1
|
Yang S, Zhang H, Zhu W, Niu T, Farooqui HFM, Wang J, Yang M, Liu E, Wang S. Peripheral blood heat shock protein 27 correlates with information processing speed and executive function, potentially serving as a marker for mild cognitive impairment in patients with type 2 diabetes mellitus. Diabetol Metab Syndr 2025; 17:167. [PMID: 40410804 PMCID: PMC12103020 DOI: 10.1186/s13098-025-01747-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/16/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND AND AIMS Previous study found that interleukin 1β (IL-1β) is associated with diabetic cognitive dysfunction. Heat shock protein 27 (HSP27) is one of the factors related to IL-1β associated inflammation. Here, we aim to investigate the role of HSP27 in mild cognitive impairment (MCI) in patients with type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS In this study, individuals with T2DM with and without MCI were recruited and categorized into Control and MCI groups. Plasma HSP27 levels were assessed and compared between the Control and MCI groups. Furthermore, the relationship between HSP27 and diabetic dysfunction was elucidated through association and regression analyses. Finally, diagnostic values were determined using ROC curves. RESULTS In humans, individuals with T2DM and MCI exhibit decreased levels of HSP27 compared to those without MCI. Notably, the levels of HSP27 are associated with neuropsychological test scores that reflect cognitive preferences. Additionally, decreased HSP27 levels serve as one of the risk factors for MCI in T2DM patients (OR = 0.355, P = 0.002). Moreover, there is a defined cut-off point for HSP27 in diagnosing MCI, set at 3.51 pg/ml, with a sensitivity of 47.2%, a specificity of 94.4%, and an area under the curve (AUC) of 0.695. CONCLUSIONS Generally speaking, HSP27 is linked to cognitive decline in individuals with T2DM. Decreased levels of HSP27 in plasma are identified as both a risk factor for MCI and a potential diagnostic biomarker for MCI in patients with T2DM. The diagnostic value of HSP27 in MCI is primarily reflected in its demonstrated true negative rate.
Collapse
Affiliation(s)
- Shufang Yang
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Haoqiang Zhang
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Wenwen Zhu
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Tong Niu
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | | | - Jue Wang
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Medical school, Dalian Medical University, Dalian, China
| | - Mingyue Yang
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Medical school, Dalian Medical University, Dalian, China
| | - Enlin Liu
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Medical school, Nantong University, Nantong, China
| | - Shaohua Wang
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.
| |
Collapse
|
2
|
Zhang Z, Novak V, Novak P, Mantzoros C, Ngo L, Lioutas V, Dai W. Intranasal insulin enhances resting-state functional connectivity in Type 2 Diabetes. PLoS One 2025; 20:e0324029. [PMID: 40392946 DOI: 10.1371/journal.pone.0324029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/09/2025] [Indexed: 05/22/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) affects cognition and resting-state functional connectivity (rsFC). Intranasal insulin (INI) has emerged as a potential treatment for T2DM-related cognitive decline. We aimed to evaluate the effect of INI treatment on rsFC with medio-prefrontal (mPFC) and left/right hippocampus (lHPC/rHPC), and their relationship with the cognition, hemoglobin A1c (HbA1c), and homeostatic model assessment of insulin resistance (HOMA-IR) and walking speed. An MRI sub-study of the MemAID trial was conducted, involving a 24-week treatment with either intranasal insulin or placebo. Blood oxygen level-dependent (BOLD) functional MRI (fMRI) images were acquired on eighteen DM subjects at baseline and eleven DM subjects (eight DM-INI patients and three DM-Placebo) at the end-of-treatment. Compared to DM-Placebo treated subjects, DM-INI patients showed increased mPFC-postcentral rsFC, lHPC-frontal rsFC, lHPC-postcentral rsFC, rHPC-frontal rsFC, and lHPC-mPFC rsFC (p < 0.05). The decreased HOMA-IR, which was observed in the MemAID trial, was associated with increased mPFC-basal ganglia rsFC (p < 0.05). This sub-study provides insights into potential mechanisms of INI effects on rsFC that require validation in a larger trial.
Collapse
Affiliation(s)
- Zongpai Zhang
- School of Computing, State University of New York at Binghamton, Binghamton, New York, United States of America
| | - Vera Novak
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Peter Novak
- Department of Neurology, Massachusetts General Brigham Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Christos Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center and Department of Medicine Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Long Ngo
- Department of Medicine, Beth Israel Deaconess Medical Center and School of Public Health, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Vasileios Lioutas
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Weiying Dai
- School of Computing, State University of New York at Binghamton, Binghamton, New York, United States of America
| |
Collapse
|
3
|
Tartau CG, Boboc IKS, Mititelu-Tartau L, Bogdan M, Buca BR, Pavel LL, Amalinei C. Exploring the Protective Effects of Traditional Antidiabetic Medications and Novel Antihyperglycemic Agents in Diabetic Rodent Models. Pharmaceuticals (Basel) 2025; 18:670. [PMID: 40430489 PMCID: PMC12114790 DOI: 10.3390/ph18050670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Type 2 Diabetes (T2D) is a complex metabolic disorder that affects multiple organs, leading to severe complications in the pancreas, kidneys, liver, and heart. Prolonged hyperglycemia, along with oxidative stress and chronic inflammation, plays a crucial role in accelerating tissue damage, significantly increasing the risk of diabetic complications such as nephropathy, hepatopathy, and cardiovascular disease. This review evaluates the protective effects of various antidiabetic treatments on organ tissues affected by T2D, based on findings from experimental animal models. Metformin, a first-line antidiabetic agent, has been widely recognized for its ability to reduce inflammation and oxidative stress, thereby mitigating diabetes-induced organ damage. Its protective role extends beyond glucose regulation, offering benefits such as improved mitochondrial function and reduced fibrosis in affected tissues. In addition to traditional therapies, new classes of antidiabetic drugs, including sodium-glucose co-transporter-2 inhibitors and glucagon-like peptide-1 (GLP-1) receptor agonists not only improve glycemic control but also exhibit nephroprotective and cardioprotective properties by reducing glomerular hyperfiltration, oxidative stress, and inflammation. Similarly, GLP-1 receptor agonists have been associated with reduced hepatic steatosis and enhanced cardiovascular function. Preclinical studies suggest that tirzepatide, a dual GLP-1/gastric inhibitory polypeptide receptor agonist may offer superior metabolic benefits compared to conventional GLP-1 agonists by improving β-cell function, enhancing insulin sensitivity, and reducing fatty liver progression. Despite promising preclinical results, differences between animal models and human physiology pose a challenge. Further clinical research is needed to confirm these effects and refine treatment strategies. Future T2D management aims to go beyond glycemic control, emphasizing organ protection and long-term disease prevention.
Collapse
Affiliation(s)
- Cosmin Gabriel Tartau
- Department of Morphofunctional Sciences I, Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.G.T.); (C.A.)
| | - Ianis Kevyn Stefan Boboc
- Department of Pharmacology, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Liliana Mititelu-Tartau
- Department of Pharmacology, Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iasi, Romania; (L.M.-T.); (B.R.B.)
| | - Maria Bogdan
- Department of Pharmacology, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Beatrice Rozalina Buca
- Department of Pharmacology, Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iasi, Romania; (L.M.-T.); (B.R.B.)
| | - Liliana Lacramioara Pavel
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800010 Galati, Romania;
| | - Cornelia Amalinei
- Department of Morphofunctional Sciences I, Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.G.T.); (C.A.)
| |
Collapse
|
4
|
Vandersmissen J, Dewachter I, Cuypers K, Hansen D. The Impact of Exercise Training on the Brain and Cognition in Type 2 Diabetes, and its Physiological Mediators: A Systematic Review. SPORTS MEDICINE - OPEN 2025; 11:42. [PMID: 40274715 PMCID: PMC12022206 DOI: 10.1186/s40798-025-00836-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/16/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Type 2 diabetes (T2DM) affects brain structure and function, and is associated with an increased risk of dementia and mild cognitive impairment. It is known that exercise training has a beneficial effect on cognition and brain structure and function, at least in healthy people, but the impact of exercise training on these aspects remains to be fully elucidated in patients with T2DM. OBJECTIVE To determine the impact of exercise training on cognition and brain structure and function in T2DM, and identify the involved physiological mediators. METHODS This paper systematically reviews studies that evaluate the effect of exercise training on cognition in T2DM, and aims to indicate the most beneficial exercise modality for improving or preserving cognition in this patient group. In addition, the possible physiological mediators and targets involved in these improvements are narratively described in the second part of this review. Papers published up until the 14th of January 2025 were searched by means of the electronic databases PubMed, Embase, and Web of Science. Studies directly investigating the effect of any kind of exercise training on the brain or cognition in patients with T2DM, or animal models thereof, were included, with the exception of human studies assessing cognition only at one time point, and studies combining exercise training with other interventions (e.g. dietary changes, cognitive training, etc.). Study quality was assessed by means of the TESTEX tool for human studies, and the CAMARADES tool for animal studies. RESULTS For the systematic part of the review, 22 papers were found to be eligible. 18 out of 22 papers (81.8%) showed a significant positive effect of exercise training on cognition in T2DM, of which two studies only showed significant improvements in the minority of the cognitive tests. Four papers (18.2%) could not find a significant effect of exercise on cognition in T2DM. Resistance and endurance exercise were found to be equally effective for achieving cognitive improvement. Machine-based power training is seemingly more effective than resistance training with body weight and elastic bands to reach cognitive improvement. In addition, BDNF, lactate, leptin, adiponectin, GSK3β, GLP-1, the AMPK/SIRT1 pathway, and the PI3K/Akt pathway were identified as plausible mediators directly from studies investigating the effect of exercise training on brain structure and function in T2DM. Via these mediators, exercise training induces multiple beneficial brain changes, such as increased neuroplasticity, increased insulin sensitivity, and decreased inflammation. CONCLUSION Overall, exercise training beneficially affects cognition and brain structure and function in T2DM, with resistance and endurance exercise having similar effects. However, there is a need for additional studies, and more methodological consistency between different studies in order to define an exercise program optimal for improving cognition in T2DM. Furthermore, we were able to define several mediators involved in the effect of exercise training on cognition in T2DM, but further research is necessary to unravel the entire process.
Collapse
Affiliation(s)
- Jitske Vandersmissen
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, Hasselt University, Wetenschapspark 7, 3590, Diepenbeek, Belgium.
| | - Ilse Dewachter
- Biomedical Research Institute, BIOMED, Hasselt University, 3590, Diepenbeek, Belgium
| | - Koen Cuypers
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, Hasselt University, Wetenschapspark 7, 3590, Diepenbeek, Belgium
- Movement Control and Neuroplasticity Research Group, Department of Movement Sciences, Group Biomedical Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Dominique Hansen
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, Hasselt University, Wetenschapspark 7, 3590, Diepenbeek, Belgium
- Heart Centre Hasselt, Jessa Hospital, 3500, Hasselt, Belgium
| |
Collapse
|
5
|
Chu J, Song J, Fan Z, Zhang R, Wang Q, Yi K, Gong Q, Liu B. Investigating the Effect and Mechanism of 3-Methyladenine Against Diabetic Encephalopathy by Network Pharmacology, Molecular Docking, and Experimental Validation. Pharmaceuticals (Basel) 2025; 18:605. [PMID: 40430426 PMCID: PMC12115123 DOI: 10.3390/ph18050605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Diabetic encephalopathy (DE), a severe neurological complication of diabetes mellitus (DM), is characterized by cognitive dysfunction. 3-Methyladenine (3-MA), a methylated adenine derivative, acts as a biomarker for DNA methylation and exhibits hypoglycemic and neuroprotective properties. However, the pharmacological mechanisms underlying 3-MA's therapeutic effects on diabetic microvascular complications remain incompletely understood, owing to the intricate and multifactorial pathogenesis of DE. Methods: This study employed network pharmacology and molecular docking techniques to predict potential targets and signaling pathways of 3-MA against DE, with subsequent validation through animal experiments to elucidate the molecular mechanisms of 3-MA in DE treatment. Results: Network pharmacological analysis identified two key targets of 3-MA in DE modulation: AKT and GSK3β. Molecular docking confirmed a strong binding affinity between 3-MA and AKT/GSK3β. In animal experiments, 3-MA significantly reduced blood glucose levels in diabetic mice, ameliorated learning and memory deficits, and preserved hippocampal neuronal integrity. Furthermore, we found that 3-MA inhibited apoptosis by regulating the expression of Bax and BCL-2. Notably, 3-MA also downregulated the expression of amyloid precursor protein (APP) and Tau while enhancing the expression of phosphorylated AKT and GSK-3β. Conclusions: Our findings may contribute to elucidating the therapeutic mechanisms of 3-MA in diabetic microangiopathy and provide potential therapeutic targets through activation of the AKT/GSK-3β pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Quan Gong
- Department of Medcine, Yangtze University, Jingzhou 434023, China; (J.C.)
| | - Benju Liu
- Department of Medcine, Yangtze University, Jingzhou 434023, China; (J.C.)
| |
Collapse
|
6
|
Zhang D, He X, Wang Y, Wang X, Han X, Liu H, Xing Y, Jiang B, Xiu Z, Bao Y, Dong Y. Hesperetin-Enhanced Metformin to Alleviate Cognitive Impairment via Gut-Brain Axis in Type 2 Diabetes Rats. Int J Mol Sci 2025; 26:1923. [PMID: 40076550 PMCID: PMC11900253 DOI: 10.3390/ijms26051923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Diabetes constitutes a risk factor for cognitive impairment, whereas insulin resistance serves as the shared pathogenesis underlying both diabetes and cognitive decline. The use of metformin for treating cognitive impairment remains controversial. The present study found that hesperetin, a flavanone derived from citrus peel, enhanced metformin's efficacy in reducing blood sugar levels, improving insulin sensitivity, and ameliorating cognitive impairment in diabetic rats. Additionally, it reduced the required dosage of metformin to one-third of its conventional dose. Transcriptome analysis and 16S rRNA sequencing revealed that the activation of insulin and cyclic-adenosine monophosphate response element binding protein (CREB)/brain-derived neurotrophic factor (BDNF) pathways benefited from the regulation of gut microbiota and the promotion of short-chain fatty acid (SCFA) producers such as Romboutsia. Furthermore, this study demonstrated that hesperetin supplementation counteracted the upregulation of β-site amyloid precursor protein cleaving enzyme 1 (BACE1), a pathological factor of Alzheimer's disease (AD) that was induced by metformin. Our findings reveal that hesperetin can be used in supplementary treatment for cognitive impairment associated with diabetes.
Collapse
Affiliation(s)
- Danyang Zhang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Xiaoshi He
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Yinbo Wang
- Dianxi Research Institute, Dalian University of Technology, Baoshan 678000, China;
| | - Xiaoyu Wang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Xiao Han
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Haodong Liu
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Yan Xing
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Bo Jiang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Zhilong Xiu
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Yongming Bao
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
- School of Ocean Science and Technology, Dalian University of Technology, Panjin 124221, China
| | - Yuesheng Dong
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| |
Collapse
|
7
|
Zhang Y, Zhang R, Wang X, Shi L, Zhu H, Liu J. Potential role of ghrelin in neuroprotection and cognitive function: implications for diabetic cognitive impairment. PeerJ 2025; 13:e18898. [PMID: 39995985 PMCID: PMC11849504 DOI: 10.7717/peerj.18898] [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: 09/25/2024] [Accepted: 01/03/2025] [Indexed: 02/26/2025] Open
Abstract
Ghrelin is a class of brain and intestinal peptides. It regulates food intake and body glucose levels and maintains cellular homeostasis. In recent years, research has revealed that ghrelin may positively impact learning and memory. Despite ghrelin's multiple functions in the central nervous system, its use as a therapeutic agent for neurologic dysfunction remains unclear. Diabetic cognitive impairment (DCI) is a severe neurological complication of diabetes mellitus. Its incidence is increasing as a comorbidity in endocrinology and neurology. Additionally, it is a risk factor for Alzheimer' s disease (AD). Ghrelin levels are altered in patients with diabetes mellitus combined with cognitive impairment. Furthermore, modulation of ghrelin levels improved cognitive function in rats with DCI. These findings suggest the potential therapeutic importance of ghrelin in the pathogenesis of DCI. This article presents a comprehensive review of the pathogenesis of DCI and its potential modulation by ghrelin and its mimics. Furthermore, this study elucidates the therapeutic prospects of ghrelin and its mimics for DCI, aiming to identify novel therapeutic targets and research avenues for the prevention and management of DCI in the future.
Collapse
Affiliation(s)
- Yuhan Zhang
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xi’an, Xianyang, China
| | - Ruihua Zhang
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xi’an, Xianyang, China
| | - Xin Wang
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xi’an, Xianyang, China
| | - Leilei Shi
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xi’an, Xianyang, China
| | - Hongzhe Zhu
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xi’an, Xianyang, China
| | - Jiping Liu
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xi’an, Xianyang, China
- Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xianyang, China
- Shaanxi Key Laboratory for Safety Monitoring of Food and Drug, Xianyang, China
- Engineering Research Center of Brain Health Industry of Chinese Medicine, Universities of Shaanxi Province, Xianyang, China
| |
Collapse
|
8
|
Chib S, Dutta BJ, Chalotra R, Abubakar M, Kumar P, Singh TG, Singh R. Role of Flavonoids in Mitigating the Pathological Complexities and Treatment Hurdles in Alzheimer's Disease. Phytother Res 2025; 39:747-775. [PMID: 39660432 DOI: 10.1002/ptr.8406] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024]
Abstract
With the passage of time, people step toward old age and become more prone to several diseases associated with the age. One such is Alzheimer's disease (AD) which results into neuronal damage and dementia with the progression of age. The existing therapeutics has been hindered by various enkindles like less eminent between remote populations, affordability issues and toxicity profiles. Moreover, lack of suitable therapeutic option further worsens the quality of life in older population. Developing an efficient therapeutic intervention to cure AD is still a challenge for medical fraternity. Recently, alternative approaches attain the attention of researchers to focus on plant-based therapy in mitigating AD. In this context, flavonoids gained centrality as a feasible treatment in modifying various neurological deficits. This review mainly focuses on the pathological facets and economic burden of AD. Furthermore, we have explored the possible mechanism of flavonoids with the preclinical and clinical aspects for curing AD. Flavonoids being potential therapeutic, target the pathogenic factors of AD such as oxidative stress, inflammation, metal toxicity, Aβ accumulation, modulate neurotransmission and insulin signaling. In this review, we emphasized on potential neuroprotective effects of flavonoids in AD pathology, with focus on both experimental and clinical findings. While preclinical studies suggest promising therapeutic benefits, clinical data remains limited and inconclusive. Thus, further high-quality clinical trials are necessary to validate the efficacy of flavonoids in AD. The study aim is to promote the plant-based therapies and encourage people to add flavonoids to regular diet to avail the beneficial effects in preventive therapy for AD.
Collapse
Affiliation(s)
- Shivani Chib
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Bhaskar Jyoti Dutta
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - Rishabh Chalotra
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Md Abubakar
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | | | - Randhir Singh
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| |
Collapse
|
9
|
Padhy DS, Aggarwal P, Velayutham R, Banerjee S. Aerobic exercise and metformin attenuate the cognitive impairment in an experimental model of type 2 diabetes mellitus: focus on neuroinflammation and adult hippocampal neurogenesis. Metab Brain Dis 2025; 40:92. [PMID: 39775196 DOI: 10.1007/s11011-024-01489-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder that increases the prevalence of cognitive impairment in the geriatric population. Aerobic exercise is an excellent non-pharmacological therapeutic strategy to prevent Alzheimer's disease, the most common form of dementia. The exact molecular mechanism of aerobic exercise (Exe) as an intervention to counter cognitive decline is far from clear. Metformin is a first-line agent against T2DM with neuroprotective properties. The present study assessed the role of treadmill exercise in combination with a low dose of metformin (Met; 70 mg/kg) in cognitive impairment and its associated molecular mechanism in T2DM rats. The experimental model of T2DM-associated cognitive decline was created by administration of a high-fat diet (HFD) with a low dose of streptozotocin (STZ; 35 mg/kg). Neurobehavioral assessments were performed to evaluate spatial recognition and fear-conditioned memory across the groups: control, HFD + STZ, HFD + STZ + Exe, and HFD + STZ + Exe + Met. In addition, we performed immunohistochemistry and western blotting on the rat hippocampal tissue from the above groups for protein expression studies. T2DM rats showed a significant cognitive decline compared to the control group, which improved in the long-term exercise and metformin co-administered animals. The level of neuroinflammation was significantly elevated in the hippocampal tissue of T2DM rats compared to the control and lowered after exercise and metformin treatment. T2DM reduced mature neurons and neurogenesis while increasing astrogliosis and microgliosis, ameliorated by exercise and metformin treatment. Moreover, T2DM impaired hippocampal neurogenesis by reducing the canonical Wnt/β-catenin pathway, which got upregulated in exercise and metformin-co-administered rats. Long-term aerobic exercise with metformin treatment ameliorated neuroinflammation and promoted adult hippocampal neurogenesis via upregulating the canonical Wnt/β-catenin pathway in T2DM rats.
Collapse
Affiliation(s)
- Dibya Sundar Padhy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Kolkata, Kolkata, West Bengal, 700054, India
| | - Punita Aggarwal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Kolkata, Kolkata, West Bengal, 700054, India
| | - Ravichandiran Velayutham
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER)- Kolkata, Kolkata, West Bengal, 700054, India.
| | - Sugato Banerjee
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Kolkata, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
10
|
Abdalla MMI. Advancing diabetes management: Exploring pancreatic beta-cell restoration's potential and challenges. World J Gastroenterol 2024; 30:4339-4353. [PMID: 39494103 PMCID: PMC11525866 DOI: 10.3748/wjg.v30.i40.4339] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/05/2024] [Accepted: 09/24/2024] [Indexed: 10/16/2024] Open
Abstract
Diabetes mellitus, characterized by chronic hyperglycemia due to insulin deficiency or resistance, poses a significant global health burden. Central to its pathogenesis is the dysfunction or loss of pancreatic beta cells, which are res-ponsible for insulin production. Recent advances in beta-cell regeneration research offer promising strategies for diabetes treatment, aiming to restore endogenous insulin production and achieve glycemic control. This review explores the physiological basis of beta-cell function, recent scientific advan-cements, and the challenges in translating these findings into clinical applications. It highlights key developments in stem cell therapy, gene editing technologies, and the identification of novel regenerative molecules. Despite the potential, the field faces hurdles such as ensuring the safety and long-term efficacy of regen-erative therapies, ethical concerns around stem cell use, and the complexity of beta-cell differentiation and integration. The review highlights the importance of interdisciplinary collaboration, increased funding, the need for patient-centered approaches and the integration of new treatments into comprehensive care strategies to overcome these challenges. Through continued research and collaboration, beta-cell regeneration holds the potential to revolutionize diabetes care, turning a chronic condition into a manageable or even curable disease.
Collapse
Affiliation(s)
- Mona Mohamed Ibrahim Abdalla
- Department of Human Biology, School of Medicine, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| |
Collapse
|
11
|
Dash UK, Mazumdar D, Singh S. High Mobility Group Box Protein (HMGB1): A Potential Therapeutic Target for Diabetic Encephalopathy. Mol Neurobiol 2024; 61:8188-8205. [PMID: 38478143 DOI: 10.1007/s12035-024-04081-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/28/2024] [Indexed: 09/21/2024]
Abstract
HMGB (high mobility group B) is one of the ubiquitous non-histone nuclear protein superfamilies that make up the HMG (high mobility group) protein group. HMGB1 is involved in a variety of physiological and pathological processes in the human body, including a structural role in the cell nucleus as well as replication, repair, DNA transcription, and assembly of nuclear proteins. It functions as a signaling regulator in the cytoplasm and a pro-inflammatory cytokine in the extracellular environment. Among several studies, HMGB1 protein is also emerging as a crucial factor involved in the development and progression of diabetic encephalopathy (DE) along with other factors such as hyperglycaemia-induced oxidative and nitrosative stress. Diabetes' chronic side effect is DE, which manifests as cognitive and psychoneurological dysfunction. The HMGB1 is released outside to the extracellular medium in diabetes condition through active or passive routes, where it functions as a damage-associated molecular pattern (DAMP) molecule to activate several signaling pathways by interacting with receptors for advanced glycosylation end-products (RAGE)/toll like receptors (TLR). HMGB1 reportedly activates inflammatory pathways, disrupts the blood-brain barrier, causes glutamate toxicity and oxidative stress, and promotes neuroinflammation, contributing to the development of cognitive impairment and neuronal damage which is suggestive of the involvement of HMGB1 in the enhancement of the diabetes-induced encephalopathic condition. Additionally, HMGB1 is reported to induce insulin resistance, further exacerbating the metabolic dysfunction associated with diabetes mellitus (DM). Thus, the present review explores the possible pathways associated with DM-induced hyperactivation of HMGB1 ultimately leading to DE.
Collapse
Affiliation(s)
- Udit Kumar Dash
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, 495009, India
| | - Debashree Mazumdar
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, 495009, India
| | - Santosh Singh
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, 495009, India.
| |
Collapse
|
12
|
Ni W, Niu Y, Cao S, Fan C, Fan J, Zhu L, Wang X. Intermittent hypoxia exacerbates anxiety in high-fat diet-induced diabetic mice by inhibiting TREM2-regulated IFNAR1 signaling. J Neuroinflammation 2024; 21:166. [PMID: 38956653 PMCID: PMC11218348 DOI: 10.1186/s12974-024-03160-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) and obstructive sleep apnea (OSA) are mutual risk factors, with both conditions inducing cognitive impairment and anxiety. However, whether OSA exacerbates cognitive impairment and anxiety in patients with T2DM remains unclear. Moreover, TREM2 upregulation has been suggested to play a protective role in attenuating microglia activation and improving synaptic function in T2DM mice. The aim of this study was to explore the regulatory mechanisms of TREM2 and the cognitive and anxiety-like behavioral changes in mice with OSA combined with T2DM. METHODS A T2DM with OSA model was developed by treating mice with a 60% kcal high-fat diet (HFD) combined with intermittent hypoxia (IH). Spatial learning memory capacity and anxiety in mice were investigated. Neuronal damage in the brain was determined by the quantity of synapses density, the number and morphology of brain microglia, and pro-inflammatory factors. For mechanism exploration, an in vitro model of T2DM combined with OSA was generated by co-treating microglia with high glucose (HG) and IH. Regulation of TREM2 on IFNAR1-STAT1 pathway was determined by RNA sequencing and qRT-PCR. RESULTS Our results showed that HFD mice exhibited significant cognitive dysfunction and anxiety-like behavior, accompanied by significant synaptic loss. Furthermore, significant activation of brain microglia and enhanced microglial phagocytosis of synapses were observed. Moreover, IH was found to significantly aggravate anxiety in the HFD mice. The mechanism of HG treatment may potentially involve the promotion of TREM2 upregulation, which in turn attenuates the proinflammatory microglia by inhibiting the IFNAR1-STAT1 pathway. Conversely, a significant reduction in TREM2 in IH-co-treated HFD mice and HG-treated microglia resulted in the further activation of the IFNAR1-STAT1 pathway and consequently increased proinflammatory microglial activation. CONCLUSIONS HFD upregulated the IFNAR1-STAT1 pathway and induced proinflammatory microglia, leading to synaptic damage and causing anxiety and cognitive deficits. The upregulated TREM2 inT2DM mice brain exerted a negative regulation of the IFNAR1-STAT1 pathway. Mice with T2DM combined with OSA exacerbated anxiety via the downregulation of TREM2, causing heightened IFNAR1-STAT1 pathway activation and consequently increasing proinflammatory microglia.
Collapse
MESH Headings
- Animals
- Mice
- Diet, High-Fat/adverse effects
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/genetics
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/genetics
- Anxiety/etiology
- Anxiety/metabolism
- Signal Transduction/physiology
- Signal Transduction/drug effects
- Hypoxia/metabolism
- Hypoxia/complications
- Male
- Mice, Inbred C57BL
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/psychology
- Receptor, Interferon alpha-beta/metabolism
- Receptor, Interferon alpha-beta/genetics
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Microglia/metabolism
- STAT1 Transcription Factor/metabolism
- Sleep Apnea, Obstructive/complications
- Sleep Apnea, Obstructive/metabolism
- Sleep Apnea, Obstructive/psychology
Collapse
Affiliation(s)
- Wenyu Ni
- Qidong People's Hospital, Affiliated Qidong Hospital of Nantong University, Qidong Liver Cancer Institute, No.9, Seyuan Road, Chongchuan District, Nantong, Jiangsu, 226000, China
| | - Yun Niu
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Sitong Cao
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Chunsun Fan
- Qidong People's Hospital, Affiliated Qidong Hospital of Nantong University, Qidong Liver Cancer Institute, No.9, Seyuan Road, Chongchuan District, Nantong, Jiangsu, 226000, China
| | - Jian Fan
- Qidong People's Hospital, Affiliated Qidong Hospital of Nantong University, Qidong Liver Cancer Institute, No.9, Seyuan Road, Chongchuan District, Nantong, Jiangsu, 226000, China
| | - Li Zhu
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| | - Xueting Wang
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.
- Medical Research Center Affiliated Hospital 2 of Nantong University, Nantong, China.
| |
Collapse
|
13
|
Rajendran K, Krishnan UM. Mechanistic insights and emerging therapeutic stratagems for Alzheimer's disease. Ageing Res Rev 2024; 97:102309. [PMID: 38615895 DOI: 10.1016/j.arr.2024.102309] [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: 07/24/2023] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
Alzheimer's disease (AD), a multi-factorial neurodegenerative disorder has affected over 30 million individuals globally and these numbers are expected to increase in the coming decades. Current therapeutic interventions are largely ineffective as they focus on a single target. Development of an effective drug therapy requires a deep understanding of the various factors influencing the onset and progression of the disease. Aging and genetic factors exert a major influence on the development of AD. Other factors like post-viral infections, iron overload, gut dysbiosis, and vascular dysfunction also exacerbate the onset and progression of AD. Further, post-translational modifications in tau, DRP1, CREB, and p65 proteins increase the disease severity through triggering mitochondrial dysfunction, synaptic loss, and differential interaction of amyloid beta with different receptors leading to impaired intracellular signalling. With advancements in neuroscience tools, new inter-relations that aggravate AD are being discovered including pre-existing diseases and exposure to other pathogens. Simultaneously, new therapeutic strategies involving modulation of gene expression through targeted delivery or modulation with light, harnessing the immune response to promote clearance of amyloid deposits, introduction of stem cells and extracellular vesicles to replace the destroyed neurons, exploring new therapeutic molecules from plant, marine and biological sources delivered in the free state or through nanoparticles and use of non-pharmacological interventions like music, transcranial stimulation and yoga. Polypharmacology approaches involving combination of therapeutic agents are also under active investigation for superior therapeutic outcomes. This review elaborates on various disease-causing factors, their underlying mechanisms, the inter-play between different disease-causing players, and emerging therapeutic options including those under clinical trials, for treatment of AD. The challenges involved in AD therapy and the way forward have also been discussed.
Collapse
Affiliation(s)
- Kayalvizhi Rajendran
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India
| | - Uma Maheswari Krishnan
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India; School of Arts, Sciences, Humanities & Education, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India.
| |
Collapse
|
14
|
Chen Q, Zhu S, Shang J, Fang Q, Xue Q, Hua J. Trends in Cognitive Function Before and After Diabetes Onset: The China Health and Retirement Longitudinal Study. Neurology 2024; 102:e209165. [PMID: 38447106 DOI: 10.1212/wnl.0000000000209165] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/30/2023] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Individuals with prevalent diabetes were known to have a higher risk of dementia and lower cognitive function. However, trends of cognitive function before diabetes and in the short term after new-onset diabetes remain unclear. METHODS This study included participants without baseline diabetes from the China Health and Retirement Longitudinal Study. Cognitive tests were conducted at baseline (wave 1) and at least one time from wave 2 (2013) to wave 4 (2018). Cognitive function was assessed using a global cognition score which was the summary measure of 4 cognitive tests. A linear mixed model was constructed to fit the trends in cognitive function before and after diabetes onset and the trends among nondiabetes. The threshold of statistical significance was p < 0.05. RESULTS During the 7-year follow-up, 1,207 (9.7% of 12,422, 59.1 ± 8.6 years, 39.9% male participants) participants developed new-onset diabetes. The cognitive function of both the without diabetes group and the diabetes group declined annually during the follow-up. The annual decline rate of the diabetes group before diabetes onset was similar to that of the without diabetes group during the whole follow-up period. After diabetes onset, participants experienced statistically significant faster cognitive declines in global cognition (-0.023 SD/year; 95% CI -0.043 to -0.004; p = 0.019) and visuospatial abilities test (-0.036 SD/year; -0.061 to -0.011; p = 0.004), but not in tests of episodic memory (-0.018 SD/year; -0.041 to 0.004; p = 0.116), attention and calculation (-0.017 SD/year; -0.037 to 0.003; p = 0.090), or orientation (0.001 SD/year; -0.018 to 0.020; p = 0.894), compared with the cognitive slope before diabetes. In subgroup analysis, compared with those who developed diabetes between 45-54 years, those developing diabetes older (55-64 years, p for interaction = 0.701; 65-74 years, p for interaction = 0.996) did not demonstrate different rates of global cognitive decline after diabetes. DISCUSSION Individuals experienced faster rate of cognitive decline in a few years after diabetes onset, but not during the prediabetes period. Age did not modify the effect of diabetes on postdiabetes cognitive decline. Efforts in eliminating the adverse impacts on cognition should be started on diagnosis of diabetes.
Collapse
Affiliation(s)
- Qingmei Chen
- From the Departments of Physical Medicine and Rehabilitation (Q.C.), Psychiatry (J.S.), and Neurology (Q.F., Q.X., J.H.), The First Affiliated Hospital of Soochow University, Suzhou; Department of Neurology (S.Z.), The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital; and Department of Epidemiology and Biostatistics (J.H.), School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Sijia Zhu
- From the Departments of Physical Medicine and Rehabilitation (Q.C.), Psychiatry (J.S.), and Neurology (Q.F., Q.X., J.H.), The First Affiliated Hospital of Soochow University, Suzhou; Department of Neurology (S.Z.), The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital; and Department of Epidemiology and Biostatistics (J.H.), School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jing Shang
- From the Departments of Physical Medicine and Rehabilitation (Q.C.), Psychiatry (J.S.), and Neurology (Q.F., Q.X., J.H.), The First Affiliated Hospital of Soochow University, Suzhou; Department of Neurology (S.Z.), The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital; and Department of Epidemiology and Biostatistics (J.H.), School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Qi Fang
- From the Departments of Physical Medicine and Rehabilitation (Q.C.), Psychiatry (J.S.), and Neurology (Q.F., Q.X., J.H.), The First Affiliated Hospital of Soochow University, Suzhou; Department of Neurology (S.Z.), The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital; and Department of Epidemiology and Biostatistics (J.H.), School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Qun Xue
- From the Departments of Physical Medicine and Rehabilitation (Q.C.), Psychiatry (J.S.), and Neurology (Q.F., Q.X., J.H.), The First Affiliated Hospital of Soochow University, Suzhou; Department of Neurology (S.Z.), The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital; and Department of Epidemiology and Biostatistics (J.H.), School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jianian Hua
- From the Departments of Physical Medicine and Rehabilitation (Q.C.), Psychiatry (J.S.), and Neurology (Q.F., Q.X., J.H.), The First Affiliated Hospital of Soochow University, Suzhou; Department of Neurology (S.Z.), The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital; and Department of Epidemiology and Biostatistics (J.H.), School of Public Health, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
15
|
Rakshe PS, Dutta BJ, Chib S, Maurya N, Singh S. Unveiling the interplay of AMPK/SIRT1/PGC-1α axis in brain health: Promising targets against aging and NDDs. Ageing Res Rev 2024; 96:102255. [PMID: 38490497 DOI: 10.1016/j.arr.2024.102255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 03/17/2024]
Abstract
The escalating prevalence of neurodegenerative diseases (NDDs) within an aging global population presents a pressing challenge. The multifaceted pathophysiological mechanisms underlying these disorders, including oxidative stress, mitochondrial dysfunction, and neuroinflammation, remain complex and elusive. Among these, the AMPK/SIRT1/PGC-1α pathway emerges as a pivotal network implicated in neuroprotection against these destructive processes. This review sheds light on the potential therapeutic implications of targeting this axis, specifically emphasizing the promising role of flavonoids in mitigating NDD-related complications. Expanding beyond conventional pharmacological approaches, the exploration of non-pharmacological interventions such as exercise and calorie restriction (CR), coupled with the investigation of natural compounds, offers a beacon of hope. By strategically elucidating the intricate connections within these pathways, this review aims to pave the ways for novel multi-target agents and interventions, fostering a renewed optimism in the quest to combat and manage the debilitating impacts of NDDs on global health and well-being.
Collapse
Affiliation(s)
- Pratik Shankar Rakshe
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India
| | - Bhaskar Jyoti Dutta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India
| | - Shivani Chib
- Department of Pharmacology, Central University of Punjab, Badal - Bathinda Rd, Ghudda, Punjab, India
| | - Niyogita Maurya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India.
| |
Collapse
|
16
|
Wang H, Jayasankar N, Thamaraikani T, Viktor P, Mohany M, Al-Rejaie SS, Alammar HK, Anad E, Alhili F, Hussein SF, Amin AH, Lakshmaiya N, Ahsan M, Bahrami A, Akhavan-Sigari R. Quercetin modulates expression of serum exosomal long noncoding RNA NEAT1 to regulate the miR-129-5p/BDNF axis and attenuate cognitive impairment in diabetic mice. Life Sci 2024; 340:122449. [PMID: 38253310 DOI: 10.1016/j.lfs.2024.122449] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 01/24/2024]
Abstract
AIMS Cognitive impairment poses a considerable health challenge in the context of type 2 diabetes mellitus (T2DM), emphasizing the need for effective interventions. This study delves into the therapeutic efficacy of quercetin, a natural flavonoid, in mitigating cognitive impairment induced by T2DM in murine models. MATERIALS AND METHODS Serum exosome samples were obtained from both T2DM-related and healthy mice for transcriptome sequencing, enabling the identification of differentially expressed mRNAs and long noncoding RNAs (lncRNAs). Subsequent experiments were conducted to ascertain the binding affinity between mmu-miR-129-5p, NEAT1 and BDNF. The structural characteristics and dimensions of isolated exosomes were scrutinized, and the expression levels of exosome-associated proteins were quantified. Primary mouse hippocampal neurons were cultured for in vitro validation, assessing the expression of pertinent genes as well as neuronal vitality, proliferation, and apoptosis capabilities. For in vivo validation, a T2DM mouse model was established, and quercetin treatment was administered. Changes in various parameters, cognitive ability, and the expression of insulin-related proteins, along with pivotal signaling pathways, were monitored. KEY FINDINGS Analysis of serum exosomes from T2DM mice revealed dysregulation of NEAT1, mmu-miR-129-5p, and BDNF. In vitro investigations demonstrated that NEAT1 upregulated BDNF expression by inhibiting mmu-miR-129-5p. Overexpression of mmu-miR-129-5p or silencing NEAT1 resulted in the downregulation of insulin-related protein expression, enhanced apoptosis, and suppressed neuronal proliferation. In vivo studies validated that quercetin treatment significantly ameliorated T2DM-related cognitive impairment in mice. SIGNIFICANCE These findings suggest that quercetin holds promise in inhibiting hippocampal neuron apoptosis and improving T2DM-related cognitive impairment by modulating the NEAT1/miR-129-5p/BDNF pathway within serum exosomes.
Collapse
Affiliation(s)
- Hui Wang
- Department of Plastic Surgery, The Fourth Affiliated Hospital Zhejiang University School of Medicine, Yiwu 322000, China
| | - Narayanan Jayasankar
- Department of Pharmacology, SRM Institute of Science and Technology, SRM College of Pharmacy, Kattankulathur 603203, Tamil Nadu, India
| | - Tamilanban Thamaraikani
- Department of Pharmacology, SRM Institute of Science and Technology, SRM College of Pharmacy, Kattankulathur 603203, Tamil Nadu, India
| | - Patrik Viktor
- Keleti Károly Faculty of Business and Management, Óbuda University, Tavaszmező, H-1084 Budapest, Hungary
| | - Mohamed Mohany
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Salim S Al-Rejaie
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | | | - Enaam Anad
- Department of Medical Laboratory Technics, Al-Noor University College, Nineveh, Iraq
| | - Farah Alhili
- Medical Technical College, Al-Farahidi University, Iraq
| | - Sinan F Hussein
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Ali H Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Natrayan Lakshmaiya
- Department of Mechanical Engineering, Saveetha School of Engineering, SIMATS, Chennai, Tamil Nadu, India
| | - Muhammad Ahsan
- Department of Measurements and Control Systems, Silesian University of Technology, Gliwice, Poland; Joint Doctoral School, Silesian University of Technology, Akademicka 2A, Gliwice, Poland.
| | - Abolfazl Bahrami
- Biomedical Center for Systems Biology Science Munich, Ludwig-Maximilians-University, Munich, Germany.
| | - Reza Akhavan-Sigari
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw, Poland; Department of Neurosurgery, University Medical Center Tuebingen, Germany
| |
Collapse
|
17
|
Zhu YH, Hu P, Luo YX, Yao XQ. Knowledge mapping of trends and hotspots in the field of exercise and cognition research over the past decade. Aging Clin Exp Res 2024; 36:19. [PMID: 38308660 PMCID: PMC10838253 DOI: 10.1007/s40520-023-02661-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/09/2023] [Indexed: 02/05/2024]
Abstract
Exercise elicits a wide range of physiological responses in mammalian tissues that enhance a broad range of functions, particularly in improving cognitive performance. However, the field lacks a comprehensive bibliometric analysis that clarifies its knowledge structure and research hotspots. This study aims to address this gap and map the research landscape regarding the role of exercise in cognitive function enhancement. Firstly, the frequencies and co-occurrence of keywords were analysed to identify six main clusters: aging, cognitive impairment, rehabilitation, obesity, fatigue, and hippocampus. Secondly, reference timeline co-citation analysis revealed that hippocampus and aging were the major bursts with high intensity and long attention span while children had recently emerged as a topical subject. Finally, the evolution of themes from 2012 to 2022 was analysed, and found that older adults had been the leading research theme for exercise affecting cognition. Childhood obesity was an emerging theme that attracted increasing research attention in recent years while the hippocampus research theme expanded rapidly during the decade but remained a niche topic with less relevance to others. This research identified and summarised research priorities and evolutionary trends in exercise to improve cognition by constructing knowledge networks through visual analysis. It provides researchers with a comprehensive insight into the current state of the field to facilitate further research.
Collapse
Affiliation(s)
- Ying-Hai Zhu
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Hu
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ya-Xi Luo
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Xiu-Qing Yao
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Chongqing Municipality Clinical Research Center for Geriatric Medicine, Chongqing, China.
| |
Collapse
|
18
|
Tian N, Song L, Hou T, Fa W, Dong Y, Liu R, Ren Y, Liu C, Zhu M, Zhang H, Wang Y, Cong L, Du Y, Qiu C. Association of Triglyceride-Glucose Index With Cognitive Function and Brain Atrophy: A Population-Based Study. Am J Geriatr Psychiatry 2024; 32:151-162. [PMID: 37827915 DOI: 10.1016/j.jagp.2023.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 10/14/2023]
Abstract
OBJECTIVE To investigate the associations of triglyceride-glucose (TyG) index, a reliable surrogate marker for insulin resistance, with the function of various cognitive domains and brain structures among older adults. DESIGN A population-based cross-sectional study. SETTING Older adults living in the rural communities in China. PARTICIPANTS About 4,541 rural-dwelling dementia-free participants (age ≥65 years; 56.37% women) undertook examinations in March-September 2018 for MIND-China. MEASUREMENTS TyG index was calculated as ln[fasting triglyceride (mg/dL) × fasting glucose (mg/dL)/2]. A neuropsychological test battery was used to assess memory, attention, verbal fluency, and executive function. Volumetric brain measures were assessed on magnetic resonance imaging (MRI) in a subsample (n = 1,019). Data were analyzed with restricted cubic spline and multivariable general linear models. RESULTS An inverted J-shaped association was observed between TyG index and z-scores of multiple cognitive domains, such that among individuals with TyG index ≥8.57 (median), a higher TyG index was significantly associated with lower z-scores of memory, attention, verbal fluency, executive function, and global cognition (all p < 0.05); among people with TyG index <8.57, a higher TyG index was significantly associated with a higher executive function z-score (p < 0.05), but not with any of the other examined cognitive domains. In the MRI subsample, a higher TyG index was significantly associated with lower volumes of total brain tissue, gray matter, and white matter as well as greater cerebrospinal fluid volume (p < 0.05), but not with white matter hyperintensity volume. CONCLUSIONS Insulin resistance, as indicated by a high TyG index, was associated with poor function in multiple cognitive domains and global brain atrophy.
Collapse
Affiliation(s)
- Na Tian
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China; Medical Science and Technology Innovation Center (NT, YD), Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Lin Song
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Tingting Hou
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Wenxin Fa
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Yi Dong
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Rui Liu
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Yifei Ren
- Department of Neurology (YR, YD), Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Cuicui Liu
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Min Zhu
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Heng Zhang
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Yongxiang Wang
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China; Institute of Brain Science and Brain-Inspired Research (YW, YD), Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China; Aging Research Center and Center for Alzheimer Research (YW, CQ), Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet-Stockholm University, 17165 Solna, Sweden
| | - Lin Cong
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Yifeng Du
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China; Medical Science and Technology Innovation Center (NT, YD), Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China; Department of Neurology (YR, YD), Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China; Institute of Brain Science and Brain-Inspired Research (YW, YD), Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China.
| | - Chengxuan Qiu
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Aging Research Center and Center for Alzheimer Research (YW, CQ), Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet-Stockholm University, 17165 Solna, Sweden
| |
Collapse
|
19
|
Zhan M, Liu X, Xia X, Yang Y, Xie Y, Zhang L, Lin C, Zhu J, Ding W, Xu S. Promotion of neuroinflammation by the glymphatic system: a new insight into ethanol extracts from Alisma orientale in alleviating obesity-associated cognitive impairment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155147. [PMID: 37864890 DOI: 10.1016/j.phymed.2023.155147] [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: 08/27/2023] [Revised: 09/20/2023] [Accepted: 10/12/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Obesity is one of the critical risk factors for cognitive dysfunction. The glymphatic system (GS) plays a key role in the pathogenesis of cognitive deficits. Alisma orientale has been shown to have anti-inflammatory and antihyperlipidemic effects, whereas its effects and underlying mechanisms on obesity-associated cognitive impairment (OACI) are unclear. PURPOSE This work aims to decipher the mechanism of ethanol extracts from Alisma orientale (EEAO) in restoring cognitive impairment in HFD-induced obese mice through a GS approach. METHODS The restoration of abnormal glucose/lipid metabolism and excess adipose deposition by EEAO were assayed by biochemical analysis and visually displayed by a micro-CT scanner and Oil Red O staining. Biochemical assays and Western blotting (WB) were used to measure cerebral blood flow (CBF), free fatty acid (FFAs) levels and the structural integrity of the blood-brain barrier (BBB). Microglial activation and neuroinflammation were assessed with immunohistochemistry staining, ELISA and WB. Moreover, GS function was determined by immunofluorescence staining, fluorescence tracer imaging and WB. Finally, the neuropathological features and cognitive functions were detested with immunohistochemistry staining, immunofluorescence and Morris Water Maze. RESULTS EEAO not only alleviated body weight, cerebral lipid accumulation and serum FFAs in HFD-induced obese mice, but also increased CBF and BBB integrity. EEAO suppressed microglial activation and lipid deposition in the hippocampus and reduced the level of inflammatory cytokines including IL-6, IL-1β and TNF-α in brain tissue. Interestingly, long-term HFD-induced GS dysfunction was significantly restored after EEAO intervention, and neuropathological lesions and cognitive deficits were also markedly rescued. CONCLUSION EEAO rescued the cognitive deficits of OACI by inhibiting neuroinflammation and restoring GS dysfunction, indicating a potential remedy for OACI.
Collapse
Affiliation(s)
- Meng Zhan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiao Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiuwen Xia
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Youjun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ya Xie
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lu Zhang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chunqiao Lin
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiushuang Zhu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Weijun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Shijun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
20
|
Dutta BJ, Rakshe PS, Maurya N, Chib S, Singh S. Unlocking the therapeutic potential of natural stilbene: Exploring pterostilbene as a powerful ally against aging and cognitive decline. Ageing Res Rev 2023; 92:102125. [PMID: 37979699 DOI: 10.1016/j.arr.2023.102125] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/20/2023]
Abstract
The therapeutic potential of natural stilbenes, with a particular focus on pterostilbene (PTE), has emerged as a promising avenue of research targeting age-associated conditions encompassing cardiovascular diseases (CVD), diabetes mellitus (DM), and cognitive decline. This comprehensive investigation delves into the intricate mechanisms through which PTE, a polyphenolic compound abundant in grapes and blueberries, exerts its advantageous effects as an anti-aging agent. Central to its action is the modulation of hallmark aging processes, including oxidative damage, inflammatory responses, telomere attrition, and cellular senescence. PTE's ability to effectively penetrate the blood-brain barrier amplifies its potential for safeguarding neural health, thereby facilitating the regulation of neuronal signalling cascades, synaptic plasticity, and mitochondrial functionality. Through engagement with sirtuin proteins, it orchestrates cellular resilience, longevity, and metabolic equilibrium. Encouraging findings from preclinical studies portray PTE as a robust candidate for counteracting age-linked cognitive decline, augmenting memory consolidation, and potentially ameliorating neurodegenerative maladies such as Alzheimer's disease (AD). The synthesis of current scientific insights accentuates the promising translational prospects of PTE as a potent, naturally derived therapeutic agent against cognitive impairments associated with aging. Consequently, these collective findings lay a solid groundwork for forthcoming clinical inquiries and innovative therapeutic interventions in this realm.
Collapse
Affiliation(s)
- Bhaskar Jyoti Dutta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India
| | - Pratik Shankar Rakshe
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India
| | - Niyogita Maurya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India
| | - Shivani Chib
- Department of Pharmacology, Central University of Punjab, Badal - Bathinda Rd, Ghudda, Punjab, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India.
| |
Collapse
|
21
|
Li X, Gao Y, Li B, Zhao W, Cai Q, Yin W, Zeng S, Li X, Gao H, Cheng M. Integrated proteomics and metabolomics analysis of D-pinitol function during hippocampal damage in streptozocin-induced aging-accelerated mice. Front Mol Neurosci 2023; 16:1251513. [PMID: 38025258 PMCID: PMC10664147 DOI: 10.3389/fnmol.2023.1251513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Diabetes can cause hippocampal damage and lead to cognitive impairment. Diabetic cognitive impairment (DCI) is a chronic complication of diabetes associated with a high disability rate; however, its pathogenesis and therapeutic targets are unclear. We aimed to explore the mechanism of hippocampal damage during diabetes and evaluate the potential role of D-pinitol (DP) in protecting hippocampal tissue and improving cognitive dysfunction. Methods DP (150 mg/kg/day) was administered intragastrically to streptozocin-induced aging-accelerated mice for 8 weeks. Hippocampal tissues were examined using tandem mass tag (TMT)-based proteomics and liquid chromatography-mass spectrometry (LC-MS)/MS-based non-targeted metabolomic analysis. Differentially expressed proteins (DEPs) and differentially regulated metabolites (DRMs) were screened for further analysis, and some DEPs were verified using western blotting. Results Our results showed that 329 proteins had significantly altered hippocampal expression in untreated diabetic mice (DM), which was restored to normal after DP treatment in 72 cases. In total, 207 DRMs were identified in the DM group, and the expression of 32 DRMs was restored to normal post-DP treatment. These proteins and metabolites are involved in metabolic pathways (purine metabolism, arginine and proline metabolism, and histidine metabolism), actin cytoskeleton regulation, oxidative phosphorylation, and Rap1-mediated signaling. Conclusions Our study may help to better understand the mechanism of diabetic hippocampal damage and cognitive impairment and suggest a potential therapeutic target.
Collapse
Affiliation(s)
- Xiaoxia Li
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
- Department of Diabetes, The Third People's Hospital of Gansu Province, Lanzhou, China
| | - Yuan Gao
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Baoying Li
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Health Management Center (East Area), Qilu Hospital of Shandong University, Jinan, China
| | - Wenqian Zhao
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Qian Cai
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Wenbin Yin
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Shudong Zeng
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Xiaoli Li
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haiqing Gao
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Mei Cheng
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| |
Collapse
|
22
|
Mei M, Liu M, Mei Y, Zhao J, Li Y. Sphingolipid metabolism in brain insulin resistance and neurological diseases. Front Endocrinol (Lausanne) 2023; 14:1243132. [PMID: 37867511 PMCID: PMC10587683 DOI: 10.3389/fendo.2023.1243132] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Sphingolipids, as members of the large lipid family, are important components of plasma membrane. Sphingolipids participate in biological signal transduction to regulate various important physiological processes such as cell growth, apoptosis, senescence, and differentiation. Numerous studies have demonstrated that sphingolipids are strongly associated with glucose metabolism and insulin resistance. Insulin resistance, including peripheral insulin resistance and brain insulin resistance, is closely related to the occurrence and development of many metabolic diseases. In addition to metabolic diseases, like type 2 diabetes, brain insulin resistance is also involved in the progression of neurodegenerative diseases including Alzheimer's disease and Parkinson's disease. However, the specific mechanism of sphingolipids in brain insulin resistance has not been systematically summarized. This article reviews the involvement of sphingolipids in brain insulin resistance, highlighting the role and molecular biological mechanism of sphingolipid metabolism in cognitive dysfunctions and neuropathological abnormalities of the brain.
Collapse
Affiliation(s)
- Meng Mei
- Department of Pharmacy, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Maochang Liu
- Department of Pharmacy, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Mei
- Department of Pharmacy, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zhao
- Administrative Office, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Li
- Department of Pharmacy, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Rezaei MH, Madadizadeh E, Aminaei M, Abbaspoor M, Schierbauer J, Moser O, Khoramipour K, Chamari K. Leptin Signaling Could Mediate Hippocampal Decumulation of Beta-Amyloid and Tau Induced by High-Intensity Interval Training in Rats with Type 2 Diabetes. Cell Mol Neurobiol 2023; 43:3465-3478. [PMID: 37378849 PMCID: PMC11409991 DOI: 10.1007/s10571-023-01357-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/29/2023] [Indexed: 06/29/2023]
Abstract
Leptin (LEP) can cross the blood-brain barrier and facilitate cross-talk between the adipose tissue and central nerve system (CNS). This study aimed to investigate the effect of 8-week high-intensity interval training (HIIT) on the LEP signaling in the hippocampus of rats with type 2 diabetes. 20 rats were randomly divided into four groups: (i) control (Con), (ii) type 2 diabetes (T2D), (iii) exercise (EX), and (iv) type 2 diabetes + exercise (T2D + EX). The rats in the T2D and T2D + EX were fed a high-fat diet for two months, then a single dose of STZ (35 mg/kg) was injected to induce diabetes. The EX and T2D + EX groups performed 4-10 intervals of treadmill running at 80-100% of Vmax. Serum and hippocampal levels of LEP as well as hippocampal levels of LEP receptors (LEP-R), Janus kinase 2 (JAK-2), signal transducer and activator of transcription 3 (STAT-3), activated protein kinase (AMP-K), proxy zoster receptor α (PGC-1α), beta-secretase 1 (BACE1), Beta-Amyloid (Aβ), Phosphoinositide 3-kinases (PI3K), protein kinase B (AKT), mammalian target of rapamycin (mTOR), Glycogen Synthase Kinase 3 Beta (GSK3β), and hyperphosphorylated tau proteins (TAU) were measured. One-way ONOVA and Tukey post-hoc tests were used to analyze the data. Serum and hippocampal levels of LEP as well as hippocampal levels of LEP-R, JAK-2, STAT-3, AMP-K, PGC1α, PI3K, AKT, and mTOR were increased while hippocampal levels of BACE1, GSK3B, TAU, and Aβ were decreased in T2D + EX compared with T2D group. Serum LEP and hippocampal levels of LEP, LEP-R, JAK-2, STAT-3, AMP-K, PGC1α, PI3K, AKT, and mTOR were decreased. Conversely hippocampal levels of BACE1, GSK3B, TAU, and Aβ were increased in T2D group compared with CON group. HIIT could improve LEP signaling in the hippocampus of rats with type 2 diabetes and decrease the accumulation of Tau and Aβ, which may reduce the risk of memory impairments.
Collapse
Affiliation(s)
- Maryam Hossein Rezaei
- Department of Exercise Physiology, Faculty of Physical Education, Shahid Bahonar University, Kerman, Iran
| | - Elham Madadizadeh
- Department of Exercise Physiology, Faculty of Physical Education, Shahid Bahonar University, Kerman, Iran
| | - Mohsen Aminaei
- Department of Exercise Physiology, Faculty of Physical Education, Shahid Bahonar University, Kerman, Iran
| | - Mehdi Abbaspoor
- Department of Exercise Physiology, Faculty of Physical Education, Shahid Bahonar University, Kerman, Iran
| | - Janis Schierbauer
- Exercise Physiology and Metabolism (Sports Medicine), BaySpo-Bayreuth Centre of Sports Science, University of Bayreuht, Bayreuth, Germany
| | - Othmar Moser
- Exercise Physiology and Metabolism (Sports Medicine), BaySpo-Bayreuth Centre of Sports Science, University of Bayreuht, Bayreuth, Germany
- Interdisciplinary Metabolic Medicine Trials Unit, Medical University of Graz, Graz, Austria
| | - Kayvan Khoramipour
- Neuroscience Research Center, Institute of Neuropharmacology and Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Karim Chamari
- Aspetar Qatar Orthopaedic and Sports Medicine Hospital, Doha, Qatar
| |
Collapse
|
24
|
Chao Y, Gu T, Zhang Z, Wu T, Wang J, Bi Y. The role of miRNAs carried by extracellular vesicles in type 2 diabetes and its complications. J Diabetes 2023; 15:838-852. [PMID: 37583355 PMCID: PMC10590682 DOI: 10.1111/1753-0407.13456] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 07/30/2023] [Indexed: 08/17/2023] Open
Abstract
Diabetes poses severe global public health problems and places heavy burdens on the medical and economic systems of society. Type 2 diabetes (T2D) accounts for 90% of these cases. Diabetes also often accompanies serious complications that threaten multiple organs such as the brain, eyes, kidneys, and the cardiovascular system. MicroRNAs (miRNAs) carried by extracellular vesicles (EV-miRNAs) are considered to mediate cross-organ and cross-cellular communication and have a vital role in the pathophysiology of T2D. They also offer promising sources of diabetes-related biomarkers and serve as effective therapeutic targets. Here, we briefly reviewed studies of EV-miRNAs in T2D and related complications. Specially, we innovatively explore the targeting nature of miRNA action due to the target specificity of vesicle binding, aiding mechanism understanding as well as the detection and treatment of diseases.
Collapse
Affiliation(s)
- Yining Chao
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Branch of National Clinical Research Centre for Metabolic DiseasesNanjingChina
| | - Tianwei Gu
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Branch of National Clinical Research Centre for Metabolic DiseasesNanjingChina
| | - Zhou Zhang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Branch of National Clinical Research Centre for Metabolic DiseasesNanjingChina
| | - Tianyu Wu
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Branch of National Clinical Research Centre for Metabolic DiseasesNanjingChina
| | - Jin Wang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Branch of National Clinical Research Centre for Metabolic DiseasesNanjingChina
| | - Yan Bi
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Branch of National Clinical Research Centre for Metabolic DiseasesNanjingChina
| |
Collapse
|
25
|
Guo Y, Liang R, Ren J, Cheng L, Wang M, Chai H, Cheng X, Yang Y, Sun Y, Li J, Zhao S, Hou W, Zhang J, Liu F, Wang R, Niu Q, Yu H, Yang S, Bai J, Zhang H, Qin X, Xia N. Cognitive status and its risk factors in patients with hypertension and diabetes in a low-income rural area of China: A cross-sectional study. Int J Geriatr Psychiatry 2023; 38:e6010. [PMID: 37794769 DOI: 10.1002/gps.6010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 09/25/2023] [Indexed: 10/06/2023]
Abstract
OBJECTIVES The proportion of older people with dementia in China is gradually increasing with the increase in the aging population over recent years. Hypertension and diabetes are common non-communicable diseases among rural populations in China. However, it remains unclear whether these conditions affect the occurrence and development of cognitive impairment as there is limited research on cognitive status and its risk factors among residents of rural areas. METHODS A multi-stage stratified cluster random sampling method was used to select 5400 participants from rural permanent residents. A self-designed structured questionnaire was used to investigate demographic data of the participants. Cognitive function was assessed using the Montreal Cognitive Function Assessment Scale (MoCA). The results were analyzed using chi-square test, ANOVA and multiple linear regression analysis. RESULTS A total of 5028 participants returned the survey, giving a response rate of 93.1%. Higher education (odds ratio (OR) = 3.2, 95% confidence interval (CI) 2.87-3.54, p < 0.001), higher income (OR = 1.61, 95% CI 1.16-2.07, p < 0.001), and dietary control (OR = 0.66, 95%CI 0.34-0.98, p < 0.001) were protective factors. A visual representation of the relationship between annual income and MoCA score showed an inverted U-curve, the group with an annual income of 6000-7999 RMB had a maximum OR of 1.93 (95%CI 0.12-2.74, p < 0.001). While difficulty in maintaining sleep were risk factors for cognitive impairment (OR = -2.28, 95% CI-4.18-0.39, p = 0.018). CONCLUSIONS Participants with middle incomes had better cognitive status than those with the highest incomes. Higher education, proper diet control and good sleep are beneficial to the cognitive status of residents in rural areas.
Collapse
Affiliation(s)
- Yuyan Guo
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Ruifeng Liang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Jingjuan Ren
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Liting Cheng
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China
- Jinzhong Center for Disease Control and Prevention, Health Commission of Shanxi Province, Jinzhong, China
| | - Mengqin Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Huilin Chai
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Xiaoyu Cheng
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Yaowen Yang
- Health Commission Supervision & Inspection Center, Health Commission of Shanxi Province, Taiyuan, China
| | - Yajuan Sun
- Evaluation Center for Medical Service and Administration, Health Commission of Shanxi Province, Taiyuan, China
| | - Jiantao Li
- Department of Health Economics, School of Management, Shanxi Medical University, Taiyuan, China
| | - Shuhong Zhao
- Evaluation Center for Medical Service and Administration, Health Commission of Shanxi Province, Taiyuan, China
| | - Wenjing Hou
- Evaluation Center for Medical Service and Administration, Health Commission of Shanxi Province, Taiyuan, China
| | - Jianhua Zhang
- Health Commission and Sports Bureau of Yangqu County, Taiyuan, China
| | - Feng Liu
- Yangqu People's Hospital, Taiyuan, China
| | - Rong Wang
- Yangqu People's Hospital, Taiyuan, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China
| | - Hongmei Yu
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Shoulin Yang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Jianying Bai
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Hongmei Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Xiaojiang Qin
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Na Xia
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
26
|
Ran Q, Tian H, Lin J, Wang H, Wang B, Chen Z, Song D, Gong C. Mesenchymal Stem Cell-Derived Exosomes: A Novel Approach to Diabetes-Associated Cognitive Impairment. J Inflamm Res 2023; 16:4213-4228. [PMID: 37753267 PMCID: PMC10519429 DOI: 10.2147/jir.s429532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023] Open
Abstract
The progression of diabetes frequently results in a myriad of neurological disorders, including ischemic stroke, depression, blood-brain barrier impairment, and cognitive dysfunction. Notably, diabetes-associated cognitive impairment, a prevalent comorbidity during the course of diabetes, progressively affects patients' cognitive abilities and may reciprocally influence diabetes management, thereby severely impacting patients' quality of life. Extracellular vesicles, particularly nanoscale exosomes, have garnered considerable attention in recent years. These exosomes carry and transfer various functional molecules, such as proteins, lipids, and diverse non-coding RNAs, serving as novel regulators and communicators in intercellular interactions. Of particular interest, mesenchymal stem cell-derived exosomes (MSC-Exos) have been reported to traverse the blood-brain barrier and ameliorate intracerebral pathologies. This review elucidates the role of MSC-Exos in diabetes-related cognitive impairment, with a focus on their applications as biomarkers, modulation of neuronal regeneration and synaptic plasticity, anti-inflammatory properties, antioxidative effects, and their involvement in regulating the functionality of β-amyloid proteins during the course of cognitive impairment. The immense therapeutic potential of MSC-Exos in the treatment of diabetes-induced cognitive dysfunction is emphasized.
Collapse
Affiliation(s)
- Qingsen Ran
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| | - He Tian
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| | - Jian Lin
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| | - Han Wang
- Department of Gastroenterology, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| | - Bo Wang
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| | - Zhixin Chen
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| | - Da Song
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| | - Chunzhu Gong
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| |
Collapse
|
27
|
Tian Y, Jing G, Zhang M. Insulin-degrading enzyme: Roles and pathways in ameliorating cognitive impairment associated with Alzheimer's disease and diabetes. Ageing Res Rev 2023; 90:101999. [PMID: 37414154 DOI: 10.1016/j.arr.2023.101999] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/12/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Accumulation of amyloid-β in the central nervous system is a common feature of Alzheimer's disease (AD) and diabetes-related cognitive impairment. Since the insulin-degrading enzyme (IDE) can break down amyloid-β plaques, there is considerable interest in using this enzyme to treat both neurological disorders. In this review, we have summarized the pre-clinical and clinical research on the potential application of IDE for the improvement of cognitive impairment. Furthermore, we have presented an overview of the main pathways that can be targeted to mitigate the progression of AD and the cognitive impairment caused by diabetes.
Collapse
Affiliation(s)
- Yue Tian
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Guangchan Jing
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Mengren Zhang
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
28
|
Little K, Singh A, Del Marco A, Llorián-Salvador M, Vargas-Soria M, Turch-Anguera M, Solé M, Bakker N, Scullion S, Comella JX, Klaassen I, Simó R, Garcia-Alloza M, Tiwari VK, Stitt AW. Disruption of cortical cell type composition and function underlies diabetes-associated cognitive decline. Diabetologia 2023; 66:1557-1575. [PMID: 37351595 PMCID: PMC10317904 DOI: 10.1007/s00125-023-05935-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/28/2023] [Indexed: 06/24/2023]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes is associated with increased risk of cognitive decline although the pathogenic basis for this remains obscure. Deciphering diabetes-linked molecular mechanisms in cells of the cerebral cortex could uncover novel therapeutic targets. METHODS Single-cell transcriptomic sequencing (scRNA-seq) was conducted on the cerebral cortex in a mouse model of type 2 diabetes (db/db mice) and in non-diabetic control mice in order to identify gene expression changes in distinct cell subpopulations and alterations in cell type composition. Immunohistochemistry and metabolic assessment were used to validate the findings from scRNA-seq and to investigate whether these cell-specific dysfunctions impact the neurovascular unit (NVU). Furthermore, the behavioural and cognitive alterations related to these dysfunctions in db/db mice were assessed via Morris water maze and novel object discrimination tests. Finally, results were validated in post-mortem sections and protein isolates from individuals with type 2 diabetes. RESULTS Compared with non-diabetic control mice, the db/db mice demonstrated disrupted brain function as revealed by losses in episodic and spatial memory and this occurred concomitantly with dysfunctional NVU, neuronal circuitry and cerebral atrophy. scRNA-seq of db/db mouse cerebral cortex revealed cell population changes in neurons, glia and microglia linked to functional regulatory disruption including neuronal maturation and altered metabolism. These changes were validated through immunohistochemistry and protein expression analysis not just in the db/db mouse cerebral cortex but also in post-mortem sections and protein isolates from individuals with type 2 diabetes (74.3 ± 5.5 years) compared with non-diabetic control individuals (87.0 ± 8.5 years). Furthermore, metabolic and synaptic gene disruptions were evident in cortical NVU cell populations and associated with a decrease in vascular density. CONCLUSIONS/INTERPRETATION Taken together, our data reveal disruption in the cellular and molecular architecture of the cerebral cortex induced by diabetes, which can explain, at least in part, the basis for progressive cognitive decline in individuals with type 2 diabetes. DATA AVAILABILITY The single-cell sequencing data that supports this study are available at GEO accession GSE217665 ( https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE217665 ).
Collapse
Affiliation(s)
- Karis Little
- The Wellcome‑Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Aditi Singh
- The Wellcome‑Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Angel Del Marco
- Division of Physiology, School of Medicine, University of Cadiz, Cadiz, Spain
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| | - María Llorián-Salvador
- The Wellcome‑Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, Northern Ireland, UK
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Diabetes and Metabolism Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron University Hospital, Barcelona, Spain
| | - Maria Vargas-Soria
- Division of Physiology, School of Medicine, University of Cadiz, Cadiz, Spain
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| | - Mireia Turch-Anguera
- Diabetes and Metabolism Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron University Hospital, Barcelona, Spain
- Cell Signaling and Apoptosis Group, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
- Departament de Bioquímica i Biologia Molecular i Institut de Neurociències, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- Centro de Investigación en Red en Enfermedades Neurodegenerativas (CIBERNED - ISCII), Madrid, Spain
| | - Montse Solé
- Diabetes and Metabolism Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron University Hospital, Barcelona, Spain
- Cell Signaling and Apoptosis Group, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
- Departament de Bioquímica i Biologia Molecular i Institut de Neurociències, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- Centro de Investigación en Red en Enfermedades Neurodegenerativas (CIBERNED - ISCII), Madrid, Spain
| | - Noëlle Bakker
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Sarah Scullion
- The Wellcome‑Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Joan X Comella
- Diabetes and Metabolism Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron University Hospital, Barcelona, Spain
- Cell Signaling and Apoptosis Group, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
- Departament de Bioquímica i Biologia Molecular i Institut de Neurociències, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- Centro de Investigación en Red en Enfermedades Neurodegenerativas (CIBERNED - ISCII), Madrid, Spain
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Rafael Simó
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Diabetes and Metabolism Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron University Hospital, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM-ISCIII), Madrid, Spain
| | - Monica Garcia-Alloza
- Division of Physiology, School of Medicine, University of Cadiz, Cadiz, Spain.
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain.
| | - Vijay K Tiwari
- The Wellcome‑Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, Northern Ireland, UK.
- Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark.
- Danish Institute for Advanced Study (DIAS), Odense M, Denmark.
- Department of Clinical Genetics, Odense University Hospital, Odense C, Denmark.
| | - Alan W Stitt
- The Wellcome‑Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, Northern Ireland, UK.
| |
Collapse
|
29
|
Shin YK, Seol GH. Effects of linalyl acetate on oxidative stress, inflammation and endothelial dysfunction: can linalyl acetate prevent mild cognitive impairment? Front Pharmacol 2023; 14:1233977. [PMID: 37576815 PMCID: PMC10416234 DOI: 10.3389/fphar.2023.1233977] [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: 06/03/2023] [Accepted: 07/21/2023] [Indexed: 08/15/2023] Open
Abstract
Mild cognitive impairment (MCI) is a major public health challenge with an increasing prevalence. Although the mechanisms underlying the development of MCI remain unclear, MCI has been reported to be associated with oxidative stress, inflammatory responses, and endothelial dysfunction, suggesting that agents that reduce these factors may be key to preventing MCI. Currently, no agents have been approved for the treatment of MCI, with the efficacy of commonly prescribed cholinesterase inhibitors remaining unclear. Relatively safe natural products that can prevent the development of MCI are of great interest. Linalyl acetate (LA), the major component of clary sage and lavender essential oils, has been shown to have a variety of pharmacological effects, including anti-hypertensive, anti-diabetic, neuroprotective, anti-inflammatory, and antioxidant properties, which may have the potential for the prevention of MCI. The present review briefly summarizes the pathogenesis of MCI related to oxidative stress, inflammatory responses, and endothelial dysfunction as well as the benefits of LA against these MCI-associated factors. The PubMed and Google Scholar databases were used to search the relevant literature. Further clinical research may lead to the development of new strategies for preventing MCI, particularly in high-risk populations with oxidative stress, inflammatory responses, and endothelial dysfunction (e.g., patients with hypertension and/or diabetes mellitus).
Collapse
Affiliation(s)
- You Kyoung Shin
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea
| | - Geun Hee Seol
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea
- BK21 FOUR Program of Transdisciplinary Major in Learning Health Systems, Graduate School, Korea University, Seoul, Republic of Korea
| |
Collapse
|
30
|
Hajizadeh Tekmeh H, Vanizor Kural B, Kör S, Arıkan Malkoç M, Yuluğ E, Kutlu A, Abidin İ, Orem A. How does l-theanine treatment affect the levels of serum and hippocampal BDNF, insulin and adipocytokines in diabetic rats? Biochem Biophys Res Commun 2023; 667:95-103. [PMID: 37210871 DOI: 10.1016/j.bbrc.2023.05.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 05/13/2023] [Indexed: 05/23/2023]
Abstract
BACKGROUND Diabetes Mellitus (DM), a metabolic disease characterized by the increased blood glucose level, insulin deficiency or ineffectiveness, may cause structural and functional disorders in the brain. l-Theanine (LTN) has the relaxing, psychoactive, antidepressant, anti-inflammatory and antinecrotic properties, and regulates the functions of hippocampus (HP) in brain. In the present study, the aim was to identify the effects LTN on the levels of BDNF, insulin and adipocytokines (TNF-α, leptin, adiponectin and resistin) in both HP and serum of diabetic rats. METHODS 32 male Wistar rats were divided into four groups (n = 8/group): Control, LTN, DM and DM + LTN. Diabetes was induced by by nicotinamide/streptozotocin. 200 mg/kg/day LTN treatment was applied for 28 days. The serum and hippocampal levels of the parameters were determined by using commercial ELISA kits. Additionally, HP tissues examined histopathologically. RESULTS LTN treatment significantly decreased leptin and adiponectin levels in HP tissues in diabetic rats (p < 0.05). Although it decreased the insulin level in both serum and HP, this was not statistically significant. No significant effect on other parameters was observed (p > 0.05). In histopathological analysis, although the damage was reduced by LTN in all sections of HP, this change was significant mainly in CA3 region (p < 0.05). CONCLUSION It was concluded that LTN has the ability to reduce hippocampal degeneration and modulates adipocytokines in diabetic rats.
Collapse
Affiliation(s)
- Hamed Hajizadeh Tekmeh
- Department of Medical Biochemistry, Graduate School of Health Sciences, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Birgul Vanizor Kural
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Sevil Kör
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Meltem Arıkan Malkoç
- Vocational School of Health Sciences, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Esin Yuluğ
- Department of Histology and Embryology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Abdulkadir Kutlu
- Department of Histology and Embryology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - İsmail Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Asım Orem
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| |
Collapse
|
31
|
Li H, Ren J, Li Y, Wu Q, Wei J. Oxidative stress: The nexus of obesity and cognitive dysfunction in diabetes. Front Endocrinol (Lausanne) 2023; 14:1134025. [PMID: 37077347 PMCID: PMC10107409 DOI: 10.3389/fendo.2023.1134025] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/02/2023] [Indexed: 04/05/2023] Open
Abstract
Obesity has been associated with oxidative stress. Obese patients are at increased risk for diabetic cognitive dysfunction, indicating a pathological link between obesity, oxidative stress, and diabetic cognitive dysfunction. Obesity can induce the biological process of oxidative stress by disrupting the adipose microenvironment (adipocytes, macrophages), mediating low-grade chronic inflammation, and mitochondrial dysfunction (mitochondrial division, fusion). Furthermore, oxidative stress can be implicated in insulin resistance, inflammation in neural tissues, and lipid metabolism disorders, affecting cognitive dysfunction in diabetics.
Collapse
Affiliation(s)
- Huimin Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Ren
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Yusi Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Wu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junping Wei
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
32
|
Shpakov AO, Zorina II, Derkach KV. Hot Spots for the Use of Intranasal Insulin: Cerebral Ischemia, Brain Injury, Diabetes Mellitus, Endocrine Disorders and Postoperative Delirium. Int J Mol Sci 2023; 24:3278. [PMID: 36834685 PMCID: PMC9962062 DOI: 10.3390/ijms24043278] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
A decrease in the activity of the insulin signaling system of the brain, due to both central insulin resistance and insulin deficiency, leads to neurodegeneration and impaired regulation of appetite, metabolism, endocrine functions. This is due to the neuroprotective properties of brain insulin and its leading role in maintaining glucose homeostasis in the brain, as well as in the regulation of the brain signaling network responsible for the functioning of the nervous, endocrine, and other systems. One of the approaches to restore the activity of the insulin system of the brain is the use of intranasally administered insulin (INI). Currently, INI is being considered as a promising drug to treat Alzheimer's disease and mild cognitive impairment. The clinical application of INI is being developed for the treatment of other neurodegenerative diseases and improve cognitive abilities in stress, overwork, and depression. At the same time, much attention has recently been paid to the prospects of using INI for the treatment of cerebral ischemia, traumatic brain injuries, and postoperative delirium (after anesthesia), as well as diabetes mellitus and its complications, including dysfunctions in the gonadal and thyroid axes. This review is devoted to the prospects and current trends in the use of INI for the treatment of these diseases, which, although differing in etiology and pathogenesis, are characterized by impaired insulin signaling in the brain.
Collapse
Affiliation(s)
- Alexander O. Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| | | | | |
Collapse
|
33
|
Calluna vulgaris Crude Extract Reverses Liver Steatosis and Insulin Resistance-Associated-Brain Lesion Induced by CCl4 Administration. SEPARATIONS 2023. [DOI: 10.3390/separations10020094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Fatty liver (FL) is one of the most prevalent diseases in the world, characterized by insulin resistance and hyperlipidemia, which consequently lead to neurodegenerative disorders through the induction of oxidative stress-inflammatory axis, which alters the neurotransmitters’ levels. Calluna vulgaris (CV), also known as heather, has anti-inflammatory and antidepressant properties, making it a promising candidate for treating steatosis and brain lesions. This study aimed to assess the prophylactic and therapeutic effect of CV extract on brain dysfunction associated with steatosis. FL was induced in rats by CCl4 oral administration (50 µL/Kg in olive oil three times/week) for six weeks. The protection group received 200 mg/kg CV extract orally for two weeks before and two weeks during FL induction, while the treatment group was orally administered CV extract after FL induction for one month. The biochemical parameters revealed that CCl4 administration induced hepatotoxicity as blood-liver function parameters (AST, ALT, ALP, protein, and LDH) were increased by 1.8, 1.4, 2, 2.4, and 1.2-fold, respectively. Moreover, insulin resistance was characterized by a two-fold increase in the glucose, insulin, and lipid profile when compared to control one, at p < 0.05. Steatosis liver demonstrated a two-fold increase in all following parameters— acetaldehyde (AC), prooxidant (TBARS), acetylcholine esterase (AChE), monoamine oxidase (MAO), hyaluronidase, and ATPase—when compared to control one, at p < 0.05. CCl4 administration led to brain lesions where the brain level of TBARS, insulin, cholesterol, AChE, and MAO was progressively increased by 2, 1.6, 2.2, 4, and 1.6-fold, respectively, that was associated with reduced glucose (8-fold) and GSH (2-fold) than that of control level, at p < 0.05. CV extract as a prophylactic and therapeutic agent increased GSH and decreased TBARS of both the liver and brain than that of induced group, at p < 0.05, normalized the activities of AChE and MAO, and increased insulin sensitivity where they successfully decreased the HOMA-IR, glucose, TG, and cholesterol compared to than that of induced group, at p < 0.05. This positive effect of CV extract contributed to the presence of polyphenolic compounds such as catechins (5.501 ± 0.056 µg/g extract), gallic (3.525 ± 0.143 µg/g) extract, and protocatechuic acid (2.719 ± 0.132 µg/g extract). Therefore, we concluded that FL induced brain dysfunction through the formation of ROS and elevation of insulin and lipid inside the brain tissue, which alter the amount of neurotransmitter and cellular energy production. Rich in polyphenolic compounds, CV extract functions as an antioxidant, antidiabetic, hepatoprotective, inhibitor of neurotransmitter catabolizing enzymes, and a regulator for energy production. Therefore, it can be used as a preventative or treatment for NAFLD and brain damage.
Collapse
|
34
|
Ding H, Liu C, Zhang S, Li B, Xu Q, Shi B, Li S, Dong S, Ma X, Zhang Y, Zhong M, Zhang G. Sleeve gastrectomy attenuated diabetes-related cognitive decline in diabetic rats. Front Endocrinol (Lausanne) 2022; 13:1015819. [PMID: 36407319 PMCID: PMC9669300 DOI: 10.3389/fendo.2022.1015819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To investigate the effects of sleeve gastrectomy (SG) on diabetes-related cognitive decline (DCD) in rats with diabetic mellitus (DM). METHODS AND METHODS Forty Wistar rats were randomly divided into control (CON) group (n=10), diabetes mellitus (DM) group (n=10), sham operation (SHAM) group (n=10) and SG group (n=10). DM model was established by high-fat diet (HFD) combined with intraperitoneal injection of streptozocin (STZ). Behavioral evaluation was given using Morris water maze test and Y-maze. In addition, PET-CT, TUNEL assay, histological analysis, transmission electron microscopy (TEM), immunohistochemistry (IHC) and Western blot analysis were used to evaluate the alleviating effects and potential mechanisms of SG on DCD in DM rats. RESULTS Compared with the sham group, SG induced significant improvement in the metabolic indices such as blood glucose and body weight. Besides, it could attenuate the insulin resistance compared with SHAM group. In addition, SG could improve the cognitive function of DM rats, which were featured by significant decrease in the escape latency (P<0.05), and significant increase in the time in target quadrant and platform crossings (P<0.05) compared with the SHAM group. SG induced significant elevation in the spontaneous alternation compared with SHAM group (P<0.05). Moreover, SG could improve the arrangement and biosynthesis of hippocampus neuron. Moreover, SG triggered the inhibition of apoptosis of hippocampus neurons, and Western blot analysis showed SG induced significant increase in the ratios of Bcl-2/Bax and Caspase3/cleaved Caspase 3. TEM demonstrated SG could significantly improve the microstructure of hippocampus neurons compared with the SHAM group. Western blot and IHC confirmed the significant decrease in the phosphorylation of tau at Ser404 and Ser396 sites in the SG group. Furthermore, SG activated the PI3K signaling pathway by elevating the phosphorylation of PI3K and Akt and GSK3β compared with the SHAM group. CONCLUSION SG attenuated the DCD in DM rats, which may be related to the activation of PI3K signaling pathway.
Collapse
Affiliation(s)
- Huanxin Ding
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuxuan Liu
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuo Zhang
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bingjun Li
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Qian Xu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Bowen Shi
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Songhan Li
- Department of Breast Disease, Peking University People’s Hospital, Beijing, China
| | - Shuohui Dong
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaomin Ma
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Yun Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Mingwei Zhong
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Guangyong Zhang
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Guangyong Zhang,
| |
Collapse
|