 58) and those with suboptimal (n = 56) metabolic health. The suboptimal group showed signs of insulin resistance and dyslipidemia (false discovery rate-corrected p < .05) and had older global brainAGE and local brainAGE, with deviations most prominent in cerebellar, ventromedial prefrontal, and medial temporal regions (familywise error-corrected p < .05). Longitudinal analysis revealed group differences but no significant time or interaction effects on brainAGE measures. CONCLUSIONS Suboptimal metabolic status is linked to accelerated brain aging, particularly in brain regions rich in insulin receptors. These findings highlight the importance of metabolic health in maintaining brain function and suggest that promoting metabolic well-being may help extend health span.
Collapse
Affiliation(s)
- Shalaila S Haas
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Fahim Abbasi
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California
| | - Kathleen Watson
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California
| | - Thalia Robakis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alison Myoraku
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California
| | - Sophia Frangou
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Djavad Mowafaghian Centre for Brain Health, Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Natalie Rasgon
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California.
| |
Collapse
|
14
|
Zheng M, Wang C, Hu M, Li Q, Li J, Quan S, Zhang X, Gu L. Research progress on the association of insulin resistance with type 2 diabetes mellitus and Alzheimer's disease. Metab Brain Dis 2024; 40:35. [PMID: 39570454 DOI: 10.1007/s11011-024-01472-y] [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: 03/24/2024] [Accepted: 09/09/2024] [Indexed: 11/22/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder that is characterized by insulin resistance and hyperglycemia. It is also known to be a risk factor for Alzheimer's disease (AD). Insulin plays a crucial role in regulating the body's metabolism and is responsible for activating the Phosphoinotide-3-Kinase (PI3K)/Protein Kinase B (Akt) signaling pathway. This pathway is activated when insulin binds to the insulin receptor on nerve cells, and it helps regulate the metabolism of glucose and lipids. Dysfunction in the insulin signaling pathway can lead to a decrease in brain insulin levels and insulin sensitivity, thereby inducing disruptions in insulin signal transduction and leading to disorders in brain energy metabolism. Moreover, these dysfunctions also contribute to the accumulation of β-amyloid (Aβ) deposition and the hyperphosphorylation of Tau protein, both of which are characteristic features of AD. Therefore, this article focuses on insulin resistance to reveal the complex mechanism between brain insulin resistance and AD occurrence in T2DM. On this basis, this article further summarizes the biological effects and mechanisms of antidiabetic drugs on the two diseases, aiming to provide new ideas for the discovery of drugs for the treatment of T2DM combined with AD.
Collapse
Affiliation(s)
- Miao Zheng
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, Zhejiang, P.R. China
| | - Can Wang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, Zhejiang, P.R. China
| | - Min Hu
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, Zhejiang, P.R. China
| | - Qin Li
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, Zhejiang, P.R. China
| | - Jinhua Li
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, Zhejiang, P.R. China
| | - Shengli Quan
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, Zhejiang, P.R. China
| | - Xinyue Zhang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, Zhejiang, P.R. China.
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy (Institute of Materia Medica), Hangzhou Medical College, No.182, Tianmushan road, Xihu District, Hangzhou, 310013, Zhejiang, P.R. China.
| | - Lili Gu
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, Zhejiang, P.R. China.
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy (Institute of Materia Medica), Hangzhou Medical College, No.182, Tianmushan road, Xihu District, Hangzhou, 310013, Zhejiang, P.R. China.
| |
Collapse
|
15
|
Ben Shaul T, Frenkel D, Gurevich T. The Interplay of Stress, Inflammation, and Metabolic Factors in the Course of Parkinson's Disease. Int J Mol Sci 2024; 25:12409. [PMID: 39596474 PMCID: PMC11594997 DOI: 10.3390/ijms252212409] [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: 09/29/2024] [Revised: 11/09/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative condition for which there are symptomatic treatments but no disease-modifying therapies (DMTs). Extensive research over the years has highlighted the need for a multi-target DMT approach in PD that recognizes the various risk factors and their intricate interplay in contributing to PD-related neurodegeneration. Widespread risk factors, such as emotional stress and metabolic factors, have increasingly become focal points of exploration. Our review aims to summarize interactions between emotional stress and selected key players in metabolism, such as insulin, as potential mechanisms underlying neurodegeneration in PD.
Collapse
Affiliation(s)
- Tal Ben Shaul
- Movement Disorders Center, Neurological Institute, Tel Aviv Medical Center, Tel Aviv 6423906, Israel;
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dan Frenkel
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Ramat-Aviv, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tanya Gurevich
- Movement Disorders Center, Neurological Institute, Tel Aviv Medical Center, Tel Aviv 6423906, Israel;
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
16
|
Alagiakrishnan K, Halverson T. Role of Peripheral and Central Insulin Resistance in Neuropsychiatric Disorders. J Clin Med 2024; 13:6607. [PMID: 39518747 PMCID: PMC11547162 DOI: 10.3390/jcm13216607] [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: 10/07/2024] [Revised: 10/27/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Insulin acts on different organs, including the brain, which helps it regulate energy metabolism. Insulin signaling plays an important role in the function of different cell types. In this review, we have summarized the key roles of insulin and insulin receptors in healthy brains and in different brain disorders. Insulin signaling, as well as insulin resistance (IR), is a major contributor in the regulation of mood, behavior, and cognition. Recent evidence showed that both peripheral and central insulin resistance play a role in the pathophysiology, clinical presentation, and management of neuropsychiatric disorders like Cognitive Impairment/Dementia, Depression, and Schizophrenia. Many human studies point out Insulin Resistance/Metabolic Syndrome can increase the risk of dementia especially Alzheimer's dementia (AD). IR has been shown to play a role in AD development but also in its progression. This review article discusses the pathophysiological pathways and mechanisms of insulin resistance in major neuropsychiatric disorders. The extent of insulin resistance can be quantified using IR biomarkers like insulin levels, HOMA-IR index, and Triglyceride glucose-body mass index (TyG-BMI) levels. IR has been shown to precede neurodegeneration. Human trials showed current treatment with certain antidiabetic drugs, as well as life style management, like weight loss and exercise for IR, have shown promise in the management of cognitive/neuropsychiatric disorders. This may pave the pathway to the development of new therapeutic approaches to these challenging disorders of dementia and psychiatric diseases. Recent clinical trials are showing some encouraging evidence for these pharmacological and nonpharmacological approaches for IR in psychiatric and cognitive disorders, even though more research is needed to apply this evidence into clinical practice. Early identification and management of IR may help as a strategy to potentially alter neuropsychiatric disorders onset as well as its progression.
Collapse
Affiliation(s)
| | - Tyler Halverson
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada;
| |
Collapse
|
17
|
Ruisch IH, Widomska J, De Witte W, Mota NR, Fanelli G, Van Gils V, Jansen WJ, Vos SJB, Fóthi A, Barta C, Berkel S, Alam KA, Martinez A, Haavik J, O'Leary A, Slattery D, Sullivan M, Glennon J, Buitelaar JK, Bralten J, Franke B, Poelmans G. Molecular landscape of the overlap between Alzheimer's disease and somatic insulin-related diseases. Alzheimers Res Ther 2024; 16:239. [PMID: 39465382 PMCID: PMC11514822 DOI: 10.1186/s13195-024-01609-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
Alzheimer's disease (AD) is a multifactorial disease with both genetic and environmental factors contributing to its etiology. Previous evidence has implicated disturbed insulin signaling as a key mechanism that plays a role in both neurodegenerative diseases such as AD and comorbid somatic diseases such as diabetes mellitus type 2 (DM2). In this study, we analysed available genome-wide association studies (GWASs) of AD and somatic insulin-related diseases and conditions (SID), i.e., DM2, metabolic syndrome and obesity, to identify genes associated with both AD and SID that could increase our insights into their molecular underpinnings. We then performed functional enrichment analyses of these genes. Subsequently, using (additional) GWAS data, we conducted shared genetic etiology analyses between AD and SID, on the one hand, and blood and cerebrospinal fluid (CSF) metabolite levels on the other hand. Further, integrating all these analysis results with elaborate literature searches, we built a molecular landscape of the overlap between AD and SID. From the landscape, multiple functional themes emerged, including insulin signaling, estrogen signaling, synaptic transmission, lipid metabolism and tau signaling. We also found shared genetic etiologies between AD/SID and the blood/CSF levels of multiple metabolites, pointing towards "energy metabolism" as a key metabolic pathway that is affected in both AD and SID. Lastly, the landscape provided leads for putative novel drug targets for AD (including MARK4, TMEM219, FKBP5, NDUFS3 and IL34) that could be further developed into new AD treatments.
Collapse
Affiliation(s)
- I Hyun Ruisch
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joanna Widomska
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ward De Witte
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nina R Mota
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Giuseppe Fanelli
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Veerle Van Gils
- Department of Psychiatry & Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Willemijn J Jansen
- Department of Psychiatry & Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Stephanie J B Vos
- Department of Psychiatry & Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Abel Fóthi
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Csaba Barta
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Simone Berkel
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Kazi A Alam
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Aurora Martinez
- Department of Biomedicine, University of Bergen, Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's Disease, University of Bergen, Neuro-SysMed Center, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Aet O'Leary
- Department of Psychiatry, University Hospital, Frankfurt, Germany
| | - David Slattery
- Department of Psychiatry, Psychosomatics and Psychotherapy, Goethe-Universität, Frankfurt, Germany
| | - Mairéad Sullivan
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Jeffrey Glennon
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Janita Bralten
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Barbara Franke
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Geert Poelmans
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands.
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
18
|
Pramanik R, Dey A, Chakrabarty AK, Banerjee D, Narwaria A, Sharma S, Rai RK, Katiyar CK, Dubey SK. Diabetes mellitus and Alzheimer's disease: Understanding disease mechanisms, their correlation, and promising dual activity of selected herbs. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118402. [PMID: 38821139 DOI: 10.1016/j.jep.2024.118402] [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: 08/11/2023] [Revised: 05/12/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE This review explores the link between Type 2 Diabetes Mellitus (T2DM) and diabetes-induced Alzheimer's disease (AD). It emphasizes the shared pathophysiological links and mechanisms between the two conditions, focusing on reduced insulin levels and receptors, impaired glucose metabolism, insulin resistance, mitochondrial dysfunction, and oxidative damage in AD-affected brains-paralleling aspects of T2DM. The review suggests AD as a "diabetes of the brain," supported by cognitive enhancement through antidiabetic interventions. It focuses on the traditionally used Indian herbs as a means to manage both conditions while addressing developmental challenges. AIM OF THE STUDY This study explores the DM-AD connection, reviewing medicinal herbs with protective potential for both ailments, considering traditional uses and developmental challenges. MATERIALS AND METHODS Studied research, reviews, and ethnobotanical and scientific data from electronic databases and traditional books. RESULTS The study analyzes the pathophysiological links between DM and AD, emphasizing their interconnected factors. Eight Ayurvedic plants with dual protective effects against T2DM and AD are thoroughly reviewed with preclinical/clinical evidence. Historical context, phytoconstituents, and traditional applications are explored. Innovative formulations using these plants are examined. Challenges stemming from phytoconstituents' physicochemical properties are highlighted, prompting novel formulation development, including nanotechnology-based delivery systems. The study uncovers obstacles in formulating treatments for these diseases. CONCLUSION The review showcases the dual potential of chosen medicinal herbs against both diseases, along with their traditional applications, endorsing their use. It addresses formulation obstacles, proposing innovative delivery technologies for herbal therapies, while acknowledging their constraints. The review suggests the need for heightened investment and research in this area.
Collapse
Affiliation(s)
- Rima Pramanik
- R&D Healthcare Division, Emami Ltd, 13, BT Road, Belgharia, Kolkata, 700056, India
| | - Anuradha Dey
- R&D Healthcare Division, Emami Ltd, 13, BT Road, Belgharia, Kolkata, 700056, India
| | | | - Dipankar Banerjee
- R&D Healthcare Division, Emami Ltd, 13, BT Road, Belgharia, Kolkata, 700056, India
| | - Avinash Narwaria
- R&D Healthcare Division, Emami Ltd, 13, BT Road, Belgharia, Kolkata, 700056, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Rajiva Kumar Rai
- R&D Healthcare Division, Emami Ltd, 13, BT Road, Belgharia, Kolkata, 700056, India
| | - Chandra Kant Katiyar
- R&D Healthcare Division, Emami Ltd, 13, BT Road, Belgharia, Kolkata, 700056, India
| | - Sunil Kumar Dubey
- R&D Healthcare Division, Emami Ltd, 13, BT Road, Belgharia, Kolkata, 700056, India.
| |
Collapse
|
19
|
Rhea EM, Babin A, Thomas P, Omer M, Weaver R, Hansen K, Banks WA, Talbot K. Brain uptake pharmacokinetics of albiglutide, dulaglutide, tirzepatide, and DA5-CH in the search for new treatments of Alzheimer's and Parkinson's diseases. Tissue Barriers 2024; 12:2292461. [PMID: 38095516 PMCID: PMC11583597 DOI: 10.1080/21688370.2023.2292461] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/29/2023] [Accepted: 12/02/2023] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND A number of peptide incretin receptor agonists (IRAs) show promise as therapeutics for Alzheimer's disease (AD) and Parkinson's disease (PD). Transport across the blood-brain barrier (BBB) is one way for IRAs to act directly within the brain. To determine which IRAs are high priority candidates for treating these disorders, we have studied their brain uptake pharmacokinetics. METHODS We quantitatively measure the ability of four IRAs to cross the BBB. We injected adult male CD-1 mice intravenously with 125I- or 14C-labeled albiglutide, dulaglutide, DA5-CH, or tirzepatide and used multiple-time regression analyses to measure brain kinetics up to 1 hour. For those IRAs failing to enter the brain 1 h after intravenous injection, we also investigated their ability to enter over a longer time frame (i.e., 6 h). RESULTS Albiglutide and dulaglutide had the fastest brain uptake rates within 1 hour. DA5-CH appears to enter the brain rapidly, reaching equilibrium quickly. Tirzepatide does not appear to cross the BBB within 1 h after iv injection but like albumin, did so slowly over 6 h, presumably via the extracellular pathways. CONCLUSIONS We find that IRAs can cross the BBB by two separate processes; one that is fast and one that is slow. Three of the four IRAs investigated here have fast rates of transport and should be taken into consideration for testing as AD and PD therapeutics as they would have the ability to act quickly and directly on the brain as a whole.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Alice Babin
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
| | - Peter Thomas
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
| | - Mohamed Omer
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
| | - Riley Weaver
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
| | - Kim Hansen
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
| | - William A Banks
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Konrad Talbot
- Departments of Neurosurgery, Pathology and Human Anatomy, and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
20
|
Radosinska D, Gaal Kovalcikova A, Gardlik R, Chomova M, Snurikova D, Radosinska J, Vrbjar N. Oxidative Stress Markers and Na,K-ATPase Enzyme Kinetics Are Altered in the Cerebellum of Zucker Diabetic Fatty fa/fa Rats: A Comparison with Lean fa/+ and Wistar Rats. BIOLOGY 2024; 13:759. [PMID: 39452068 PMCID: PMC11505095 DOI: 10.3390/biology13100759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024]
Abstract
Type 2 diabetes mellitus has been referred to as being closely related to oxidative stress, which may affect brain functions and brain glucose metabolism due to its high metabolic activity and lipid-rich content. Na,K-ATPase is an essential enzyme maintaining intracellular homeostasis, with properties that can sensitively mirror various pathophysiological conditions such as diabetes. The goal of this study was to determine oxidative stress markers as well as Na,K-ATPase activities in the cerebellum of Zucker diabetic fatty (ZDF) rats depending on diabetes severity. The following groups of male rats were used: Wistar, ZDF Lean (fa/+), and ZDF (fa/fa) rats, arbitrarily divided according to glycemia into ZDF obese (ZO, less severe diabetes) and ZDF diabetic (ZOD, advanced diabetes) groups. In addition to basic biometry and biochemistry, oxidative stress markers were assessed in plasma and cerebellar tissues. The Na, K-ATPase enzyme activity was measured at varying ATP substrate concentrations. The results indicate significant differences in basic biometric and biochemical parameters within all the studied groups. Furthermore, oxidative damage was greater in the cerebellum of both ZDF (fa/fa) groups compared with the controls. Interestingly, Na,K-ATPase enzyme activity was highest to lowest in the following order: ZOD > ZO > Wistar > ZDF lean rats. In conclusion, an increase in systemic oxidative stress resulting from diabetic conditions has a significant impact on the cerebellar tissue independently of diabetes severity. The increased cerebellar Na,K-ATPase activity may reflect compensatory mechanisms in aged ZDF (fa/fa) animals, rather than indicating cerebellar neurodegeneration: a phenomenon that warrants further investigation.
Collapse
Affiliation(s)
- Dominika Radosinska
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia;
| | - Alexandra Gaal Kovalcikova
- Department of Pediatrics, National Institute of Children’s Diseases, Faculty of Medicine, Comenius University in Bratislava, 833 40 Bratislava, Slovakia;
| | - Roman Gardlik
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia;
| | - Maria Chomova
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, 813 72 Bratislava, Slovakia;
| | - Denisa Snurikova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 841 04 Bratislava, Slovakia; (D.S.); (N.V.)
| | - Jana Radosinska
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, 811 08 Bratislava, Slovakia
| | - Norbert Vrbjar
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 841 04 Bratislava, Slovakia; (D.S.); (N.V.)
| |
Collapse
|
21
|
Di Domenico F, Lanzillotta C, Perluigi M. Redox imbalance and metabolic defects in the context of Alzheimer disease. FEBS Lett 2024; 598:2047-2066. [PMID: 38472147 DOI: 10.1002/1873-3468.14840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024]
Abstract
Redox reactions play a critical role for intracellular processes, including pathways involved in metabolism and signaling. Reactive oxygen species (ROS) act either as second messengers or generators of protein modifications, fundamental mechanisms for signal transduction. Disturbance of redox homeostasis is associated with many disorders. Among these, Alzheimer's disease is a neurodegenerative pathology that presents hallmarks of oxidative damage such as increased ROS production, decreased activity of antioxidant enzymes, oxidative modifications of macromolecules, and changes in mitochondrial homeostasis. Interestingly, alteration of redox homeostasis is closely associated with defects of energy metabolism, involving both carbohydrates and lipids, the major energy fuels for the cell. As the brain relies exclusively on glucose metabolism, defects of glucose utilization represent a harmful event for the brain. During aging, a progressive perturbation of energy metabolism occurs resulting in brain hypometabolism. This condition contributes to increase neuronal cell vulnerability ultimately resulting in cognitive impairment. The current review discusses the crosstalk between alteration of redox homeostasis and brain energy defects that seems to act in concert in promoting Alzheimer's neurodegeneration.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
22
|
Presta M, Zoratto F, Mulder D, Ottomana AM, Pisa E, Arias Vásquez A, Slattery DA, Glennon JC, Macrì S. Hyperglycemia and cognitive impairments anticipate the onset of an overt type 2 diabetes-like phenotype in TALLYHO/JngJ mice. Psychoneuroendocrinology 2024; 167:107102. [PMID: 38896988 DOI: 10.1016/j.psyneuen.2024.107102] [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: 02/20/2024] [Revised: 04/30/2024] [Accepted: 06/08/2024] [Indexed: 06/21/2024]
Abstract
Type 2 Diabetes mellitus (T2DM) is a metabolic disorder characterized by chronic hyperglycemia, resulting from deficits in insulin secretion, insulin action, or both. Whilst the role of insulin in the peripheral nervous system has been ascertained in countless studies, its role in the central nervous system (CNS) is emerging only recently. Brain insulin has been lately associated with brain disorders like Alzheimer's disease, obsessive compulsive disorder, and attention deficit hyperactivity disorder. Thus, understanding the role of insulin as a common risk factor for mental and somatic comorbidities may disclose novel preventative and therapeutic approaches. We evaluated general metabolism (glucose tolerance, insulin sensitivity, energy expenditure, lipid metabolism, and polydipsia) and cognitive capabilities (attention, cognitive flexibility, and memory), in adolescent, young adult, and adult male and female TALLYHO/JngJ mice (TH, previously reported to constitute a valid experimental model of T2DM due to impaired insulin signaling). Adult TH mice have also been studied for alterations in gut microbiota diversity and composition. While TH mice exhibited profound deficits in cognitive flexibility and altered glucose metabolism, we observed that these alterations emerged either much earlier (males) or independent of (females) a comprehensive constellation of symptoms, isomorphic to an overt T2DM-like phenotype (insulin resistance, polydipsia, higher energy expenditure, and altered lipid metabolism). We also observed significant sex-dependent alterations in gut microbiota alpha diversity and taxonomy in adult TH mice. Deficits in insulin signaling may represent a common risk factor for both T2DM and CNS-related deficits, which may stem from (partly) independent mechanisms.
Collapse
Affiliation(s)
- Martina Presta
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy; Department of Physiology and Pharmacology, Sapienza University of Rome, Rome 00185, Italy
| | - Francesca Zoratto
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Danique Mulder
- Donders Institute for Brain, Cognition and Behaviour, Departments of Psychiatry and Human Genetics, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Angela Maria Ottomana
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy; Neuroscience Unit, Department of Medicine, University of Parma, Parma 43100, Italy
| | - Edoardo Pisa
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Alejandro Arias Vásquez
- Donders Institute for Brain, Cognition and Behaviour, Departments of Psychiatry and Human Genetics, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Jeffrey C Glennon
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland
| | - Simone Macrì
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy.
| |
Collapse
|
23
|
Ari C, D'Agostino DP, Cha BJ. Neuroregeneration Improved by Sodium-D,L-Beta-Hydroxybutyrate in Primary Neuronal Cultures. Pharmaceuticals (Basel) 2024; 17:1160. [PMID: 39338322 PMCID: PMC11435142 DOI: 10.3390/ph17091160] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Ketone bodies are considered alternative fuels for the brain when glucose availability is limited. To determine the neuroregenerative potential of D,L-sodium-beta-hydroxybutyrate (D/L-BHB), Sprague Dawley rat primary cortical neurons were exposed to simulated central nervous system injury using a scratch assay. The neuronal cell migration, cell density and degree of regeneration in the damaged areas (gaps) in the absence (control) and presence of BHB (2 mM) were documented with automated live-cell imaging by the CytoSMART system over 24 h, which was followed by immunocytochemistry, labeling synapsin-I and β3-tubulin. The cell density was significantly higher in the gaps with BHB treatment after 24 h compared to the control. In the control, only 1.5% of the measured gap areas became narrower over 24 h, while in the BHB-treated samples 49.23% of the measured gap areas became narrower over 24 h. In the control, the gap expanded by 63.81% post-injury, while the gap size decreased by 10.83% in response to BHB treatment, compared to the baseline. The cell density increased by 97.27% and the gap size was reduced by 74.64% in response to BHB, compared to the control. The distance travelled and velocity of migrating cells were significantly higher with BHB treatment, while more synapsin-I and β3-tubulin were found in the BHB-treated samples after 24 h, compared to the control. The results demonstrate that D/L-BHB enhanced neuronal migration and molecular processes associated with neural regeneration and axonogenesis. These results may have clinical therapeutic applications in the future for nervous system injuries, such as for stroke, concussion and TBI patients.
Collapse
Affiliation(s)
- Csilla Ari
- Behavioral Neuroscience Laboratory, Department of Psychology, University of South Florida, Tampa, FL 33620, USA
- Ketone Technologies LLC, Tampa, FL 33612, USA
| | - Dominic P D'Agostino
- Ketone Technologies LLC, Tampa, FL 33612, USA
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
- Institute for Human and Machine Cognition, Ocala, FL 34471, USA
| | - Byeong J Cha
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
| |
Collapse
|
24
|
Hurtado-Carneiro V, LeBaut-Ayuso Y, Velázquez E, Flores-Lamas C, Fernández-de la Rosa R, García-García L, Gómez-Oliver F, Ruiz-Albusac JM, Pozo MÁ. Effects of chronic treatment with metformin on brain glucose hypometabolism and central insulin actions in transgenic mice with tauopathy. Heliyon 2024; 10:e35752. [PMID: 39170185 PMCID: PMC11337050 DOI: 10.1016/j.heliyon.2024.e35752] [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: 01/31/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Brain glucose hypometabolism and insulin alterations are common features of many neurological diseases. Herein we sought to corroborate the brain glucose hypometabolism that develops with ageing in 12-months old Tau-VLW transgenic mice, a model of tauopathy, as well as to determine whether this model showed signs of altered peripheral glucose metabolism. Our results demonstrated that 12-old months Tau mice exhibited brain glucose hypometabolism as well as basal hyperglycemia, impaired glucose tolerance, hyperinsulinemia, and signs of insulin resistance. Then, we further studied the effect of chronic metformin treatment (9 months) in Tau-VLW mice from 9 to 18 months of age. Longitudinal PET neuroimaging studies revealed that chronic metformin altered the temporal profile in the progression of brain glucose hypometabolism associated with ageing. Besides, metformin altered the content and/or phosphorylation of key components of the insulin signal transduction pathway in the frontal cortex leading to significant changes in the content of the active forms. Thus, metformin increased the expression of pAKT-Y474 while reducing pmTOR-S2448 and pGSK3β. These changes might be related, at least partially, to a slow progression of ageing, neurological damage, and cognitive decline. Metformin also improved the peripheral glucose tolerance and the ability of the Tau-VLW mice to maintain their body weight through ageing. Altogether our study shows that the tau-VLW mice could be a useful model to study the potential interrelationship between tauopathy and central and peripheral glucose metabolism alterations. More importantly our results suggest that chronic metformin treatment may have direct beneficial central effects by post-transcriptional modulation of key components of the insulin signal transduction pathway.
Collapse
Affiliation(s)
| | - Yannick LeBaut-Ayuso
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Esther Velázquez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Cinthya Flores-Lamas
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | | | - Luis García-García
- Pluridisciplinary Institute, Complutense University, IdISSC, Madrid, Spain
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Complutense University, Madrid, Spain
| | - Francisca Gómez-Oliver
- Pluridisciplinary Institute, Complutense University, IdISSC, Madrid, Spain
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Complutense University, Madrid, Spain
| | - Juan Miguel Ruiz-Albusac
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Miguel Ángel Pozo
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
- Pluridisciplinary Institute, Complutense University, IdISSC, Madrid, Spain
| |
Collapse
|
25
|
Liu Y, Yang X, Gao J, Xie C, Tian C, Gao T. Association between triglyceride glucose index and cognitive decline: A meta-analysis. J Affect Disord 2024; 359:70-77. [PMID: 38735580 DOI: 10.1016/j.jad.2024.05.049] [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: 02/06/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND The triglyceride glucose (TyG) index, a novel surrogate indicator for insulin resistance (IR), is believed to be associated with various diseases. However, its connection with cognitive decline remains controversy. METHODS The PubMed, EMBASE, Cochrane Library, Web of Science, and Medline databases were systematically searched up to October 2023 to assess the association between the TyG index and the risk of cognitive decline. Effect estimates and 95 % confidence intervals (CIs) were calculated using a random-effects model. RESULTS Our review included 3 cohort studies and 9 case-control/cross-sectional studies with a total of 5,603,350 participants. In comparison to a low TyG index, a higher TyG index was connected to an elevated risk of cognitive decline (RR/HR = 1.14, 95 % CI [1.11, 1.17], P < 0.05; OR = 1.75, 95 % CI [1.34, 2.29], P < 0.05). Furthermore, the dose-response analysis from the case-control/cross-sectional studies revealed a 1.42 times higher risk of cognitive decline per 1 mg/dl increment of the TyG index (OR = 1.42, 95 % CI [1.19, 1.69], P < 0.05). LIMITATIONS The inclusion of observational studies in the meta-analysis demonstrated a lower hierarchy of evidence compared to randomized controlled trials. Moreover, we incorporated a restricted number of studies and identified significant heterogeneity among them, potentially attributed to the presence of numerous confounding variables. CONCLUSION TyG index is related to cognitive decline. In view of some of the limitations of this study, further research will be necessary to confirm this relationship.
Collapse
Affiliation(s)
- Yuqin Liu
- School of Public Health, Qingdao University, 38 Dengzhou Road, Qingdao 266021, China.
| | - Xingxiang Yang
- School of Public Health, Qingdao University, 38 Dengzhou Road, Qingdao 266021, China.
| | - Jie Gao
- School of Public Health, Qingdao University, 38 Dengzhou Road, Qingdao 266021, China.
| | - Chenqi Xie
- School of Public Health, Qingdao University, 38 Dengzhou Road, Qingdao 266021, China.
| | - Chunyan Tian
- School of Public Health, Qingdao University, 38 Dengzhou Road, Qingdao 266021, China
| | - Tianlin Gao
- School of Public Health, Qingdao University, 38 Dengzhou Road, Qingdao 266021, China.
| |
Collapse
|
26
|
Abdel-Kareem NM, Elshazly SM, Abd El Fattah MA, Aldahish AA, Zaitone SA, Ali SK, Abd El-Haleim EA. Nifedipine Improves the Ketogenic Diet Effect on Insulin-Resistance-Induced Cognitive Dysfunction in Rats. Pharmaceuticals (Basel) 2024; 17:1054. [PMID: 39204160 PMCID: PMC11359371 DOI: 10.3390/ph17081054] [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: 07/05/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Insulin resistance, induced by high fructose consumption, affects cognitive function negatively. Nifedipine may be suggested for neurological disorders. This study aimed to assess the effect of nifedipine with either a normal diet (ND) or a ketogenic diet (KD) in cognitive dysfunction. Male Wistar rats received 10% fructose in drinking water for 8 weeks to induce insulin resistance. Rats received nifedipine (5.2 mg/kg/day; p.o.) later with ND or KD for an additional five weeks. One and two-way ANOVAs were used in analyzing the data. Reversion to the ND improved insulin resistance and lipid profile, besides brain-derived neurotrophic factor (BDNF), glycogen synthase kinase-3 beta (GSK3β), and insulin-degrading enzyme (IDE) levels. Rats fed KD alone and those that received nifedipine with KD did not show similar improvement in the previously mentioned parameters as the ND group. However, nifedipine-ND rats showed improvement in cognitive behavior and insulin resistance. Treatment with nifedipine-KD ameliorated GSK3β, amyloid β (Aβ), and tau protein levels. As the nifedipine-KD combination succeeded in diminishing the accumulated Aβ and tau protein, KD may be used for a while due to its side effects, then nifedipine treatment could be continued with an ND. This conclusion is based on the finding that this combination mitigated insulin resistance with the associated improved behavior.
Collapse
Affiliation(s)
- Nancy M. Abdel-Kareem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University—Arish Branch, Arish 45511, Egypt
| | - Shimaa M. Elshazly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - May A. Abd El Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt (E.A.A.E.-H.)
| | - Afaf A. Aldahish
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, Abha 61441, Saudi Arabia;
| | - Sawsan A. Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Sahar K. Ali
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia;
| | - Enas A. Abd El-Haleim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt (E.A.A.E.-H.)
| |
Collapse
|
27
|
Liang X, Xing Z, Lai K, Li X, Gui S, Li Y. Sex differences in the association between metabolic score for insulin resistance and the reversion to normoglycemia in adults with prediabetes: a cohort study. Diabetol Metab Syndr 2024; 16:183. [PMID: 39080757 PMCID: PMC11288094 DOI: 10.1186/s13098-024-01430-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/24/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND The metabolic score for insulin resistance (MetS-IR) has become a valid indicator to evaluate insulin resistance. Our investigation sought gender differences in the correlation between MetS-IR and the reversion from prediabetes to normoglycemic status. METHODS This retrospective research, carried out in 32 areas across 11 cities with several centers in China, encompassed 15,423 participants with prediabetes. We employed a Cox proportional hazards regression model to examine the link between MetS-IR and the reversion to normoglycemic status. We also applied cubic spline functions and smooth curve fitting to detect non-linear relationships. Additionally, we embarked on a range of sensitivity analyses. RESULTS The study included 15,423 participants, with 10,009 males (64.90%) and 5,414 females (35.10%). The average follow-up time was 2.96 ± 0.93 years, and 6,623 individuals (42.94%) reversed normoglycemia. A non-linear correlation was discovered among MetS-IR and reversion to normoglycemic status in men, with a turning point at 55.48. For a one-unit rise in MetS-IR below this point, the chance of reversal to normoglycemic levels declined by 3% (HR = 0.97, 95% CI:0.96-0.97, P < 0.0001). In women, the association was linear, with every unit rise in MetS-IR leading to a 3% reduction in transitioning to normal glycemic levels. (HR = 0.97, 95% CI: 0.97-0.98, p < 0.0001). CONCLUSION A negative correlation was discovered between MetS-IR and reversion to normoglycemic status in adults with prediabetes. Specifically, a non-linear association was observed for males, while females exhibited a linear correlation.
Collapse
Affiliation(s)
- Xiaomin Liang
- Department of Critical Care Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zemao Xing
- Department of Critical Care Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Kai Lai
- Department of Critical Care Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiaohong Li
- Department of Critical Care Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Shuiqing Gui
- Department of Critical Care Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.
| | - Ying Li
- Department of Critical Care Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.
| |
Collapse
|
28
|
Bai W, An S, Jia H, Xu J, Qin L. Relationship between triglyceride-glucose index and cognitive function among community-dwelling older adults: a population-based cohort study. Front Endocrinol (Lausanne) 2024; 15:1398235. [PMID: 39104819 PMCID: PMC11298491 DOI: 10.3389/fendo.2024.1398235] [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: 03/09/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Background The global increase in the aging population presents considerable challenges, particularly regarding cognitive impairment, a major concern for public health. This study investigates the association between the triglyceride-glucose (TyG) index, a measure of insulin resistance, and the risk of cognitive impairment in the elderly. Methods This prospective cohort study enrolled 2,959 participants aged 65 and above from the 2015 and 2020 waves of the China Health and Retirement Longitudinal Study (CHARLS). The analysis employed a logistic regression model to assess the correlation between the TyG index and cognitive impairment. Results The study included 2,959 participants, with a mean age of 71.2 ± 5.4 years, 49.8% of whom were female. The follow-up in 2020 showed a decrease in average cognitive function scores from 8.63 ± 4.61 in 2015 to 6.86 ± 5.45. After adjusting for confounding factors, a significant association was observed between TyG index quartiles and cognitive impairment. Participants in the highest quartile (Q4) of baseline TyG had a higher risk of cognitive impairment compared to those in the lowest quartile (Q1) (odds ratio [OR]: 1.97, 95% confidence intervals [CI]: 1.28-2.62, P<0.001). Conclusion The study highlights a significant connection between elevated TyG index levels and cognitive impairment among older adults in China. These findings suggest that targeted interventions to reduce the TyG index could mitigate cognitive impairment and potentially lower the incidence of dementia.
Collapse
Affiliation(s)
- Weimin Bai
- Department of Emergency, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Shuang An
- Department of Pediatric Rehabilitation, Henan Children’s Hospital Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Hui Jia
- Department of Convalescent Four Areas Nine Departments, Navy Qingdao Special Service Recuperation Center, Qingdao, China
| | - Juan Xu
- Department of General Surgery, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Lijie Qin
- Department of Emergency, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| |
Collapse
|
29
|
Boccardi V, Travaglini EG, Sciacca E, Mancinetti F, Murasecco I, Guazzarini AG, Bastiani P, Ruggiero C, Mecocci P. Dysglycemia, gender, and cognitive performance in older persons living with mild cognitive impairment: findings from a cross-sectional, population-based study. Aging Clin Exp Res 2024; 36:145. [PMID: 39012506 PMCID: PMC11252216 DOI: 10.1007/s40520-024-02806-7] [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: 04/01/2024] [Accepted: 07/02/2024] [Indexed: 07/17/2024]
Abstract
OBJECTIVE This study aims to examine the relationship between dysglycemia - also known as pre-diabetes or impaired glucose tolerance- and cognitive abilities in an older population living Mild Cognitive Impairment (MCI) and stratified by gender. STUDY DESIGN This is a retrospective study with data gathered from a large Italian clinical-based database. MAIN OUTCOME MEASURES The evaluation of cognitive performances by the Mini-Mental State Examination and the Addenbrooke's Cognitive Examination Revised rating scale as tests of screening and a comprehensive neuropsychological evaluation of several cognitive areas. RESULTS The study comprised 682 subjects (445 F/237 M) with a mean age of 76.08 ± 9.03 (range: 66-93) years. In all population, subjects with dysglycemia 193 (28.3%) had significantly poorer performance in memory (p = 0.006) and logic reasoning (p = 0.007) when compared with subjects without dysglycemia. The linear regression analyses revealed significant differences in the correlates of cognitive domains between gender groups. Independent of multiple covariates, women with dysglycemia showed worse performances in attention and short-term memory domains as compared with men. Even in the absence of dysglycemia women were more likely to show lower score in screening test of general cognition and attention. CONCLUSIONS Our findings suggest that dysglycemia in older individuals with MCI is associated with declines in specific cognitive domains, potentially influenced by gender. Implementing a comprehensive approach involving risk stratification and preventive strategies may be more effective in averting further cognitive decline in this high-risk population.
Collapse
Affiliation(s)
- Virginia Boccardi
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, Perugia, 06132, Italy.
| | - Emma Giulia Travaglini
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, Perugia, 06132, Italy
| | - Emanuela Sciacca
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, Perugia, 06132, Italy
| | - Francesca Mancinetti
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, Perugia, 06132, Italy
| | - Ilenia Murasecco
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, Perugia, 06132, Italy
| | - Anna Giulia Guazzarini
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, Perugia, 06132, Italy
| | - Patrizia Bastiani
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, Perugia, 06132, Italy
| | - Carmelinda Ruggiero
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, Perugia, 06132, Italy
| | - Patrizia Mecocci
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, Perugia, 06132, Italy
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
30
|
Samadzadeh S, Adnan R, Berglova P, Barzegar M, Debrabant B, Roikjaer SG, Levy M, Petzold A, Palace J, Flanagan EP, Mariotto S, Skou ST, Froelich A, Lotan I, Messina S, Geraldes R, Asseyer S, Stiebel-Kalish H, Oertel FC, Shaygannejad V, Sahraian MA, Kim HJ, Bennett JL, Böttcher C, Zimmermann HG, Weinshenker BG, Paul F, Asgari N. Protocol of a prospective multicenter study on comorbidity impact on multiple sclerosis and antibody-mediated diseases of the central nervous system (COMMIT). Front Immunol 2024; 15:1380025. [PMID: 39021565 PMCID: PMC11253107 DOI: 10.3389/fimmu.2024.1380025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/05/2024] [Indexed: 07/20/2024] Open
Abstract
Comorbidities in patients with multiple sclerosis (MS) and antibody-mediated diseases of the central nervous system (CNS) including neuromyelitis optica spectrum disorder (NMOSD), and myelin oligodendrocyte glycoprotein (MOG)-antibody-associated disease (MOGAD) are common and may influence the course of their neurological disease. Comorbidity may contribute to neuronal injury and therefore limit recovery from attacks, accelerate disease progression, and increase disability. This study aims to explore the impact of comorbidity, particularly vascular comorbidity, and related risk factors on clinical and paraclinical parameters of MS, NMOSD and MOGAD. We propose COMMIT, a prospective multicenter study with longitudinal follow-up of patients with MS, NMOSD, and MOGAD, with or without comorbidities, as well as healthy subjects as controls. Subjects will be stratified by age, sex and ethnicity. In consecutive samples we will analyze levels of inflammation and neurodegeneration markers in both fluid and cellular compartments of the peripheral blood and cerebrospinal fluid (CSF) using multiple state-of-the-art technologies, including untargeted proteomics and targeted ultrasensitive ELISA assays and quantitative reverse transcription polymerase chain reaction (RT-qPCR) as well as high-dimensional single-cell technologies i.e., mass cytometry and single-cell RNA sequencing. Algorithm-based data analyses will be used to unravel the relationship between these markers, optical coherence tomography (OCT) and magnetic resonance imaging (MRI), and clinical outcomes including frequency and severity of relapses, long-term disability, and quality of life. The goal is to evaluate the impact of comorbidities on MS, NMOSD, and MOGAD which may lead to development of treatment approaches to improve outcomes of inflammatory demyelinating diseases of the CNS.
Collapse
Affiliation(s)
- Sara Samadzadeh
- Institute of Regional Health Research and, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- The Center for Neurological Research, Department of Neurology Slagelse Hospital, Slagelse, Denmark
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
| | - Rafl Adnan
- Institute of Regional Health Research and, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- The Center for Neurological Research, Department of Neurology Slagelse Hospital, Slagelse, Denmark
| | - Paulina Berglova
- The Center for Neurological Research, Department of Neurology Slagelse Hospital, Slagelse, Denmark
| | - Mahdi Barzegar
- Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Birgit Debrabant
- Department of Mathematics and Computer Science, Faculty of Natural Sciences, University of Southern Denmark, Odense, Denmark
| | - Stine Gundtoft Roikjaer
- Institute of Regional Health Research and, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- The Center for Neurological Research, Department of Neurology Slagelse Hospital, Slagelse, Denmark
- The Research and Implementation Unit PROgrez, Department of Physiotherapy and Occupational Therapy, Næstved-Slagelse-Ringsted Hospitals, Slagelse, Region Zealand, Denmark
| | - Michael Levy
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Axel Petzold
- The National Hospital for Neurology and Neurosurgery, and Moorfields Eye Hospital and Queen Square University College London (UCL), Institute of Neurology, London, United Kingdom
- Department of Neurology, Amsterdam The University Medical Center Utrecht (UMC), Amsterdam, Netherlands
- Department of Ophthalmology, Amsterdam The University Medical Center Utrecht (UMC), Amsterdam, Netherlands
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, National Health Service Trust, Oxford, United Kingdom
| | - Eoin P. Flanagan
- Department Neurology and Center for Multiple Sclerosis (MS), and Autoimmune Neurology, Mayo Clinic, Rochester, MN, United States
| | - Sara Mariotto
- Neurology Unit, Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Verona, Italy
| | - Soeren T. Skou
- The Research and Implementation Unit PROgrez, Department of Physiotherapy and Occupational Therapy, Næstved-Slagelse-Ringsted Hospitals, Slagelse, Region Zealand, Denmark
- Center for Muscle and Joint Health, Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Anne Froelich
- The Center for Neurological Research, Department of Neurology Slagelse Hospital, Slagelse, Denmark
- Innovation and Research Centre for Multimorbidity, Slagelse Hospital, Slagelse, Denmark
- Section of General Practice, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Itay Lotan
- Department of Neurology and Neuroimmunology Unit, Rabin Medical Center, Petah Tikva, Israel
- Tel Aviv University Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Silvia Messina
- Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, National Health Service Trust, Oxford, United Kingdom
| | - Ruth Geraldes
- Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, National Health Service Trust, Oxford, United Kingdom
| | - Susanna Asseyer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- NeuroCure Clinical Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Hadas Stiebel-Kalish
- Tel Aviv University Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Ophthalmology, Neuro-Ophthalmology Unit, Rabin Medical Center, Petah Tikva, Israel
- Eye Laboratory, Felsenstein Research Institute, Tel Aviv, Israel
| | - Frederike Cosima Oertel
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- NeuroCure Clinical Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Vahid Shaygannejad
- Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Ali Sahraian
- Multiple Sclerosis (MS) Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ho Jin Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang, Republic of Korea
| | - Jeffrey L. Bennett
- Department of Neurology and Ophthalmology, Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Chotima Böttcher
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Hanna G. Zimmermann
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- NeuroCure Clinical Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Einstein Center Digital Future, Berlin, Germany
| | - Brian G. Weinshenker
- Department of Neurology, University of Virginia, Charlottesville, VA, United States
| | - Friedemann Paul
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- NeuroCure Clinical Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nasrin Asgari
- Institute of Regional Health Research and, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- The Center for Neurological Research, Department of Neurology Slagelse Hospital, Slagelse, Denmark
- Open Patient data Explorative Network, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
31
|
Guo B, Li QY, Liu XJ, Luo GH, Wu YJ, Nie J. Diabetes mellitus and Alzheimer's disease: Vacuolar adenosine triphosphatase as a potential link. Eur J Neurosci 2024; 59:2577-2595. [PMID: 38419188 DOI: 10.1111/ejn.16286] [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: 12/01/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 03/02/2024]
Abstract
Globally, the incidence of diabetes mellitus (DM) and Alzheimer's disease (AD) is increasing year by year, causing a huge economic and social burden, and their pathogenesis and aetiology have been proven to have a certain correlation. In recent years, more and more studies have shown that vacuolar adenosine triphosphatases (v-ATPases) in eukaryotes, which are biomolecules regulating lysosomal acidification and glycolipid metabolism, play a key role in DM and AD. This article describes the role of v-ATPase in DM and AD, including its role in glycolysis, insulin secretion and insulin resistance (IR), as well as its relationship with lysosomal acidification, autophagy and β-amyloid (Aβ). In DM, v-ATPase is involved in the regulation of glucose metabolism and IR. v-ATPase is closely related to glycolysis. On the one hand, v-ATPase affects the rate of glycolysis by affecting the secretion of insulin and changing the activities of key glycolytic enzymes hexokinase (HK) and phosphofructokinase 1 (PFK-1). On the other hand, glucose is the main regulator of this enzyme, and the assembly and activity of v-ATPase depend on glucose, and glucose depletion will lead to its decomposition and inactivation. In addition, v-ATPase can also regulate free fatty acids, thereby improving IR. In AD, v-ATPase can not only improve the abnormal brain energy metabolism by affecting lysosomal acidification and autophagy but also change the deposition of Aβ by affecting the production and degradation of Aβ. Therefore, v-ATPase may be the bridge between DM and AD.
Collapse
Affiliation(s)
- Bin Guo
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Qi-Ye Li
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xue-Jia Liu
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Guo-Hui Luo
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ya-Juan Wu
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jing Nie
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
32
|
Martinez-Feduchi P, Jin P, Yao B. Epigenetic modifications of DNA and RNA in Alzheimer's disease. Front Mol Neurosci 2024; 17:1398026. [PMID: 38726308 PMCID: PMC11079283 DOI: 10.3389/fnmol.2024.1398026] [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: 03/08/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder and the most common form of dementia. There are two main types of AD: familial and sporadic. Familial AD is linked to mutations in amyloid precursor protein (APP), presenilin-1 (PSEN1), and presenilin-2 (PSEN2). On the other hand, sporadic AD is the more common form of the disease and has genetic, epigenetic, and environmental components that influence disease onset and progression. Investigating the epigenetic mechanisms associated with AD is essential for increasing understanding of pathology and identifying biomarkers for diagnosis and treatment. Chemical covalent modifications on DNA and RNA can epigenetically regulate gene expression at transcriptional and post-transcriptional levels and play protective or pathological roles in AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
33
|
Momina SS, Gandla K. Flavonoid-Rich Trianthema decandra Ameliorates Cognitive Dysfunction in the Hyperglycemic Rats. Biochem Genet 2024:10.1007/s10528-024-10744-2. [PMID: 38570442 DOI: 10.1007/s10528-024-10744-2] [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: 12/31/2023] [Accepted: 02/14/2024] [Indexed: 04/05/2024]
Abstract
The present study was aimed at the evaluation of neuroprotective ability of methanolic extract of Trianthema decandra (METD) against hyperglycemia-related cognitive impairment in rats. The extract of T. decandra was standardized by TLC and HPTLC methods. To verify the identity and purity of isolated compounds, they were segregated and characterized using various techniques, including UV-visible spectrophotometry, FT-IR, H-NMR, and Mass spectroscopy. α-Amylase and α-glucosidase inhibition property of the extracts were assessed in-vitro. The screening of the neuroprotective effects of METD in hyperglycemic rats was done utilizing Morri's water (MWM) and elevated plus maze (EPM) model, as well as acetylcholinesterase (AChE) activity. The extracts of Trianthema decandra and its chemical constituents, namely quercetin and phytol, demonstrated a significant protective effect on enzymes like α-amylase and α-glucosidase. Methanol and hydroalcoholic extracts have shown the strongest inhibitory activity followed by chloroform extract. Quercetin and phytol were associated with the methanolic and chloroform extracts which were identified using TLC and HPTLC techniques. During the thirty days of the study, the induction of diabetes in the rats exhibited persistent hyperglycemia, hyperlipidemia, higher escape latency during training trials and reduced time spent in target quadrant in probe trial in Morris water maze test, and increased escape latency in EPM task. Regimen of METD (200 and 400 mg/kg) in the diabetic rats reduced the glucose levels in blood, lipid, and liver profile and showed positive results on Morri's water and elevated plus maze tasks. During the investigation, it was determined that Trianthema decandra extracts and the chemical constituent's quercetin and phytol in it had anti-diabetic and neuroprotective activities.
Collapse
Affiliation(s)
- Sayyada Saleha Momina
- Department of Pharmacognosy and Phytochemistry, Chaitanya (Deemed to be University), Gandipet, HimayathNagar (Vill), Hyderabad, Telangana, 500075, India
| | - Kumaraswamy Gandla
- Department of Pharmacy, Chaitanya (Deemed to be University), Gandipet, HimayathNagar (Vill), Hyderabad, Telangana, 500075, India.
| |
Collapse
|
34
|
Choi AH, Delgado M, Chen KY, Chung ST, Courville A, Turner SA, Yang S, Airaghi K, Dustin I, McGurrin P, Wu T, Hallett M, Ehrlich DJ. A randomized feasibility trial of medium chain triglyceride-supplemented ketogenic diet in people with Parkinson's disease. BMC Neurol 2024; 24:106. [PMID: 38561682 PMCID: PMC10983636 DOI: 10.1186/s12883-024-03603-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 03/17/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND A ketogenic diet (KD) may benefit people with neurodegenerative disorders marked by mitochondrial depolarization/insufficiency, including Parkinson's disease (PD). OBJECTIVE Evaluate whether a KD supplemented by medium chain triglyceride (MCT-KD) oil is feasible and acceptable for PD patients. Furthermore, we explored the effects of MCT-KD on blood ketone levels, metabolic parameters, levodopa absorption, mobility, nonmotor symptoms, simple motor and cognitive tests, autonomic function, and resting-state electroencephalography (rsEEG). METHODS A one-week in-hospital, double-blind, randomized, placebo-controlled diet (MCT-KD vs. standard diet (SD)), followed by an at-home two-week open-label extension. The primary outcome was KD feasibility and acceptability. The secondary outcome was the change in Timed Up & Go (TUG) on day 7 of the diet intervention. Additional exploratory outcomes included the N-Back task, Unified Parkinson's Disease Rating Scale, Non-Motor Symptom Scale, and rsEEG connectivity. RESULTS A total of 15/16 subjects completed the study. The mean acceptability was 2.3/3, indicating willingness to continue the KD. Day 7 TUG time was not significantly different between the SD and KD groups. The nonmotor symptom severity score was reduced at the week 3 visit and to a greater extent in the KD group. UPDRS, 3-back, and rsEEG measures were not significantly different between groups. Blood ketosis was attained by day 4 in the KD group and to a greater extent at week 3 than in the SD group. The plasma levodopa metabolites DOPAC and dopamine both showed nonsignificant increasing trends over 3 days in the KD vs. SD groups. CONCLUSIONS An MCT-supplemented KD is feasible and acceptable to PD patients but requires further study to understand its effects on symptoms and disease. TRIAL REGISTRATION Trial Registration Number NCT04584346, registration dates were Oct 14, 2020 - Sept 13, 2022.
Collapse
Affiliation(s)
- Alexander H Choi
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
- Mid-Atlantic Permanente Medical Group, Kaiser Permanente Mid-Atlantic States, Rockville, MD, USA.
| | - Melanie Delgado
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kong Y Chen
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie T Chung
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amber Courville
- NIH Clinical Center Nutrition Department, National Institutes of Health, Bethesda, MD, USA
| | - Sara A Turner
- NIH Clinical Center Nutrition Department, National Institutes of Health, Bethesda, MD, USA
| | - Shanna Yang
- NIH Clinical Center Nutrition Department, National Institutes of Health, Bethesda, MD, USA
| | - Kayla Airaghi
- NIH Clinical Center Nutrition Department, National Institutes of Health, Bethesda, MD, USA
| | - Irene Dustin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Patrick McGurrin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Tianxia Wu
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mark Hallett
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Debra J Ehrlich
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
35
|
Xu Y, Nie J, Lu C, Hu C, Chen Y, Ma Y, Huang Y, Lu L. Effects and mechanisms of bisphenols exposure on neurodegenerative diseases risk: A systemic review. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 919:170670. [PMID: 38325473 DOI: 10.1016/j.scitotenv.2024.170670] [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: 10/18/2023] [Revised: 01/12/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Environmental bisphenols (BPs) pose a global threat to human health because of their extensive use as additives in plastic products. BP residues are increasing in various environmental media (i.e., water, soil, and indoor dust) and biological and human samples (i.e., serum and brain). Both epidemiological and animal studies have determined an association between exposure to BPs and an increased risk of neurodegenerative diseases (e.g., Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis), including cognitive abnormalities and behavioral disturbances. Hence, understanding the biological responses to different BPs is essential for prevention, and treatment. This study provides an overview of the underlying pathogenic molecular mechanisms as a valuable basis for understanding neurodegenerative disease responses to BPs, including accumulation of misfolded proteins, reduction of tyrosine hydroxylase and dopamine, abnormal hormone signaling, neuronal death, oxidative stress, calcium homeostasis, and inflammation. These findings provide new insights into the neurotoxic potential of BPs and ultimately contribute to a comprehensive health risk evaluation.
Collapse
Affiliation(s)
- Yeqing Xu
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jun Nie
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; School of Engineering, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Chenghao Lu
- College of Mathematics and Computer Science, Zhejiang A & F University, Hangzhou 311300, China
| | - Chao Hu
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; School of Engineering, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yunlu Chen
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Ying Ma
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yuru Huang
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Liping Lu
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| |
Collapse
|
36
|
Basatinya AM, Sajedianfard J, Nazifi S, Hosseinzadeh S. The analgesic effects of insulin and its disorders in streptozotocin-induced short-term diabetes. Physiol Rep 2024; 12:e16009. [PMID: 38639646 PMCID: PMC11027902 DOI: 10.14814/phy2.16009] [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: 12/29/2023] [Revised: 04/01/2024] [Accepted: 04/01/2024] [Indexed: 04/20/2024] Open
Abstract
Evidence suggests that insulin resistance plays an important role in developing diabetes complications. The association between insulin resistance and pain perception is less well understood. This study aimed to investigate the effects of peripheral insulin deficiency on pain pathways in the brain. Diabetes was induced in 60 male rats with streptozotocin (STZ). Insulin was injected into the left ventricle of the brain by intracerebroventricular (ICV) injection, then pain was induced by subcutaneous injection of 2.5% formalin. Samples were collected at 4 weeks after STZ injection. Dopamine (DA), serotonin, reactive oxygen species (ROS), and mitochondrial glutathione (mGSH) were measured by ELISA, and gene factors were assessed by RT-qPCR. In diabetic rats, the levels of DA, serotonin, and mGSH decreased in the nuclei of the thalamus, raphe magnus, and periaqueductal gray, and the levels of ROS increased. In addition, the levels of expression of the neuron-specific enolase and receptor for advanced glycation end genes increased, but the expression of glial fibrillary acidic protein expression was reduced. These results support the findings that insulin has an analgesic effect in non-diabetic rats, as demonstrated by the formalin test. ICV injection of insulin reduces pain sensation, but this was not observed in diabetic rats, which may be due to cell damage ameliorated by insulin.
Collapse
Affiliation(s)
| | - Javad Sajedianfard
- Department of Basic Sciences, School of Veterinary MedicineShiraz UniversityShirazIran
| | - Saeed Nazifi
- Department of Clinical Science, School of Veterinary MedicineShiraz UniversityShirazIran
| | - Saied Hosseinzadeh
- Department of Food Hygiene and Public Health, School of Veterinary MedicineShiraz UniversityShirazIran
| |
Collapse
|
37
|
Pinheiro FI, Araújo-Filho I, do Rego ACM, de Azevedo EP, Cobucci RN, Guzen FP. Hepatopancreatic metabolic disorders and their implications in the development of Alzheimer's disease and vascular dementia. Ageing Res Rev 2024; 96:102250. [PMID: 38417711 DOI: 10.1016/j.arr.2024.102250] [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: 12/05/2023] [Revised: 02/07/2024] [Accepted: 02/22/2024] [Indexed: 03/01/2024]
Abstract
Dementia has been faced with significant public health challenges and economic burdens that urges the need to develop safe and effective interventions. In recent years, an increasing number of studies have focused on the relationship between dementia and liver and pancreatic metabolic disorders that result in diseases such as diabetes, obesity, hypertension and dyslipidemia. Previous reports have shown that there is a plausible correlation between pathologies caused by hepatopancreatic dysfunctions and dementia. Glucose, insulin and IGF-1 metabolized in the liver and pancreas probably have an important influence on the pathophysiology of the most common dementias: Alzheimer's and vascular dementia. This current review highlights recent studies aimed at identifying convergent mechanisms, such as insulin resistance and other diseases, linked to altered hepatic and pancreatic metabolism, which are capable of causing brain changes that ultimately lead to dementia.
Collapse
Affiliation(s)
- Francisco I Pinheiro
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Department of Surgical, Federal University of Rio Grande do Norte, Natal 59010-180, Brazil; Institute of Education, Research and Innovation of the Liga Norte Rio-Grandense Against Cancer
| | - Irami Araújo-Filho
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Department of Surgical, Federal University of Rio Grande do Norte, Natal 59010-180, Brazil; Postgraduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | - Amália C M do Rego
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Institute of Education, Research and Innovation of the Liga Norte Rio-Grandense Against Cancer
| | - Eduardo P de Azevedo
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil
| | - Ricardo N Cobucci
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Postgraduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil; Postgraduate Program in Science Applied to Women`s Health, Medical School, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | - Fausto P Guzen
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Postgraduate Program in Health and Society, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró, Brazil; Postgraduate Program in Physiological Sciences, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró, Brazil.
| |
Collapse
|
38
|
Chamberland É, Moravveji S, Doyon N, Duchesne S. A computational model of Alzheimer's disease at the nano, micro, and macroscales. Front Neuroinform 2024; 18:1348113. [PMID: 38586183 PMCID: PMC10995318 DOI: 10.3389/fninf.2024.1348113] [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: 12/01/2023] [Accepted: 02/26/2024] [Indexed: 04/09/2024] Open
Abstract
Introduction Mathematical models play a crucial role in investigating complex biological systems, enabling a comprehensive understanding of interactions among various components and facilitating in silico testing of intervention strategies. Alzheimer's disease (AD) is characterized by multifactorial causes and intricate interactions among biological entities, necessitating a personalized approach due to the lack of effective treatments. Therefore, mathematical models offer promise as indispensable tools in combating AD. However, existing models in this emerging field often suffer from limitations such as inadequate validation or a narrow focus on single proteins or pathways. Methods In this paper, we present a multiscale mathematical model that describes the progression of AD through a system of 19 ordinary differential equations. The equations describe the evolution of proteins (nanoscale), cell populations (microscale), and organ-level structures (macroscale) over a 50-year lifespan, as they relate to amyloid and tau accumulation, inflammation, and neuronal death. Results Distinguishing our model is a robust foundation in biological principles, ensuring improved justification for the included equations, and rigorous parameter justification derived from published experimental literature. Conclusion This model represents an essential initial step toward constructing a predictive framework, which holds significant potential for identifying effective therapeutic targets in the fight against AD.
Collapse
Affiliation(s)
- Éléonore Chamberland
- Centre de Recherche CERVO, Institut Universitaire de Santé Mentale de Québec, Québec, QC, Canada
- Département de Mathématiques et de Statistique, Québec, QC, Canada
| | - Seyedadel Moravveji
- Centre de Recherche CERVO, Institut Universitaire de Santé Mentale de Québec, Québec, QC, Canada
- Département de Mathématiques et de Statistique, Québec, QC, Canada
| | - Nicolas Doyon
- Centre de Recherche CERVO, Institut Universitaire de Santé Mentale de Québec, Québec, QC, Canada
- Département de Mathématiques et de Statistique, Québec, QC, Canada
| | - Simon Duchesne
- Centre de Recherche CERVO, Institut Universitaire de Santé Mentale de Québec, Québec, QC, Canada
- Département de Radiologie et Médecine Nucléaire, Université Laval, Québec, QC, Canada
- Centre de Recherche de l'Institut Universitaire en Cardiologie et Pneumologie de Québec, Québec, QC, Canada
| |
Collapse
|
39
|
Hu R, Geng Y, Huang Y, Liu Z, Li F, Dong H, Ma W, Song K, Zhang M, Zhang Z, Song Y. New insights into the interaction between polycystic ovary syndrome and psychiatric disorders: A narrative review. Int J Gynaecol Obstet 2024; 164:387-420. [PMID: 37458179 DOI: 10.1002/ijgo.14988] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 01/15/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disease characterized by hyperandrogenism, ovulatory dysfunction, and ovarian polycystic changes, which combines with reproductive problems, metabolic disorders, and psychological disorders to exhibit a far-reaching impact on the physical and mental health of women. We reviewed previous research and discovered that psychiatric disorders are more common in PCOS patients and their children, potentially exacerbating the condition and creating a vicious loop. To understand the reasons, relevant articles were collected following the Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines from PubMed, Web of Science, and Cochrane Library, through December 2022. Evidence suggested that PCOS-related clinical manifestations, hyperandrogenism, insulin resistance, obesity, gut dysbiosis, and other variables may increase the risk of psychiatric disorders in patients. In turn, psychiatric disorders may aggravate the pathologic process of PCOS and increase the difficulty of the treatment. We systematically reported the mechanisms underlying the psychiatric disorders-PCOS interactions, intending to provide potential ways to break the vicious cycle and lay the groundwork for future research. However, research on PCOS and psychiatric disorders were still in initial stages, which limited the scope of this review. More studies are needed to further verify our findings.
Collapse
Affiliation(s)
- Runan Hu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuli Geng
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanjing Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuo Liu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoxu Dong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenwen Ma
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kunkun Song
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingmin Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuo Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yufan Song
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
40
|
Yakupov EZ, Zhamieva RA. [Cognitive impairments and emotional disorders and their correction in perimenopausal women]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:48-53. [PMID: 39072566 DOI: 10.17116/jnevro202412406148] [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: 07/30/2024]
Abstract
The presented literature review reveals the topic of the features of risk factors for cognitive impairment in women in the perimenopausal period (PMP), as well as the possibilities of their earliest detection and correction. The paper searches for various symptoms and predictors of the development of cognitive impairment in women in the PMP. The key features include certain difficulties in making a diagnosis at earlier stages. The relationship of metabolic disorders with factors negatively affecting the health of women in the PPP, as well as contributing to the deterioration of cognitive functions, is considered. Women are more at risk of developing cognitive impairment and represent a specific target group that requires special attention in assessing risk factors and methods for correcting cognitive disorders. To date, the relationship between gender and dementia risk still needs to be studied in more depth. Given this, menopause is an important physiological period, as it is accompanied by intense hormonal changes that may be the direct cause of cognitive decline. Many women experience mood disorders, anxiety, increased mental and/or physical fatigue, irritability, mild cognitive disorders, which requires an interdisciplinary approach by doctors to this issue. All these manifestations should be evaluated and corrected in time to avoid their progression and a decrease in the quality of life. An integrated approach to therapy, both medicinal and non-medicinal, can significantly improve the quality of life of patients in the PPP.
Collapse
Affiliation(s)
- E Z Yakupov
- LLC Scientific Research Medical Complex «Vashe Zdorovya», Kazan, Russia
| | - R A Zhamieva
- LLC Scientific Research Medical Complex «Vashe Zdorovya», Kazan, Russia
| |
Collapse
|
41
|
Gambino G, Frinchi M, Giglia G, Scordino M, Urone G, Ferraro G, Mudò G, Sardo P, Di Majo D, Di Liberto V. Impact of “Golden” tomato juice on cognitive alterations in metabolic syndrome: Insights into behavioural and biochemical changes in a high-fat diet rat model. J Funct Foods 2024; 112:105964. [DOI: 10.1016/j.jff.2023.105964] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
42
|
Na D, Lim DH, Hong JS, Lee HM, Cho D, Yu MS, Shaker B, Ren J, Lee B, Song JG, Oh Y, Lee K, Oh KS, Lee MY, Choi MS, Choi HS, Kim YH, Bui JM, Lee K, Kim HW, Lee YS, Gsponer J. A multi-layered network model identifies Akt1 as a common modulator of neurodegeneration. Mol Syst Biol 2023; 19:e11801. [PMID: 37984409 DOI: 10.15252/msb.202311801] [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: 06/05/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/22/2023] Open
Abstract
The accumulation of misfolded and aggregated proteins is a hallmark of neurodegenerative proteinopathies. Although multiple genetic loci have been associated with specific neurodegenerative diseases (NDs), molecular mechanisms that may have a broader relevance for most or all proteinopathies remain poorly resolved. In this study, we developed a multi-layered network expansion (MLnet) model to predict protein modifiers that are common to a group of diseases and, therefore, may have broader pathophysiological relevance for that group. When applied to the four NDs Alzheimer's disease (AD), Huntington's disease, and spinocerebellar ataxia types 1 and 3, we predicted multiple members of the insulin pathway, including PDK1, Akt1, InR, and sgg (GSK-3β), as common modifiers. We validated these modifiers with the help of four Drosophila ND models. Further evaluation of Akt1 in human cell-based ND models revealed that activation of Akt1 signaling by the small molecule SC79 increased cell viability in all models. Moreover, treatment of AD model mice with SC79 enhanced their long-term memory and ameliorated dysregulated anxiety levels, which are commonly affected in AD patients. These findings validate MLnet as a valuable tool to uncover molecular pathways and proteins involved in the pathophysiology of entire disease groups and identify potential therapeutic targets that have relevance across disease boundaries. MLnet can be used for any group of diseases and is available as a web tool at http://ssbio.cau.ac.kr/software/mlnet.
Collapse
Affiliation(s)
- Dokyun Na
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Do-Hwan Lim
- College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- School of Systems Biomedical Science, Soongsil University, Seoul, Republic of Korea
| | - Jae-Sang Hong
- College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Hyang-Mi Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Daeahn Cho
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Myeong-Sang Yu
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Bilal Shaker
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Jun Ren
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Bomi Lee
- College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Jae Gwang Song
- College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Yuna Oh
- Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Kyungeun Lee
- Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Kwang-Seok Oh
- Information-based Drug Research Center, Korea Research Institute of Chemical Technology, Deajeon, Republic of Korea
| | - Mi Young Lee
- Information-based Drug Research Center, Korea Research Institute of Chemical Technology, Deajeon, Republic of Korea
| | - Min-Seok Choi
- College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Han Saem Choi
- College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Yang-Hee Kim
- College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Jennifer M Bui
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Kangseok Lee
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Hyung Wook Kim
- College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Young Sik Lee
- College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jörg Gsponer
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
43
|
Ottomana AM, Presta M, O'Leary A, Sullivan M, Pisa E, Laviola G, Glennon JC, Zoratto F, Slattery DA, Macrì S. A systematic review of preclinical studies exploring the role of insulin signalling in executive function and memory. Neurosci Biobehav Rev 2023; 155:105435. [PMID: 37913873 DOI: 10.1016/j.neubiorev.2023.105435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/04/2023] [Accepted: 10/23/2023] [Indexed: 11/03/2023]
Abstract
Beside its involvement in somatic dysfunctions, altered insulin signalling constitutes a risk factor for the development of mental disorders like Alzheimer's disease and obsessive-compulsive disorder. While insulin-related somatic and mental disorders are often comorbid, the fundamental mechanisms underlying this association are still elusive. Studies conducted in rodent models appear well suited to help decipher these mechanisms. Specifically, these models are apt to prospective studies in which causative mechanisms can be manipulated via multiple tools (e.g., genetically engineered models and environmental interventions), and experimentally dissociated to control for potential confounding factors. Here, we provide a narrative synthesis of preclinical studies investigating the association between hyperglycaemia - as a proxy of insulin-related metabolic dysfunctions - and impairments in working and spatial memory, and attention. Ultimately, this review will advance our knowledge on the role of glucose metabolism in the comorbidity between somatic and mental illnesses.
Collapse
Affiliation(s)
- Angela Maria Ottomana
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; Neuroscience Unit, Department of Medicine, University of Parma, 43100 Parma, Italy
| | - Martina Presta
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | - Aet O'Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany; Chair of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Mairéad Sullivan
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland
| | - Edoardo Pisa
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Giovanni Laviola
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Jeffrey C Glennon
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland
| | - Francesca Zoratto
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Simone Macrì
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy.
| |
Collapse
|
44
|
Carter KJ, Ward AT, Al-Subu A, Wilson AD, Zevin EL, Serlin RC, Eldridge M, Wieben O, Schrage WG. An oral glucose tolerance test does not affect cerebral blood flow: role of NOS. Am J Physiol Regul Integr Comp Physiol 2023; 325:R759-R768. [PMID: 37842740 PMCID: PMC11178292 DOI: 10.1152/ajpregu.00169.2022] [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: 07/01/2022] [Revised: 09/11/2023] [Accepted: 10/06/2023] [Indexed: 10/17/2023]
Abstract
Animal data indicate that insulin triggers a robust nitric oxide synthase (NOS)-mediated dilation in cerebral arteries similar to the peripheral tissue vasodilation observed in healthy adults. Insulin's role in regulating cerebral blood flow (CBF) in humans remains unclear but may be important for understanding the links between insulin resistance, diminished CBF, and poor brain health outcomes. We tested the hypothesis that an oral glucose challenge (oral glucose tolerance test, OGTT), which increases systemic insulin and glucose, would acutely increase CBF in healthy adults due to NOS-mediated vasodilation, and that changes in CBF would be greater in anterior regions where NOS expression or activity may be greater. In a randomized, single-blind approach, 18 young healthy adults (24 ± 5 yr) underwent magnetic resonance imaging (MRI) with a placebo before and after an OGTT (75 g glucose), and 11 of these adults also completed an NG-monomethyl-l-arginine (l-NMMA) visit. Four-dimensional (4-D) flow MRI quantified macrovascular CBF and arterial spin labeling (ASL) quantified microvascular perfusion. Subjects completed baseline imaging with a placebo (or l-NMMA), then consumed an OGTT followed by MRI scans and blood sampling every 10-15 min for 90 min. Contrary to our hypothesis, total CBF (P = 0.17) and global perfusion (P > 0.05) did not change at any time point up to 60 min after the OGTT, and no regional changes were detected. l-NMMA did not mediate any effect of OGTT on CBF. These data suggest that insulin-glucose challenge does not acutely alter CBF in healthy adults.
Collapse
Affiliation(s)
- Katrina J Carter
- Department of Kinesiology, University of Wisconsin, Madison, Wisconsin, United States
| | - Aaron T Ward
- Deparment of Center for Health Disparities Research, University of Wisconsin, Madison, Wisconsin, United States
| | - Awni Al-Subu
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States
| | - Allen D Wilson
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States
| | - Erika L Zevin
- Department of Pediatrics, University School of Medicine, Indianapolis, Indiana, United States
| | - Ronald C Serlin
- Department of Educational Psychology, University of Wisconsin, Madison, Wisconsin, United States
| | - Marlowe Eldridge
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States
| | - Oliver Wieben
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin, United States
- Department of Radiology, University of Wisconsin, Madison, Wisconsin, United States
| | - William G Schrage
- Department of Kinesiology, University of Wisconsin, Madison, Wisconsin, United States
| |
Collapse
|
45
|
Bidzan L, Jurek P, Olech M, Bidzan-Wiącek M, Bidzan-Bluma I, Bidzan M. Somatic comorbidity and the progression of cognitive impairment. Front Aging Neurosci 2023; 15:1219449. [PMID: 38046465 PMCID: PMC10691469 DOI: 10.3389/fnagi.2023.1219449] [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: 05/09/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023] Open
Abstract
Background There are usually multiple factors underlying dementia in old age. Somatic comorbidity is one important element that influences the progression of cognitive impairment. Objective The goal of this study was to assess the relationship between the progression of cognitive impairment and the presence and severity of comorbidities based on a four-year observation. Material Out of 128 patients from the Clinic for Outpatients in Gdansk, who were recruited into the study based on the criteria of the Working Group on Mild Cognitive Impairment, a total of 93 participants completed the four-year observation. Only the data from participants who completed the full period of observations were analysed. The mean age of the group was M = 75.93 (SD = 9.43). The level of progression of cognitive impairment was measured using the Clinical Dementia Rating Scale - Sum of Boxes, the severity of comorbidities was measured using the modified Cumulative Illness Rating Scale, and, additionally, at the time of inclusion in the study, participants were assessed using the MMSE scale and the Activity Scale, and sociodemographic data were collected. The Generalized Estimating Equations method was employed to fit a marginal model for analyzing the data collected in a repeated measures design. The tested model elucidated the role of the overall severity of comorbidities in explaining the progression of cognitive impairment, while controlling for everyday activity and basic demographic variables. Results During the four-year observation, a significant decline in cognitive function (B = 1.86, p < 0.01) was observed in the examined sample. The statistical analysis revealed that individuals with higher overall severity of comorbidities exhibited significantly more pronounced progression of cognitive impairment over time. Regarding particular comorbidities, metabolic diseases were found to be associated with a poorer prognosis (rho = 0.41, p < 0.05). Furthermore, a time physical activity interaction was identified as predicting cognitive impairment, indicating that individuals who were more physically active at the beginning of the study exhibited significantly less pronounced progression of cognitive impairment over the course of the 4 years. Conclusion This study suggests the important roles of comorbidities and physical activity for the prognosis of mild cognitive impairment.
Collapse
Affiliation(s)
- Leszek Bidzan
- Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
- Department of Health Sciences, Pomeranian University in Słupsk, Słupsk, Poland
| | - Paweł Jurek
- Institute of Psychology, University of Gdańsk, Gdańsk, Poland
| | - Michał Olech
- Faculty of Health Sciences, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Ilona Bidzan-Bluma
- Institute of Psychology, University of Gdańsk, Gdańsk, Poland
- Institute of Pedagogy and Languages, University of Applied Sciences in Elbląg, Elbląg, Poland
| | - Mariola Bidzan
- Institute of Psychology, University of Gdańsk, Gdańsk, Poland
- Institute of Pedagogy and Languages, University of Applied Sciences in Elbląg, Elbląg, Poland
| |
Collapse
|
46
|
Ишмуратова АН, Абрамов МА, Кузнецов КО, Иванюта МВ, Шакирова ЗФ, Китапова АИ, Усмонов МД, Черноусова ЛМ, Валеева ЛИ, Кузнецова АЮ, Баисламов АС, Шайхетдинова АР, Миргалиев АА, Орозбердиев СТ, Якупова КИ. [The role of antidiabetic drugs in the treatment of Alzheimer's disease: systematic review]. PROBLEMY ENDOKRINOLOGII 2023; 69:73-83. [PMID: 37968954 PMCID: PMC10680548 DOI: 10.14341/probl13183] [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: 10/27/2022] [Revised: 04/07/2023] [Accepted: 04/07/2023] [Indexed: 11/17/2023]
Abstract
Recent studies show that Alzheimer's disease (AD) has many common links with conditions associated with insulin resistance, including neuroinflammation, impaired insulin signaling, oxidative stress, mitochondrial dysfunction and metabolic syndrome. The authors conducted an electronic search for publications in the PubMed/MEDLINE and Google Scholar databases using the keywords "amyloid beta", "Alzheimer type-3-diabetes", "intranasal insulin", "metformin", "type 2 diabetes mellitus", "incretins" and "PPARy agonists». A systematic literature search was conducted among studies published between 2005 and 2022. The authors used the following inclusion criteria: 1) Subjects who received therapy for AD and/or DM2, if the expected result concerned the risk of cognitive decline or the development of dementia; 2) The age of the study participants is > 50 years; 3) The type of studies included in this review were randomized clinical trials, population-based observational studies or case-control studies, prospective cohort studies, as well as reviews and meta-analyses; 4) The included articles were written in English. In recent years, there has been considerable interest in identifying the mechanisms of action of antidiabetic drugs and their potential use in AD. Human studies involving patients with mild cognitive impairment and Alzheimer's disease have shown that the administration of certain antidiabetic drugs, such as intranasal insulin, metformin, incretins and thiazolidinediones, can improve cognitive function and memory. The purpose of this study is to evaluate the effectiveness of antidiabetic drugs in the treatment of AD. According to the results of the study, metformin, intranasal insulin, thiazolidinediones and incretins showed a positive effect both in humans and in animal models. Recent studies show that thiazolidinediones can activate pathways in the brain that are regulated by IGF-1; however, rosiglitazone may pose a significant risk of side effects. The results of clinical studies on the use of metformin in AD are limited and contradictory.
Collapse
Affiliation(s)
| | | | | | - М. В. Иванюта
- Российский национальный исследовательский медицинский университет им. Н.И. Пирогова
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wei B, Dong Q, Ma J, Zhang A. The association between triglyceride-glucose index and cognitive function in nondiabetic elderly: NHANES 2011-2014. Lipids Health Dis 2023; 22:188. [PMID: 37932783 PMCID: PMC10629120 DOI: 10.1186/s12944-023-01959-0] [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: 09/07/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND The relationship between Insulin resistance (IR) evaluated through homeostasis model assessment insulin resistance (HOMA-IR) and cognitive function is controversial among nondiabetic individuals. No study so far has reported the association between the IR evaluated through triglyceride glucose (TyG) index and cognitive function among nondiabetics. This study aims to assess this association among US nondiabetic older elderly. METHODS Data were obtained from the 2011-2014 National Health and Nutrition Examination Survey (NHANES). Low cognitive function was evaluated using the Consortium to Establish a Registry for Alzheimer's Disease Battery for immediate word list learning (CERAD-WL) and delayed recall (CERAD-DR) test, the Animal Fluency Test (AFT), and the Digit Symbol Substitution Test (DSST). Logistic regression analyses were conducted to compute the odds ratio (OR) and 95% confidential interval (CI) to examine the association between the TyG index (continuous and quartiles) and low cognitive function. RESULTS A total of 661 nondiabetic older adults were included with a mean age of 68.62 ± 6.49 years. Compared to the 1st quartile of the TyG index, participants in the TyG index 4th quartile were associated with low cognitive function evaluated through the CERAD test (CERAD-WL and CERAD-DR) [OR: 2.62; 95% CI (1.31, 5.23); P < 0.05]. Subgroup analyses showed that females (ORQ4 VS Q1: 3.07; 95% CI (1.04, 9.05); P < 0.05) and smokers (OR Q4 VS Q1: 2.70; 95% CI (1.01, 7.26); P < 0.05) categories were related with a higher risk of low cognitive function. CONCLUSIONS A high TyG index was strongly correlated with low cognitive function evaluated through the CERAD test (CERAD-WL and CERAD-DR) among US nondiabetic older women. The management of IR in women might be beneficial to primarily prevent low cognitive function among nondiabetic older elderly.
Collapse
Affiliation(s)
- Baojian Wei
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, No.619 Changcheng Road, Daiyue District, Taian, 271000, China
| | - Qianni Dong
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, No.619 Changcheng Road, Daiyue District, Taian, 271000, China
| | - Jinlong Ma
- School of Nursing, Yanbian University, Yanji, China
| | - Aihua Zhang
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, No.619 Changcheng Road, Daiyue District, Taian, 271000, China.
| |
Collapse
|
48
|
Li Y, Chen J, Tu H, Ma Q, Wang M, Chen J, Chen M. Protective effects of GuanXinNing tablet (GXNT) on diabetic encephalopathy in zucker diabetic obesity (ZDF) rats. BMC Complement Med Ther 2023; 23:385. [PMID: 37891536 PMCID: PMC10605859 DOI: 10.1186/s12906-023-04195-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Diabetic encephalopathy (DE) is a complication of diabetes that leads to cognitive and behavioral decline. Utilizing safe and effective complementary and alternative medications for its management is a wise choice. Previous studies have shown that GuanXinNing Tablet (GXNT), an oral preparation primarily derived from two Chinese herbs, Salvia miltiorrhiza Bge. and Ligusticum chuanxiong Hort., exerts a beneficial neuroprotective effect. In this study, we explored the protective effects of GXNT on DE in male Zucker diabetic fatty (ZDF) rats induced by a high-fat diet, aiming to ascertain its significance and potential mechanisms. METHODS ZDF rats were induced to develop type 2 diabetes (T2DM) with DE by a high-fat diet and treated with GXNT for 8 weeks until they were 20 weeks old. Throughout the experiment, the animals' vital parameters, such as body weight, were continuously monitored. Cognitive function was evaluated using the Y maze test. Biochemical kits were employed to analyze blood glucose, lipids, and vascular endothelial-related factors. Cerebrovascular lesions were assessed using magnetic resonance angiography (MRA) imaging. Brain lesions were evaluated using hematoxylin and eosin (H&E) staining and ultrastructure observation. IgG and albumin (ALB) leakage were detected using immunofluorescence. RESULTS GXNT demonstrated an enhancement in the overall well-being of the animals. It notably improved cognitive and behavioral abilities, as demonstrated by extended retention time in the novel heterogeneous arm during the Y-maze test. GXNT effectively regulated glucose and lipid metabolism, reducing fasting and postprandial blood glucose, glycated hemoglobin (HbA1c), and total cholesterol (TC) levels. Additionally, it exhibited a protective effect on the vascular endothelium by reducing the serum TXB2/PGI2 ratio while elevating NO and PGI2 levels. Moreover, GXNT ameliorated stenosis and occlusion in cerebral vessel branches, increased the number of microvessels and neurons around the hippocampus, and improved microvascular occlusion in the cerebral cortex, along with addressing perivascular cell abnormalities. Immunofluorescence staining showed a decrease in the fluorescence intensity of IgG and ALB in the cerebral cortex. CONCLUSIONS GXNT demonstrated a highly satisfactory protective effect on DE in ZDF rats. Its mechanism of action could be based on the regulation of glucolipid metabolism and its protective effect on the vascular endothelium.
Collapse
Affiliation(s)
- Yajing Li
- The Department of Biopharmaceutical Technology, Zhejiang Institute of Economics and Trade, Hangzhou, 310018, China
| | - Jiaojiao Chen
- Institute of Comparative Medicine, Experimental Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310012, China
| | - Haiye Tu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Quanxin Ma
- Institute of Comparative Medicine, Experimental Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mulan Wang
- Institute of Comparative Medicine, Experimental Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- The Department of Medicine, Chiatai Qingchunbao Pharmaceutical Co., Ltd, Hangzhou, 310053, China
| | - Jie Chen
- Department of Vasculocardiology, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310006, China.
| | - Minli Chen
- Institute of Comparative Medicine, Experimental Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
49
|
Aderinto N, Olatunji G, Abdulbasit M, Ashinze P, Faturoti O, Ajagbe A, Ukoaka B, Aboderin G. The impact of diabetes in cognitive impairment: A review of current evidence and prospects for future investigations. Medicine (Baltimore) 2023; 102:e35557. [PMID: 37904406 PMCID: PMC10615478 DOI: 10.1097/md.0000000000035557] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/18/2023] [Indexed: 11/01/2023] Open
Abstract
Cognitive impairment in individuals with diabetes represents a multifaceted and increasingly prevalent health concern. This review critically examines the current evidence regarding the intricate relationship between diabetes and cognitive decline. It highlights the existing knowledge on the impact of diabetes on cognitive function, spanning from mild cognitive impairment to dementia, including vascular and Alzheimer dementia. The review underscores the need for a standardized diagnostic paradigm and explores research gaps, such as the implications of cognitive impairment in younger populations and various diabetes types. Furthermore, this review emphasizes the relevance of diabetes-related comorbidities, including hypertension and dyslipidemia, in influencing cognitive decline. It advocates for a comprehensive, interdisciplinary approach, integrating insights from neuroscience, endocrinology, and immunology to elucidate the mechanistic underpinnings of diabetes-related cognitive impairment. The second part of this review outlines prospective research directions and opportunities. It advocates for longitudinal studies to understand disease progression better and identifies critical windows of vulnerability. The search for accurate biomarkers and predictive factors is paramount, encompassing genetic and epigenetic considerations. Personalized approaches and tailored interventions are essential in addressing the substantial variability in cognitive outcomes among individuals with diabetes.
Collapse
Affiliation(s)
- Nicholas Aderinto
- Department of Medicine, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Kwara State, Nigeria
| | - Muili Abdulbasit
- Department of Medicine, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Patrick Ashinze
- Saint Francis Catholic Hospital, Okpara Inland, Warri Catholic Diocesan Hospital Commission, Delta State, Nigeria
| | - Olamide Faturoti
- Department of Medicine and Surgery, University of Ilorin, Kwara State, Nigeria
| | - Abayomi Ajagbe
- Department of Anatomy, College of Health Sciences, Nile University of Nigeria, Abuja, Nigeria
| | - Bonaventure Ukoaka
- Department of Internal Medicine, Asokoro District Hospital, Abuja, Nigeria
| | - Gbolahan Aboderin
- Department of Medicine, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| |
Collapse
|
50
|
Liang J, Liu B, Dong X, Wang Y, Cai W, Zhang N, Zhang H. Decoding the role of gut microbiota in Alzheimer's pathogenesis and envisioning future therapeutic avenues. Front Neurosci 2023; 17:1242254. [PMID: 37790586 PMCID: PMC10544353 DOI: 10.3389/fnins.2023.1242254] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/04/2023] [Indexed: 10/05/2023] Open
Abstract
Alzheimer's disease (AD) emerges as a perturbing neurodegenerative malady, with a profound comprehension of its underlying pathogenic mechanisms continuing to evade our intellectual grasp. Within the intricate tapestry of human health and affliction, the enteric microbial consortium, ensconced within the milieu of the human gastrointestinal tract, assumes a role of cardinal significance. Recent epochs have borne witness to investigations that posit marked divergences in the composition of the gut microbiota between individuals grappling with AD and those favored by robust health. The composite vicissitudes in the configuration of the enteric microbial assembly are posited to choreograph a participatory role in the inception and progression of AD, facilitated by the intricate conduit acknowledged as the gut-brain axis. Notwithstanding, the precise nature of this interlaced relationship remains enshrouded within the recesses of obscurity, poised for an exhaustive revelation. This review embarks upon the endeavor to focalize meticulously upon the mechanistic sway exerted by the enteric microbiota upon AD, plunging profoundly into the execution of interventions that govern the milieu of enteric microorganisms. In doing so, it bestows relevance upon the therapeutic stratagems that form the bedrock of AD's management, all whilst casting a prospective gaze into the horizon of medical advancements.
Collapse
Affiliation(s)
- Junyi Liang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Bin Liu
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Xiaohong Dong
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Yueyang Wang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Wenhui Cai
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Ning Zhang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Hong Zhang
- Heilongjiang Jiamusi Central Hospital, Jiamusi, Heilongjiang, China
| |
Collapse
|