1
|
Alami M, Morvaridzadeh M, El Khayari A, Boumezough K, El Fatimy R, Khalil A, Fulop T, Berrougui H. Reducing Alzheimer's disease risk with SGLT2 inhibitors: From glycemic control to neuroprotection. Ageing Res Rev 2025; 108:102751. [PMID: 40204129 DOI: 10.1016/j.arr.2025.102751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
Recent research has established a strong link between metabolic abnormalities and an increased risk of dementia. In parallel, there is growing epidemiological evidence supporting the neuroprotective effects of antidiabetic medications against cognitive impairments. Among these, sodium-glucose co-transporter (SGLT2) inhibitors have emerged as pharmacological candidates with promising potential in alleviating the burden of age-related diseases, particularly neurodegenerative diseases (NDD). SGLT2 inhibitor therapies are FDA-approved medications routinely prescribed to manage diabetes. This novel class was initially developed to address cardiovascular disorders and to reduce the risk of hypoglycemia associated with insulin-secretagogue agents. It subsequently attracted growing interest for its beneficial effects on central nervous system (CNS) disorders. However, the molecular mechanisms through which these glucose-lowering therapies mitigate cognitive decline and limit the progression of certain brain degenerative diseases remain largely unexplored. Consequently, the neuroscientific community needs further studies that gather, analyze, and critically discuss the available mechanistic evidence regarding the neuroprotective effects of SGLT2 inhibitors. This review aims to critically examine the most relevant published findings, both in vitro and in vivo, as well as human studies evaluating the impact of SGLT2 inhibitors exposure on Alzheimer's disease (AD). It seeks to integrate the current understanding of their beneficial effects at the molecular level and their role in addressing the pathophysiology and neuropathology of AD. These insights will help extend our knowledge of how SGLT2 inhibitor therapies are associated with reduced risk of dementia and thus shed light on the link between diabetes and AD.
Collapse
Affiliation(s)
- Mehdi Alami
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology, Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Mojgan Morvaridzadeh
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Abdellatif El Khayari
- Faculty of Medical Sciences, UM6P Hospitals, Mohammed VI Polytechnic University, Ben-Guerir 43150, Morocco; Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kaoutar Boumezough
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology, Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Rachid El Fatimy
- Faculty of Medical Sciences, UM6P Hospitals, Mohammed VI Polytechnic University, Ben-Guerir 43150, Morocco
| | - Abdelouahed Khalil
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Tamas Fulop
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Hicham Berrougui
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology, Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada.
| |
Collapse
|
2
|
Abdel-Lah ES, Sherkawy HS, Mohamed WH, Fawy MA, Hasan AA, Muhammed AA, Taha AF, Tony AA, Hamad N, Gamea MG. Empagliflozin and memantine combination ameliorates cognitive impairment in scopolamine + heavy metal mixture-induced Alzheimer's disease in rats: role of AMPK/mTOR, BDNF, BACE-1, neuroinflammation, and oxidative stress. Inflammopharmacology 2025:10.1007/s10787-025-01755-5. [PMID: 40325262 DOI: 10.1007/s10787-025-01755-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 04/02/2025] [Indexed: 05/07/2025]
Abstract
One of the major consequences of diabetes mellitus that has gained attention due to its rising incidence is cognitive impairment. Recent research suggested that sodium-glucose cotransporter-2 (SGLT-2) inhibitors can mitigate memory impairment linked to Alzheimer's disease and are now being explored for their cognitive benefits. However, their mechanisms were not thoroughly studied. This research investigates the hypothesis of the neuroprotective effect of empagliflozin administration against scopolamine-heavy metal mixture (SCO + HMM)-treated Alzheimer's rat models in comparison with memantine as a reference drug and the impact of their combination. Yet, the neuroprotective effects of memantine and empagliflozin combination against cognitive impairment have not been previously explored. This study employed adult male albino rats categorized into five groups. The impact of empagliflozin, memantine, and their concomitant administration on cognitive performance was assessed in a scopolamine and heavy metal mixture-treated Alzheimer's disease model in rats. The assessment of rats' cognitive behavior, memory, and spatial learning was conducted, followed by an evaluation of hippocampal brain-derived neurotrophic factor (BDNF), beta-secretase (BACE-1), oxidative stress (OS), and inflammatory marker activity. And, a western blot analysis was conducted to detect phosphorylated 5' AMP-activated protein kinase (p-AMPK), phosphorylated mammalian target of rapamycin (p-mTOR), and heme oxygenase-1 (HO-1). Hippocampal and cerebellar histopathology were thoroughly examined, in addition to the expressions of amyloid β (Aβ). The current data demonstrate the involvement of the pAMPK/mTOR/HO-1 signaling pathway in empagliflozin neuroprotection against SCO + HMM-induced AD. In addition, it reduces AD hallmarks (Aβ and BACE1), neuro-inflammation, and oxidative stress sequelae, and enhances neurogenesis and synaptic density via BDNF. This study proposes that EMPA, especially when co-administered with other conventional anti-Alzheimer therapy, may be formulated into an innovative therapeutic strategy for the enhancement of cognitive impairments associated with neurodegenerative disorders.
Collapse
Affiliation(s)
- Ebtsam S Abdel-Lah
- Department of Pharmacology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
- Department of Pharmacology, School of Veterinary Medicine, Badr University, Assiut, 11829, Egypt.
| | - Hoda S Sherkawy
- Department of Medical Biochemistry, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Wafaa H Mohamed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Aswan University, Aswan, Egypt
| | - Mariam A Fawy
- Department of Zoology, Faculty of Science, South Valley University, Qena, 83523, Egypt
| | - Asmaa A Hasan
- Department of Human Anatomy and Embryology, Faculty of Medicine, Aswan University, Aswan, 81528, Egypt
| | - Asmaa A Muhammed
- Department of Medical Physiology, Faculty of Medicine, Aswan University, Aswan, 81528, Egypt
| | - Amira F Taha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Abeer A Tony
- Department of Neuropsychiatry, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Nashwa Hamad
- Department of Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71515, Egypt
| | - Marwa G Gamea
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, 71526, Egypt
- Basic Medical Science Department, Badr University, Assiut, Egypt
| |
Collapse
|
3
|
Alami M, Zerif E, Khalil A, Hajji N, Ramassamy C, Lacombe G, Laurent B, Cohen AA, Wikowski JM, Gris D, Bunt T, van Tellingen O, Hirokawa K, Fulop T, Berrougui H. Neuroprotective effects of SGLT2 inhibitors empagliflozin and dapagliflozin on Aβ 1-42-induced neurotoxicity and neuroinflammation in cellular models of Alzheimer's disease. J Alzheimers Dis 2025; 105:464-480. [PMID: 40111935 DOI: 10.1177/13872877251329474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
BackgroundAlzheimer's disease (AD) is a chronic brain degenerative disease that leads to dementia.ObjectiveThe aim of the present study is to investigate the neuroprotective impact of sodium-glucose cotransporter-2 inhibitors (SGLT2i) (empagliflozin and dapagliflozin) on tau phosphorylation, oxidative stress, and neuroinflammation.MethodsWe used MTT (3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide) assay, annexin-V-FITC kit, and DCFH-DA (dichloro-dihydro-fluorescein diacetate) to respectively evaluate the effect of the SGLT2i (empagliflozin and dapagliflozin) on amyloid-β (Aβ)1-42-induced neuronal death, apoptosis, and oxidative stress. The expression of NLRP3-inflammasome, phospho-Tau181, glycogen synthase kinase-3 beta (GSK-3β), cyclin-dependent kinase 5 (CdK5), and histone deacetylase 6 (HDAC6), was quantified by flow cytometry. Drug distribution in the mice's brains was assessed by liquid chromatography-mass spectrometry (LC-MS).ResultsAβ1-42 significantly reduced cell viability and increased apoptosis, which was reversed by using gliflozins. SGLT2i significantly reduced Aβ1-42-induced reactive oxygen species generation, downregulated NLRP3-inflammasome, and diminished tau pathology. Mechanistically, the last effect involved the modulation of GSK-3β and CdK5 protein expression. However, the tested treatments did not modify the Aβ1-42-stimulating effect of HDAC6. Gliflozins are substrates of drug transporters ATP-binding cassette sub-family B member 1 and/or ATP binding cassette subfamily G member 2 (ABCB1 and ABCG2), and Elacridar significantly enhances their brain distribution.ConclusionsSGLT2i empagliflozin and dapagliflozin exhibited neuroprotective actions against human Aβ1-42-induced neurotoxicity.
Collapse
Affiliation(s)
- Mehdi Alami
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal, Morocco
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Echarki Zerif
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Abdelouahed Khalil
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Nabil Hajji
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, Seville, Spain
| | - Charles Ramassamy
- INRS-Centre Armand-Frappier Santé-biotechnologie, Laval, Montréal, Québec, Canada
| | - Guy Lacombe
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Benoit Laurent
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Research Center on Aging, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - Jacek M Wikowski
- Department of Embryology, Medical University of Gdańsk, Gdańsk, Poland
| | - Denis Gris
- Immunology Division, Faculty of Medicine and Health Sciences, Centre de Recherche du CHUS, University of Sherbrooke, Sherbrooke, Québec, Canada
| | - Ton Bunt
- Izumi Biosciences, Inc., Lexington, MA, USA
| | - Olaf van Tellingen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Katsuiku Hirokawa
- Department of Pathology, Institute Health and Life Science, Tokyo Medical and Dental University, Tokyo and Nito-memory Nakanosogo Hospital, Itabashi-ku, Tokyo, Japan
| | - Tamas Fulop
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Hicham Berrougui
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal, Morocco
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| |
Collapse
|
4
|
Hsiao YY, Chen YY, Kuo MJ, Chien YS, Li GY, Wu SJ, Lin WL, Chiu SF, Li CH, Lin JC, Lin CH, Huang JL, Hsieh YC, Chen SA. SGLT2i and Cardiovascular Events in Patients With Concomitant Atrial Fibrillation and Diabetes: A TriNetX Cohort Study. J Clin Endocrinol Metab 2025:dgae861. [PMID: 39998428 DOI: 10.1210/clinem/dgae861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Indexed: 02/26/2025]
Abstract
AIMS Sodium-glucose co-transporter 2 inhibitors (SGLT2i) enhance cardiovascular outcomes in individuals with type 2 diabetes mellitus (T2DM). Whether such effects also occur in T2DM patients with atrial fibrillation (AF) remains unknown. We aimed to investigate SGLT2i use on cardiovascular outcomes in patients with concomitant AF and T2DM. METHODS Patients with both AF and T2DM were identified from TriNetX, an international electronic medical record. Participants were divided into 2 groups according to their use of SGLT2i, at a 1:1 distribution through propensity score matching (PSM). The hazard ratio (HR) for clinical outcomes was determined using multivariate Cox hazards regression model. RESULTS We studied 339 792 patients with AF and T2DM, with 32 945 (9.70%) SGLT2i users. Following PSM, 17 011 patients aged 68.4 ± 7.9 years were included in each group. After a 3-year follow-up, patients treated with SGLT2i showed significantly reduced risks of stroke (adjusted HR: 0.830, P < .001), dementia (adjusted HR: 0.662, P < .001), long-standing persistent AF (adjusted HR: 0.917, P < .001), heart failure (adjusted HR: 0.833, P < .001), and all-cause mortality (adjusted HR: 0.532, P < .001). CONCLUSION The use of SGLT2i was associated with reduced risks of stroke, dementia, long-standing persistent AF, heart failure, and mortality in patients with both AF and T2DM. SGLT2i may be considered as a potential first-line therapy for this population.
Collapse
Affiliation(s)
- Yu-Yu Hsiao
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
| | - Yun-Yu Chen
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Internal Medicine, Faculty of Medicine, Institute of Clinical Medicine, National Yang-Ming Chiao Tung University School of Medicine, Taipei 112304, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University School of Medicine, Taichung 402202, Taiwan
| | - Ming-Jen Kuo
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Internal Medicine, Faculty of Medicine, Institute of Clinical Medicine, National Yang-Ming Chiao Tung University School of Medicine, Taipei 112304, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University School of Medicine, Taichung 402202, Taiwan
- Department of Data Science and Big Data Analytics, and Department of Financial Engineering, Providence University, Taichung 43301, Taiwan
| | - Yu-Shan Chien
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Internal Medicine, Faculty of Medicine, Institute of Clinical Medicine, National Yang-Ming Chiao Tung University School of Medicine, Taipei 112304, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University School of Medicine, Taichung 402202, Taiwan
- Department of Data Science and Big Data Analytics, and Department of Financial Engineering, Providence University, Taichung 43301, Taiwan
| | - Guan-Yi Li
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Internal Medicine, Faculty of Medicine, Institute of Clinical Medicine, National Yang-Ming Chiao Tung University School of Medicine, Taipei 112304, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University School of Medicine, Taichung 402202, Taiwan
| | - Shang-Ju Wu
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Internal Medicine, Faculty of Medicine, Institute of Clinical Medicine, National Yang-Ming Chiao Tung University School of Medicine, Taipei 112304, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University School of Medicine, Taichung 402202, Taiwan
- Department of Data Science and Big Data Analytics, and Department of Financial Engineering, Providence University, Taichung 43301, Taiwan
| | - Wei-Lun Lin
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Internal Medicine, Faculty of Medicine, Institute of Clinical Medicine, National Yang-Ming Chiao Tung University School of Medicine, Taipei 112304, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University School of Medicine, Taichung 402202, Taiwan
| | - Shu-Fen Chiu
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
| | - Cheng-Hung Li
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Internal Medicine, Faculty of Medicine, Institute of Clinical Medicine, National Yang-Ming Chiao Tung University School of Medicine, Taipei 112304, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University School of Medicine, Taichung 402202, Taiwan
- Department of Data Science and Big Data Analytics, and Department of Financial Engineering, Providence University, Taichung 43301, Taiwan
| | - Jiunn-Cherng Lin
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Internal Medicine, Faculty of Medicine, Institute of Clinical Medicine, National Yang-Ming Chiao Tung University School of Medicine, Taipei 112304, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University School of Medicine, Taichung 402202, Taiwan
- Department of Data Science and Big Data Analytics, and Department of Financial Engineering, Providence University, Taichung 43301, Taiwan
| | - Ching-Heng Lin
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407219, Taiwan
| | - Jin-Long Huang
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Internal Medicine, Faculty of Medicine, Institute of Clinical Medicine, National Yang-Ming Chiao Tung University School of Medicine, Taipei 112304, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University School of Medicine, Taichung 402202, Taiwan
- Department of Medical Education, Taichung Veterans General Hospital, Taichung 407219, Taiwan
| | - Yu-Cheng Hsieh
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Internal Medicine, Faculty of Medicine, Institute of Clinical Medicine, National Yang-Ming Chiao Tung University School of Medicine, Taipei 112304, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University School of Medicine, Taichung 402202, Taiwan
- Department of Data Science and Big Data Analytics, and Department of Financial Engineering, Providence University, Taichung 43301, Taiwan
| | - Shih-Ann Chen
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Internal Medicine, Faculty of Medicine, Institute of Clinical Medicine, National Yang-Ming Chiao Tung University School of Medicine, Taipei 112304, Taiwan
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University School of Medicine, Taichung 402202, Taiwan
| |
Collapse
|
5
|
Chen X, Fan M, Xiao Z, Xiong X. Dapagliflozin Improves High-Fat Diet-Induced Cognitive Impairment in Female Mice. Brain Behav 2025; 15:e70361. [PMID: 39972998 PMCID: PMC11839776 DOI: 10.1002/brb3.70361] [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: 08/02/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND High fat consumption is a known risk factor for the development of type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD). Sodium-glucose cotransporter 2 inhibitors (SGLT2is) have been found to possess anti-inflammatory and neuroprotective properties. However, the cognitive effects and mechanisms of SGLT2is on female mice fed with a high-fat diet remain unknown. OBJECTIVE This study aimed to investigate the impacts of dapagliflozin on metabolism, cognition, neuroinflammation, insulin resistance, and microglial activation in female mice fed a HFD. METHODS Dapagliflozin (1 mg/kg) was administered to HFD-fed mice for 24 weeks. Body weight, glucose tolerance, and insulin resistance were assessed. Additionally, all mice were subjected to the Morris water maze (MWM) and one-trial Y-maze tests. The levels of metabolic hormones and cytokines were analyzed using ELISA kits. The levels of phosphorylated tau (p-tau) protein in the hippocampus were measured. Microglia, insulin receptors, NLRP3, and IL-1β in the hippocampus of mice were evaluated by immunofluorescence or immunohistochemical staining. RESULTS As anticipated, dapagliflozin improved insulin resistance and glucose metabolism and reduced cognitive impairment in female mice fed with a HFD. In the hippocampus, dapagliflozin alleviated microglial activation yet did not reduce the secretion of inflammatory chemokines. Furthermore, it increased the expression of insulin receptor in the hippocampus of HFD-fed mice and decreased the expression of p-tau. CONCLUSIONS Our results provide a foundation for the clinical application of SGLT2is as an adjuvant to slow down the progression of central degenerative diseases related to metabolic disorders, such as AD.
Collapse
Affiliation(s)
- Xiaolin Chen
- Department of EndocrinologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Mingxia Fan
- Center for Animal ExperimentRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhuoni Xiao
- Reproductive Medical CenterRenmin Hospital of Wuhan UniversityWuhanChina
| | - Xiaoxing Xiong
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
6
|
Hou Y, Chen Z, Cheng J, Li G, Yin L, Gao J. The Mechanism and Treatment of Cognitive Dysfunction in Diabetes: A Review. Exp Clin Endocrinol Diabetes 2025; 133:64-72. [PMID: 39572247 DOI: 10.1055/a-2480-7826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Diabetes mellitus (DM) is one of the fastest growing diseases in terms of global incidence and seriously affects cognitive function. The incidence rate of cognitive dysfunction is up to 13% in diabetes patients aged 65-74 years and reaches 24% in those aged >75 years. The mechanisms and treatments of cognitive dysfunction associated with diabetes mellitus are complicated and varied. Previous studies suggest that hyperglycemia mainly contributes to cognitive dysfunction through mechanisms involving inflammation, autophagy, the microbial-gut-brain axis, brain-derived neurotrophic factors, and insulin resistance. Antidiabetic drugs such as metformin, liraglutide, and empagliflozin and other drugs such as fingolimod and melatonin can alleviate diabetes-induced cognitive dysfunction. Self-management, intermittent fasting, and repetitive transverse magnetic stimulation can also ameliorate cognitive impairment. In this review, we discuss the mechanisms linking diabetes mellitus with cognitive dysfunction and propose a potential treatment for cognitive decline associated with diabetes mellitus.
Collapse
Affiliation(s)
- Yangbo Hou
- Department of Neurology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhen Chen
- Department of Encephalopathy, Suqian Hospital of Chinese Medicine , Nanjing University of Traditional Chinese Medicine, Suqian, China
| | - Jiwei Cheng
- Department of Neurology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guoyi Li
- Department of Neurology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Yin
- Department of Rehabilitation, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Gao
- Department of Endocrinology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
7
|
Zhang Y, Liao X, Xu J, Yin J, Li S, Li M, Shi X, Zhang S, Li C, Xu W, Yu X, Yang Y. The Promising Potency of Sodium-Glucose Cotransporter 2 Inhibitors in the Prevention of and as Treatment for Cognitive Impairment Among Type 2 Diabetes Patients. Biomedicines 2024; 12:2783. [PMID: 39767690 PMCID: PMC11673520 DOI: 10.3390/biomedicines12122783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM), accounting for the majority of diabetes mellitus prevalence, is associated with an increased risk of cognition decline and deterioration of cognition function in diabetic patients. The sodium-glucose cotransporter 2 (SGLT2), located in the renal proximal tubule, plays a role in urine glucose reabsorption. SGLT2 inhibitors (SGLT2i), have shown potential benefits beyond cardiac and renal improvement in preventing and treating cognitive impairment (CI), including mild cognitive impairment, Alzheimer's disease and vascular dementia in T2DM patients. Studies suggest that SGLT2i may ameliorate diabetic CI through metabolism pathways, inflammation, oxidative stress, neurotrophic factors and AChE inhibition. Clinical trials and meta-analyses have reported significant and insignificant results. Given their vascular effects, SGLT2i may offer unique protection against vascular CI. This review compiles mechanisms and clinical evidence, emphasizing the need for future analysis, evaluation, trials and meta-analyses to verify and recommend optimal SGLT2i selection and dosage for specific patients.
Collapse
Affiliation(s)
- Yibin Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaobin Liao
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jialu Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Jiaxin Yin
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Shan Li
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Mengni Li
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Xiaoli Shi
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Shujun Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Chunyu Li
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Weijie Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Xuefeng Yu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Yan Yang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| |
Collapse
|
8
|
Wilar G, Suhandi C, Wathoni N, Fukunaga K, Kawahata I. Nanoparticle-Based Drug Delivery Systems Enhance Treatment of Cognitive Defects. Int J Nanomedicine 2024; 19:11357-11378. [PMID: 39524925 PMCID: PMC11550695 DOI: 10.2147/ijn.s484838] [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: 06/29/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Nanoparticle-based drug delivery presents a promising solution in enhancing therapies for neurological diseases, particularly cognitive impairment. These nanoparticles address challenges related to the physicochemical profiles of drugs that hinder their delivery to the central nervous system (CNS). Benefits include improved solubility due to particle size reduction, enhanced drug penetration across the blood-brain barrier (BBB), and sustained release mechanisms suitable for long-term therapy. Successful application of nanoparticle delivery systems requires careful consideration of their characteristics tailored for CNS delivery, encompassing particle size and distribution, surface charge and morphology, loading capacity, and drug release kinetics. Literature review reveals three main types of nanoparticles developed for cognitive function enhancement: polymeric nanoparticles, lipid-based nanoparticles, and metallic or inorganic nanoparticles. Each type and its production methods possess distinct advantages and limitations. Further modifications such as coating agents or ligand conjugation have been explored to enhance their brain cell uptake. Evidence supporting their development shows improved efficacy outcomes, evidenced by enhanced cognitive function assessments, modulation of pro-oxidant markers, and anti-inflammatory activities. Despite these advancements, clinical trials validating the efficacy of nanoparticle systems in treating cognitive defects are lacking. Therefore, these findings underscore the need for researchers to expedite clinical testing to provide robust evidence of the potential of nanoparticle-based drug delivery systems.
Collapse
Affiliation(s)
- Gofarana Wilar
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | - Cecep Suhandi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Ichiro Kawahata
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
| |
Collapse
|
9
|
Manral K, Singh A, Singh Y. Nanotechnology as a potential treatment for diabetes and its complications: A review. Diabetes Metab Syndr 2024; 18:103159. [PMID: 39612615 DOI: 10.1016/j.dsx.2024.103159] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND AND AIM Diabetes mellitus is a chronic metabolic disorder that causes multiple complications in various organs, such as the kidney, liver and cardiovascular system. These complications are the main causes of morbidity and mortality in patients with diabetes. Nanotechnology offers new opportunities for the therapy of diabetes and its multiple complications through site-specific and precise drug delivery. This review summarizes the various studies demonstrating the potential applications of different nanoparticles in diabetes-associated complications. METHOD A literature search was conducted using PubMed, Google Scholar and Scopus databases, focusing on the role of nanoparticles in the improved delivery of various hypoglycemic agents for the treatment of microvascular and macrovascular diabetic complications. RESULTS Numerous studies have shown that nanoparticles, such as nanoliposomes, polymeric micelles, dendrimers and metallic nanoparticles, improve the delivery of various hypoglycemic agents. Moreover, nanoparticles have been found to be safer, with improved pharmacokinetic and pharmacodynamic profiles. CONCLUSION This review outlines the significant role of nanotechnology in diabetes and related complications and its superiority over conventional drug delivery.
Collapse
Affiliation(s)
- Kanika Manral
- Department of Pharmaceutical Sciences, Faculty of Technology Sir J.C Bose Technical Campus Bhimtal, Kumaun University Nainital, 263136, India.
| | - Anita Singh
- Department of Pharmaceutical Sciences, Faculty of Technology Sir J.C Bose Technical Campus Bhimtal, Kumaun University Nainital, 263136, India.
| | - Yuvraj Singh
- Department of Pharmacy, BITS-Pilani, Hyderabad Campus, Medchal, Hyderabad, 500078, India.
| |
Collapse
|
10
|
Hong B, Lee H, Choi A, Kim WJ, Cho YM, Yon DK, Shin JY. Sodium-glucose cotransporter-2 inhibitors versus dipeptidyl peptidase IV inhibitors and risk of dementia among patients with type 2 diabetes and comorbid mental disorders: A population-based cohort study. DIABETES & METABOLISM 2024; 50:101581. [PMID: 39349097 DOI: 10.1016/j.diabet.2024.101581] [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: 05/08/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 10/02/2024]
Abstract
AIM To evaluate whether the use of sodium-glucose cotransporter-2 (SGLT2) inhibitors which have shown potential neuroprotective effects, is associated with lower risk of dementia in patients with type 2 diabetes (T2D) and comorbid mental disorders, who are considerably more susceptible to dementia. METHODS Using the nationwide healthcare data of South Korea between 2010 and 2022, we conducted a retrospective cohort study among patients with T2D and comorbid mental disorders initiating SGLT2 inhibitors versus active comparator (Dipeptidyl Peptidase IV (DPP4) inhibitors). Hazard ratios (HRs) and rate differences (RDs) per 1000 person-years of incident dementia were estimated after weighting by propensity score fine stratification method. RESULTS Over a 4.8-year median follow-up, SGLT2 inhibitors were associated with a 12 % lower risk of dementia compared with DPP4 inhibitors (11.31 vs. 12.86 events per 1000 person years; HR 0.88, 95 % CI 0.84 to 0.92; RD -1.55, -2.13 to -0.97). The results were consistent when stratified by age, sex, individual component, severe mental disorders, presence of insulin, history of cardiovascular disease, or history of hypertension. CONCLUSIONS SGLT2 inhibitors versus DPP4 inhibitors were associated with a lower risk of incident dementia in patients with T2D and comorbid mental disorders. Further randomized controlled trials are required to confirm our findings.
Collapse
Affiliation(s)
- Bin Hong
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Hyesung Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea; Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
| | - Ahhyung Choi
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Woo Jung Kim
- Department of Psychiatry, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Republic of Korea; Institute of Behavioral Sciences in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute for Innovation in Digital Healthcare, Yonsei University, Seoul, Republic of Korea
| | - Young Min Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Dong Keon Yon
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Ju-Young Shin
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea; Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea.
| |
Collapse
|
11
|
Hong B, Bea S, Ko HY, Kim WJ, Cho YM, Shin JY. Sodium-Glucose Cotransporter-2 Inhibitors, Dulaglutide, and Risk for Dementia : A Population-Based Cohort Study. Ann Intern Med 2024; 177:1319-1329. [PMID: 39186787 DOI: 10.7326/m23-3220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND Both sodium-glucose cotransporter-2 (SGLT2) inhibitors and glucagon-like peptide-1 receptor agonists (GLP-1 RAs) may have neuroprotective effects in patients with type 2 diabetes (T2D). However, their comparative effectiveness in preventing dementia remains uncertain. OBJECTIVE To compare the risk for dementia between SGLT2 inhibitors and dulaglutide (a GLP-1 RA). DESIGN Target trial emulation study. SETTING Nationwide health care data of South Korea obtained from the National Health Insurance Service between 2010 and 2022. PATIENTS Patients aged 60 years or older who have T2D and are initiating treatment with SGLT2 inhibitors or dulaglutide. MEASUREMENTS The primary outcome was the presumed clinical onset of dementia. The date of onset was defined as the year before the date of dementia diagnosis, assuming that the time between the onset of dementia and diagnosis was 1 year. The 5-year risk ratios and risk differences comparing SGLT2 inhibitors with dulaglutide were estimated in a 1:2 propensity score-matched cohort adjusted for confounders. RESULTS Overall, 12 489 patients initiating SGLT2 inhibitor treatment (51.9% dapagliflozin and 48.1% empagliflozin) and 1075 patients initiating dulaglutide treatment were included. In the matched cohort, over a median follow-up of 4.4 years, the primary outcome event occurred in 69 participants in the SGLT2 inhibitor group and 43 in the dulaglutide group. The estimated risk difference was -0.91 percentage point (95% CI, -2.45 to 0.63 percentage point), and the estimated risk ratio was 0.81 (CI, 0.56 to 1.16). LIMITATION Residual confounding is possible; there was no adjustment for hemoglobin A1c levels or duration of diabetes; the study is not representative of newer drugs, including more effective GLP-1 RAs; and the onset of dementia was not measured directly. CONCLUSION Under conventional statistical criteria, a risk for dementia between 2.5 percentage points lower and 0.6 percentage point greater for SGLT2 inhibitors than for dulaglutide was estimated to be highly compatible with the data from this study. However, whether these findings generalize to newer GLP-1 RAs is uncertain. Thus, further studies incorporating newer drugs within these drug classes and better addressing residual confounding are required. PRIMARY FUNDING SOURCE Ministry of Food and Drug Safety of South Korea.
Collapse
Affiliation(s)
- Bin Hong
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea (B.H., H.Y.K.)
| | - Sungho Bea
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea, and Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts (S.B.)
| | - Hwa Yeon Ko
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea (B.H., H.Y.K.)
| | - Woo Jung Kim
- Department of Psychiatry, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin; Institute of Behavioral Sciences in Medicine, Yonsei University College of Medicine, Seoul; and Institute for Innovation in Digital Healthcare, Yonsei University, Seoul, South Korea (W.J.K.)
| | - Young Min Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea (Y.M.C.)
| | - Ju-Young Shin
- School of Pharmacy and Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, and Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea (J.-Y.S.)
| |
Collapse
|
12
|
Gholami M, Coleman-Fuller N, Salehirad M, Darbeheshti S, Motaghinejad M. Neuroprotective Effects of Sodium-Glucose Cotransporter-2 (SGLT2) Inhibitors (Gliflozins) on Diabetes-Induced Neurodegeneration and Neurotoxicity: A Graphical Review. Int J Prev Med 2024; 15:28. [PMID: 39239308 PMCID: PMC11376549 DOI: 10.4103/ijpvm.ijpvm_5_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/20/2024] [Indexed: 09/07/2024] Open
Abstract
Diabetes is a chronic endocrine disorder that negatively affects various body systems, including the nervous system. Diabetes can cause or exacerbate various neurological disorders, and diabetes-induced neurodegeneration can involve several mechanisms such as mitochondrial dysfunction, activation of oxidative stress, neuronal inflammation, and cell death. In recent years, the management of diabetes-induced neurodegeneration has relied on several types of drugs, including sodium-glucose cotransporter-2 (SGLT2) inhibitors, also called gliflozins. In addition to exerting powerful effects in reducing blood glucose, gliflozins have strong anti-neuro-inflammatory characteristics that function by inhibiting oxidative stress and cell death in the nervous system in diabetic subjects. This review presents the molecular pathways involved in diabetes-induced neurodegeneration and evaluates the clinical and laboratory studies investigating the neuroprotective effects of gliflozins against diabetes-induced neurodegeneration, with discussion about the contributing roles of diverse molecular pathways, such as mitochondrial dysfunction, oxidative stress, neuro-inflammation, and cell death. Several databases-including Web of Science, Scopus, PubMed, Google Scholar, and various publishers, such as Springer, Wiley, and Elsevier-were searched for keywords regarding the neuroprotective effects of gliflozins against diabetes-triggered neurodegenerative events. Additionally, anti-neuro-inflammatory, anti-oxidative stress, and anti-cell death keywords were applied to evaluate potential neuronal protection mechanisms of gliflozins in diabetes subjects. The search period considered valid peer-reviewed studies published from January 2000 to July 2023. The current body of literature suggests that gliflozins can exert neuroprotective effects against diabetes-induced neurodegenerative events and neuronal dysfunction, and these effects are mediated via activation of mitochondrial function and prevention of cell death processes, oxidative stress, and inflammation in neurons affected by diabetes. Gliflozins can confer neuroprotective properties in diabetes-triggered neurodegeneration, and these effects are mediated by inhibiting oxidative stress, inflammation, and cell death.
Collapse
Affiliation(s)
- Mina Gholami
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Natalie Coleman-Fuller
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, USA
| | - Mahsa Salehirad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepideh Darbeheshti
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Motaghinejad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Kumar Nelson V, Jha NK, Nuli MV, Gupta S, Kanna S, Gahtani RM, Hani U, Singh AK, Abomughaid MM, Abomughayedh AM, Almutary AG, Iqbal D, Al Othaim A, Begum SS, Ahmad F, Mishra PC, Jha SK, Ojha S. Unveiling the impact of aging on BBB and Alzheimer's disease: Factors and therapeutic implications. Ageing Res Rev 2024; 98:102224. [PMID: 38346505 DOI: 10.1016/j.arr.2024.102224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/01/2024] [Accepted: 02/03/2024] [Indexed: 05/12/2024]
Abstract
Alzheimer's disease (AD) is a highly prevalent neurodegenerative condition that has devastating effects on individuals, often resulting in dementia. AD is primarily defined by the presence of extracellular plaques containing insoluble β-amyloid peptide (Aβ) and neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau protein (P-tau). In addition, individuals afflicted by these age-related illnesses experience a diminished state of health, which places significant financial strain on their loved ones. Several risk factors play a significant role in the development of AD. These factors include genetics, diet, smoking, certain diseases (such as cerebrovascular diseases, obesity, hypertension, and dyslipidemia), age, and alcohol consumption. Age-related factors are key contributors to the development of vascular-based neurodegenerative diseases such as AD. In general, the process of aging can lead to changes in the immune system's responses and can also initiate inflammation in the brain. The chronic inflammation and the inflammatory mediators found in the brain play a crucial role in the dysfunction of the blood-brain barrier (BBB). Furthermore, maintaining BBB integrity is of utmost importance in preventing a wide range of neurological disorders. Therefore, in this review, we discussed the role of age and its related factors in the breakdown of the blood-brain barrier and the development of AD. We also discussed the importance of different compounds, such as those with anti-aging properties, and other compounds that can help maintain the integrity of the blood-brain barrier in the prevention of AD. This review builds a strong correlation between age-related factors, degradation of the BBB, and its impact on AD.
Collapse
Affiliation(s)
- Vinod Kumar Nelson
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India.
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Centre of Research Impact and Outcome, Chitkara University, Rajpura 140401, Punjab, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | - Mohana Vamsi Nuli
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Sandeep Kanna
- Department of pharmaceutics, Chalapathi Institute of Pharmaceutical Sciences, Chalapathi Nagar, Guntur 522034, India
| | - Reem M Gahtani
- Departement of Clinical Laboratory Sciences, King Khalid University, Abha, Saudi Arabia
| | - Umme Hani
- Department of pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Arun Kumar Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology BHU, Varanasi, Uttar Pradesh, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ali M Abomughayedh
- Pharmacy Department, Aseer Central Hospital, Ministry of Health, Saudi Arabia
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, P.O. Box 59911, United Arab Emirates
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Ayoub Al Othaim
- Department of Medical Laboratory Sciences, College of Applied Medical Science, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - S Sabarunisha Begum
- Department of Biotechnology, P.S.R. Engineering College, Sivakasi 626140, India
| | - Fuzail Ahmad
- Respiratory Care Department, College of Applied Sciences, Almaarefa University, Diriya, Riyadh, 13713, Saudi Arabia
| | - Prabhu Chandra Mishra
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, India.
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, P.O. Box 15551, United Arab Emirates
| |
Collapse
|
14
|
Hierro-Bujalance C, Garcia-Alloza M. Empagliflozin reduces brain pathology in Alzheimer's disease and type 2 diabetes. Neural Regen Res 2024; 19:1189-1190. [PMID: 37905858 PMCID: PMC11467955 DOI: 10.4103/1673-5374.385865] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/20/2023] [Accepted: 08/20/2023] [Indexed: 11/02/2023] Open
Affiliation(s)
- Carmen Hierro-Bujalance
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
- Salus Infirmorum-Universidad de Cadiz, Cadiz, Spain
| | - Monica Garcia-Alloza
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| |
Collapse
|
15
|
Zandifar A, Panahi M, Badrfam R, Qorbani M. Efficacy of empagliflozin as adjunctive therapy to citalopram in major depressive disorder: a randomized double-blind, placebo-controlled clinical trial. BMC Psychiatry 2024; 24:163. [PMID: 38408937 PMCID: PMC10895773 DOI: 10.1186/s12888-024-05627-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 02/20/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Major depressive disorder is one of the most common psychiatric disorders, which is associated with a high disease burden. Current treatments using antidepressants have limitations, so using medication with neuromodulating and anti-inflammatory properties alongside them could be helpful. In a clinical trial, we studied the effectiveness of empagliflozin, a blood sugar-lowering drug, as an adjunctive therapy to reduce the severity of depression symptoms. METHODS A number of outpatients with moderate to severe depression (Hamilton Depression Rating Scale (HDRS) > = 17) who were not under related medication or had not taken medication for at least the last two months, had an age range of 18-60 years and had written informed consent to enter the study (N = 90) were randomly divided into two groups receiving placebo or empagliflozin (10 mg daily) combined with citalopram (40 mg daily) based on permuted block randomization method in an 8-week randomized, double-blind, placebo-controlled clinical trial. They were evaluated using the HDRS in weeks 0, 4, and 8. RESULTS HDRS scores were equal to 28.42(± 3.83), 20.20(± 3.82), and 13.42(± 3.42) in the placebo group during weeks 0,4, and 8, respectively. These scores were 27.36(± 3.77), 13.76(± 1.40), and 7.00(± 1.13), respectively, for the group treated with empagliflozin. Compared to the control group, patients treated with empagliflozin using repeated-measures ANOVA showed greater improvement in reducing the severity of depression symptoms over time (p value = 0.0001). CONCLUSIONS Considering the promising findings in this clinical trial, further study of empagliflozin as adjunctive therapy in MDD with larger sample sizes and longer follow-ups is recommended.
Collapse
Affiliation(s)
- Atefeh Zandifar
- Social Determinants of Health Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Department of Psychiatry, Imam Hossein Hospital, Alborz University of Medical Sciences, Karaj, Alborz, Iran
| | - Maryam Panahi
- Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Rahim Badrfam
- Department of Psychiatry, Imam Hossein Hospital, Alborz University of Medical Sciences, Karaj, Alborz, Iran.
| | - Mostafa Qorbani
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Chronic Diseases Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Elian V, Popovici V, Karampelas O, Pircalabioru GG, Radulian G, Musat M. Risks and Benefits of SGLT-2 Inhibitors for Type 1 Diabetes Patients Using Automated Insulin Delivery Systems-A Literature Review. Int J Mol Sci 2024; 25:1972. [PMID: 38396657 PMCID: PMC10888162 DOI: 10.3390/ijms25041972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/27/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024] Open
Abstract
The primary treatment for autoimmune Diabetes Mellitus (Type 1 Diabetes Mellitus-T1DM) is insulin therapy. Unfortunately, a multitude of clinical cases has demonstrated that the use of insulin as a sole therapeutic intervention fails to address all issues comprehensively. Therefore, non-insulin adjunct treatment has been investigated and shown successful results in clinical trials. Various hypoglycemia-inducing drugs such as Metformin, glucagon-like peptide 1 (GLP-1) receptor agonists, dipeptidyl peptidase-4 (DPP-4) inhibitors, amylin analogs, and Sodium-Glucose Cotransporters 2 (SGLT-2) inhibitors, developed good outcomes in patients with T1DM. Currently, SGLT-2 inhibitors have remarkably improved the treatment of patients with diabetes by preventing cardiovascular events, heart failure hospitalization, and progression of renal disease. However, their pharmacological potential has not been explored enough. Thus, the substantial interest in SGLT-2 inhibitors (SGLT-2is) underlines the present review. It begins with an overview of carrier-mediated cellular glucose uptake, evidencing the insulin-independent transport system contribution to glucose homeostasis and the essential roles of Sodium-Glucose Cotransporters 1 and 2. Then, the pharmacological properties of SGLT-2is are detailed, leading to potential applications in treating T1DM patients with automated insulin delivery (AID) systems. Results from several studies demonstrated improvements in glycemic control, an increase in Time in Range (TIR), a decrease in glycemic variability, reduced daily insulin requirements without increasing hyperglycemic events, and benefits in weight management. However, these advantages are counterbalanced by increased risks, particularly concerning Diabetic Ketoacidosis (DKA). Several clinical trials reported a higher incidence of DKA when patients with T1DM received SGLT-2 inhibitors such as Sotagliflozin and Empagliflozin. On the other hand, patients with T1DM and a body mass index (BMI) of ≥27 kg/m2 treated with Dapagliflozin showed similar reduction in hyperglycemia and body weight and insignificantly increased DKA incidence compared to the overall trial population. Additional multicenter and randomized studies are required to establish safer and more effective long-term strategies based on patient selection, education, and continuous ketone body monitoring for optimal integration of SGLT-2 inhibitors into T1DM therapeutic protocol.
Collapse
Affiliation(s)
- Viviana Elian
- Department of Diabetes, Nutrition and Metabolic Diseases, “Carol Davila” University of Medicine and Pharmacy, 5-7 Ion Movila Street, 020475 Bucharest, Romania; (V.E.); (G.R.)
- Department of Diabetes, Nutrition and Metabolic Diseases, “N. C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 020475 Bucharest, Romania
| | - Violeta Popovici
- “Costin C. Kiriţescu” National Institute of Economic Research—Center for Mountain Economics (INCE-CEMONT) of Romanian Academy, 725700 Vatra-Dornei, Romania
| | - Oana Karampelas
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020945 Bucharest, Romania;
| | - Gratiela Gradisteanu Pircalabioru
- eBio-Hub Research Centre, National University of Science and Technology Politehnica Bucharest, 061344 Bucharest, Romania;
- Research Institute, University of Bucharest, 061344 Bucharest, Romania
- Academy of Romanian Scientists, 54 Splaiul Independentei, 050094 Bucharest, Romania
| | - Gabriela Radulian
- Department of Diabetes, Nutrition and Metabolic Diseases, “Carol Davila” University of Medicine and Pharmacy, 5-7 Ion Movila Street, 020475 Bucharest, Romania; (V.E.); (G.R.)
- Department of Diabetes, Nutrition and Metabolic Diseases, “N. C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 020475 Bucharest, Romania
| | - Madalina Musat
- eBio-Hub Research Centre, National University of Science and Technology Politehnica Bucharest, 061344 Bucharest, Romania;
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 030167 Bucharest, Romania
- Department of Endocrinology IV, “C. I. Parhon” National Institute of Endocrinology, 011863 Bucharest, Romania
| |
Collapse
|
17
|
Yaribeygi H, Maleki M, Sathyapalan T, Rizzo M, Sahebkar A. Cognitive Benefits of Sodium-Glucose Co-Transporters-2 Inhibitors in the Diabetic Milieu. Curr Med Chem 2024; 31:138-151. [PMID: 36733247 DOI: 10.2174/0929867330666230202163513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/19/2022] [Accepted: 11/30/2022] [Indexed: 02/04/2023]
Abstract
Patients with diabetes are at higher risk of cognitive impairment and memory loss than the normal population. Thus, using hypoglycemic agents to improve brain function is important for diabetic patients. Sodium-glucose cotransporters-2 inhibitors (SGLT2i) are a class of therapeutic agents used in the management of diabetes that has some pharmacologic effects enabling them to fight against the onset and progress of memory deficits. Although the exact mediating pathways are not well understood, emerging evidence suggests that SGLT2 inhibition is associated with improved brain function. This study reviewed the possible mechanisms and provided evidence suggesting SGLT2 inhibitors could ameliorate cognitive deficits.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Mina Maleki
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, School of Medicine, University of Palermo, 90133, Palermo, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Pradhan SP, Sahu PK, Behera A. New insights toward molecular and nanotechnological approaches to antidiabetic agents for Alzheimer's disease. Mol Cell Biochem 2023; 478:2739-2762. [PMID: 36949264 DOI: 10.1007/s11010-023-04696-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 02/27/2023] [Indexed: 03/24/2023]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder affecting a major class of silver citizens. The disorder shares a mutual relationship on account of its cellular and molecular pathophysiology with type-II diabetes mellitus (DM). Chronic DM increases the risk for AD. Emerging evidence recommended that resistance in insulin production develops cognitive dysfunction, which generally leads to AD. Repurposing of antidiabetic drugs can be effective in preventing and treatment of the neurodegenerative disorder. Limitations of antidiabetic drugs restrict the repurposing of the drugs for other disorders. Therefore, nanotechnological intervention plays a significant role in the treatment of neurological disorders. In this review, we discuss the common cellular and molecular pathophysiologies between AD and type-II DM, the relevance of in vivo models of type II DM in the study of AD, and the repurposing of antidiabetic drugs and the nanodelivery systems of antidiabetic drugs against AD.
Collapse
Affiliation(s)
- Sweta Priyadarshini Pradhan
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Campus-II, Kalinga Nagar, Bhubaneswar, Odisha, India
| | - Pratap Kumar Sahu
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Campus-II, Kalinga Nagar, Bhubaneswar, Odisha, India
| | - Anindita Behera
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Campus-II, Kalinga Nagar, Bhubaneswar, Odisha, India.
| |
Collapse
|
19
|
Yaribeygi H, Maleki M, Jamialahmadi T, Shakhpazyan NK, Kesharwani P, Sahebkar A. Nanoparticles with SGLT2 inhibitory activity: Possible benefits and future. Diabetes Metab Syndr 2023; 17:102869. [PMID: 37778134 DOI: 10.1016/j.dsx.2023.102869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/12/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023]
Abstract
AIM Nano-drug delivery is a rapidly growing approach in medicine that helps design and develop newer forms of drugs with more efficacy and lower adverse effects. Sodium-glucose cotransporter-2 inhibitors are an emerging class of antidiabetic agents that reduce the blood glucose levels by damping glucose reabsorption in renal proximal tubules. METHODS AND RESULTS This mechanism might be followed by some adverse effects that could be prevented by nano-drug delivery. Although we have still limited evidence about nanoforms of sodium-glucose cotransporter-2 inhibitors, current knowledge strongly suggests that nanotechnology can help us design more effective drugs with lower side effects. In recent years, several studies have explored the possible benefits of nanoforms of sodium-glucose cotransporter-2 inhibitors. However, clinical trials are yet to be conducted. CONCLUSION In the current review, we present the latest findings on the development and benefits of nanoforms of sodium-glucose cotransporter-2 inhibitors.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Mina Maleki
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tannaz Jamialahmadi
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nikolay K Shakhpazyan
- Petrovsky National Science Center of Surgery, 2 Abrikosovsky Lane, 119991, Moscow, Russia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
20
|
Tian S, Jiang J, Wang J, Zhang Z, Miao Y, Ji X, Bi Y. Comparison on cognitive outcomes of antidiabetic agents for type 2 diabetes: A systematic review and network meta-analysis. Diabetes Metab Res Rev 2023; 39:e3673. [PMID: 37302139 DOI: 10.1002/dmrr.3673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/02/2023] [Accepted: 04/25/2023] [Indexed: 06/13/2023]
Abstract
We aimed to summarise current evidence on different antidiabetic drugs to delay cognitive impairment, including mild cognitive impairment, dementia, Alzheimer's disease (AD) and vascular dementia, among subjects with type 2 diabetes mellitus (T2DM). Medline, Cochrane and Embase databases were searched from inception to 31 July 2022. Two investigators independently reviewed and screened trials comparing antidiabetic drugs with no antidiabetic drugs, placebo, or other active antidiabetic drugs on cognitive outcomes in T2DM. Data were analysed using meta-analysis and network meta-analysis. Twenty-seven studies met the inclusion criteria, including 3 randomised controlled trials, 19 cohort studies and 5 case-control studies. Compared with non-user, SGLT-2i (OR 0.41 [95% CI 0.22-0.76]), GLP-1RA (OR 0.34 [95% CI 0.14-0.85]), thiazolidinedione (OR 0.60 [95% CI 0.51-0.69]), and DPP-4i (OR 0.78 [95% CI 0.61-0.99]) users had a decreased risk of dementia, whereas sulfonylurea (OR 1.43 [95% CI 1.11-1.82]) increased dementia risk. Network meta-analysis showed that SGLT-2i was most likely to rank best (SUCRA = 94.4%), GLP-1 RA second best (SUCRA = 92.7%), thiazolidinedione third best (SUCRA = 74.7%) and DPP-4i fourth best (SUCRA = 54.9%), while sulfonylurea second worst (SUCRA = 20.0%) for decreasing dementia outcomes, by synthesising evidence from direct and indirect comparisons of multiple intervention. Evidence suggests the effects of SGLT-2i ≈ GLP-1 RAs > thiazolidinedione > DPP-4i for delaying cognitive impairment, dementia and AD outcomes, whereas sulfonylurea was associated with the highest risk. These findings provide evidence for evaluating the optional treatment for clinical practice. PROSPERO REGISTRATION: Registration no. CRD42022347280.
Collapse
Affiliation(s)
- Sai Tian
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Jiaxuan Jiang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Jin Wang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Zhou Zhang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Yingwen Miao
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Xinlu Ji
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Yan Bi
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| |
Collapse
|
21
|
Farooq MA, Ali S, Sulayman R, Hassan A, Tahir HM, Shahzad H, Fiaz U, Mughal TA, Liaqat I, Mumtaz S, Nauroze T, Mumtaz S. Therapeutic Applications of Garlic and Turmeric for the Diabetic Wound Healing in Mice. J Burn Care Res 2023; 44:800-809. [PMID: 36331804 DOI: 10.1093/jbcr/irac169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Diabetes is involved in delayed wound healing that can be cured by natural products such as garlic, turmeric, and fibroin extracts. Alloxan monohydrate is used for inducing diabetes in mice. The percent wound contraction of garlic (150 mg/ml), turmeric (100 mg/ml), and fibroin (50 mg/ml), individually and in combinations garlic (150 mg/ml) + fibroin (50 mg/ml), turmeric (100 mg/ml) + fibroin (50 mg/ml), garlic (150 mg/ml) + turmeric (100 mg/ml), and garlic (150 mg/ml) + turmeric (100 mg/ml) + fibroin (50 mg/ml) was checked by evaluating the healing time, % wound contraction and histological analysis. The serum level of MMPs (MMP 2, MMP7, MMP 9), pro-inflammatory cytokines (TNF-α, IL-6, IL-8), and TIMPs were evaluated. With the combination of three extracts (Ga+Tu+Fi) garlic (150 mg/ml), turmeric (100 mg/ml) and fibroin (50 mg/ml), wounds healed in 12 days and had 97.3 ± 2.2% wound contraction. While the positive control (polyfax) and diabetic control (saline) wounds healed in 17- and 19-days with wound contraction of 96.7 ± 1.4% and 96.3 ± 1.1%, respectively. Histological analysis showed that the combination of Ga+Tu+Fi exhibited an increase in the growth of collagen fibers, fibroblasts number, and keratinocytes, and lessened inflammation of blood vessels. The combination of Ga+Tu+Fi significantly alleviated the serum concentration of TNF-α (14.2 ± 0.7 pg/ml), IL-6 (10.0 ± 1.0 pg/ml), IL-8 (16.0 ± 1.5 pg/ml), MMP2 (228.0 ± 18.1 pg/ml), MMP7 (271.0 ± 9.9 pg/ml), and MMP9 (141.0 ± 5.3 pg/ml) to diabetic control. The level of TIMPs (193.0 ± 9.1 pg/ml) was increased significantly with respect to diabetic control. We conclude that the combination of these biomaterials possessed high regenerative and healing capabilities and can be an effective remedy in the healing of chronic wounds in diabetic patients.
Collapse
Affiliation(s)
- Muhammad A Farooq
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shaukat Ali
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rida Sulayman
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Ali Hassan
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Hafiz M Tahir
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Hafsa Shahzad
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Umaima Fiaz
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Tafail A Mughal
- Department of Zoology, Women University of Azad Jammu Kashmir, Bagh, Pakistan
| | - Irfana Liaqat
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Samaira Mumtaz
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Tooba Nauroze
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shumaila Mumtaz
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| |
Collapse
|
22
|
Khan S, Hasan N, Khan S, Akhtar M, Akhtar M, Najmi AK. Exploring effects of Simvastatin on coagulation mediators to alleviate the advancement of high cholesterol diet triggered neurodegeneration. J Biochem Mol Toxicol 2023:e23342. [PMID: 36992618 DOI: 10.1002/jbt.23342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 01/17/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023]
Abstract
The objectives of our study were to investigate the possible effect of Simvastatin in ameliorating high cholesterol diet (HCD)-induced neurodegeneration and to also investigate its possible action on coagulation mediators. In silico and in vitro studies were performed to evaluate the impact of Simvastatin on prime coagulation mediators. HCD was used to induce neuropathology in wistar rats and histopathological and immunohistochemical studies were performed to evaluate the efficacy of Simvastatin in preventing the advancement of neurodegeneration in obese rats. Biochemical analyses were used to estimate changes in lipid profile, oxidative stress, inflammatory and coagulation markers. Simvastatin showed good theoretical affinity to coagulation proteins, significantly reversed changes in inflammatory and coagulation biomarkers which were induced by HCD. Enhanced fibrinolytic activity of Simvastatin was revealed through in vitro analysis. Immunohistoanalysis showed raised level of Nrf2. Histopathological studies also supported neuroprotective potential of Simvastatin in HCD fed rats. Simvastatin demonstrated reduced hypercoagulation, enhanced fibrinolysis and reversed neurodegeneration in HCD exposed rats suggesting its potential role in preventing the progression of neurodegeneration in obesity.
Collapse
Affiliation(s)
- Sana Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Noorul Hasan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Saba Khan
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Mymoona Akhtar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Mohammad Akhtar
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
23
|
Youssef ME, Yahya G, Popoviciu MS, Cavalu S, Abd-Eldayem MA, Saber S. Unlocking the Full Potential of SGLT2 Inhibitors: Expanding Applications beyond Glycemic Control. Int J Mol Sci 2023; 24:ijms24076039. [PMID: 37047011 PMCID: PMC10094124 DOI: 10.3390/ijms24076039] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
The number of diabetic patients has risen dramatically in recent decades, owing mostly to the rising incidence of type 2 diabetes mellitus (T2DM). Several oral antidiabetic medications are used for the treatment of T2DM including, α-glucosidases inhibitors, biguanides, sulfonylureas, meglitinides, GLP-1 receptor agonists, PPAR-γ agonists, DDP4 inhibitors, and SGLT2 inhibitors. In this review we focus on the possible effects of SGLT2 inhibitors on different body systems. Beyond the diabetic state, SGLT2 inhibitors have revealed a demonstrable ability to ameliorate cardiac remodeling, enhance myocardial function, and lower heart failure mortality. Additionally, SGLT2 inhibitors can modify adipocytes and their production of cytokines, such as adipokines and adiponectin, which enhances insulin sensitivity and delays diabetes onset. On the other hand, SGLT2 inhibitors have been linked to decreased total hip bone mineral deposition and increased hip bone resorption in T2DM patients. More data are needed to evaluate the role of SGLT2 inhibitors on cancer. Finally, the effects of SGLT2 inhibitors on neuroprotection appear to be both direct and indirect, according to scientific investigations utilizing various experimental models. SGLT2 inhibitors improve vascular tone, elasticity, and contractility by reducing oxidative stress, inflammation, insulin signaling pathways, and endothelial cell proliferation. They also improve brain function, synaptic plasticity, acetylcholinesterase activity, and reduce amyloid plaque formation, as well as regulation of the mTOR pathway in the brain, which reduces brain damage and cognitive decline.
Collapse
Affiliation(s)
- Mahmoud E Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Al Sharqia 44519, Egypt
| | - Mihaela Simona Popoviciu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| | - Marwa A Abd-Eldayem
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta 34518, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| |
Collapse
|
24
|
Zhang Z, Dalan R, Hu Z, Wang JW, Chew NW, Poh KK, Tan RS, Soong TW, Dai Y, Ye L, Chen X. Reactive Oxygen Species Scavenging Nanomedicine for the Treatment of Ischemic Heart Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202169. [PMID: 35470476 DOI: 10.1002/adma.202202169] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/08/2022] [Indexed: 06/14/2023]
Abstract
Ischemic heart disease (IHD) is the leading cause of disability and mortality worldwide. Reactive oxygen species (ROS) have been shown to play key roles in the progression of diabetes, hypertension, and hypercholesterolemia, which are independent risk factors that lead to atherosclerosis and the development of IHD. Engineered biomaterial-based nanomedicines are under extensive investigation and exploration, serving as smart and multifunctional nanocarriers for synergistic therapeutic effect. Capitalizing on cell/molecule-targeting drug delivery, nanomedicines present enhanced specificity and safety with favorable pharmacokinetics and pharmacodynamics. Herein, the roles of ROS in both IHD and its risk factors are discussed, highlighting cardiovascular medications that have antioxidant properties, and summarizing the advantages, properties, and recent achievements of nanomedicines that have ROS scavenging capacity for the treatment of diabetes, hypertension, hypercholesterolemia, atherosclerosis, ischemia/reperfusion, and myocardial infarction. Finally, the current challenges of nanomedicines for ROS-scavenging treatment of IHD and possible future directions are discussed from a clinical perspective.
Collapse
Affiliation(s)
- Zhan Zhang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 408433, Singapore
| | - Zhenyu Hu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jiong-Wei Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Nicholas Ws Chew
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Kian-Keong Poh
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Ru-San Tan
- Department of Cardiology, National Heart Centre Singapore, Singapore, 119609, Singapore
| | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macao, Taipa, Macau SAR, 999078, China
| | - Lei Ye
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Chemical and Biomolecular Engineering and Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
25
|
Cheng H, Zhang Z, Zhang B, Zhang W, Wang J, Ni W, Miao Y, Liu J, Bi Y. Enhancement of Impaired Olfactory Neural Activation and Cognitive Capacity by Liraglutide, but Not Dapagliflozin or Acarbose, in Patients With Type 2 Diabetes: A 16-Week Randomized Parallel Comparative Study. Diabetes Care 2022; 45:1201-1210. [PMID: 35263425 DOI: 10.2337/dc21-2064] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 02/11/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The comparative neuroprotective effects of different antidiabetes drugs have not been characterized in randomized controlled trials. Here, we investigated the therapeutic effects of liraglutide, dapagliflozin, or acarbose treatment on brain functional alterations and cognitive changes in patients with type 2 diabetes. RESEARCH DESIGN AND METHODS Thirty-six patients with type 2 diabetes inadequately controlled with metformin monotherapy were randomized to receive liraglutide, dapagliflozin, or acarbose treatment for 16 weeks. Brain functional MRI (fMRI) scan and a battery of cognitive assessments were evaluated pre- and postintervention in all subjects. RESULTS The 16-week treatment with liraglutide significantly enhanced the impaired odor-induced left hippocampal activation with Gaussian random field correction and improved cognitive subdomains of delayed memory, attention, and executive function (all P < 0.05), whereas dapagliflozin or acarbose did not. Structural equation modeling analysis demonstrated that such improvements of brain health and cognitive function could be partly ascribed to a direct effect of liraglutide on left hippocampal activation (β = 0.330, P = 0.022) and delayed memory (β = 0.410, P = 0.004) as well as to the metabolic ameliorations of reduced waist circumference, decreased body fat ratio, and elevated fasting insulin (all P < 0.05). CONCLUSIONS Our head-to-head study demonstrated that liraglutide enhanced impaired brain activation and restored impaired cognitive domains in patients with type 2 diabetes, whereas dapagliflozin and acarbose did not. The results expand the clinical application of liraglutide and provide a novel treatment strategy for individuals with diabetes and a high risk of cognitive decline.
Collapse
Affiliation(s)
- Haiyan Cheng
- Department of Endocrinology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China.,Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.,Department of Endocrinology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Zhou Zhang
- Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wen Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jin Wang
- Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wenyu Ni
- Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yingwen Miao
- Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiani Liu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yan Bi
- Department of Endocrinology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China.,Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
26
|
Ashrafizadeh M, Kumar AP, Aref AR, Zarrabi A, Mostafavi E. Exosomes as Promising Nanostructures in Diabetes Mellitus: From Insulin Sensitivity to Ameliorating Diabetic Complications. Int J Nanomedicine 2022; 17:1229-1253. [PMID: 35340823 PMCID: PMC8943613 DOI: 10.2147/ijn.s350250] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/07/2022] [Indexed: 12/11/2022] Open
Abstract
Diabetes mellitus (DM) is among the chronic metabolic disorders that its incidence rate has shown an increase in developed and wealthy countries due to lifestyle and obesity. The treatment of DM has always been of interest, and significant effort has been made in this field. Exosomes belong to extracellular vesicles with nanosized features (30-150 nm) that are involved in cell-to-cell communication and preserving homeostasis. The function of exosomes is different based on their cargo, and they may contain lipids, proteins, and nucleic acids. The present review focuses on the application of exosomes in the treatment of DM; both glucose and lipid levels are significantly affected by exosomes, and these nanostructures enhance lipid metabolism and decrease its deposition. Furthermore, exosomes promote glucose metabolism and affect the level of glycolytic enzymes and glucose transporters in DM. Type I DM results from the destruction of β cells in the pancreas, and exosomes can be employed to ameliorate apoptosis and endoplasmic reticulum (ER) stress in these cells. The exosomes have dual functions in mediating insulin resistance/sensitivity, and M1 macrophage-derived exosomes inhibit insulin secretion. The exosomes may contain miRNAs, and by transferring among cells, they can regulate various molecular pathways such as AMPK, PI3K/Akt, and β-catenin to affect DM progression. Noteworthy, exosomes are present in different body fluids such as blood circulation, and they can be employed as biomarkers for the diagnosis of diabetic patients. Future studies should focus on engineering exosomes derived from sources such as mesenchymal stem cells to treat DM as a novel strategy.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, 34956, Istanbul, Turkey
| | - Alan Prem Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Translational Sciences, Xsphera Biosciences Inc., Boston, MA, 02210, USA
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkey
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
27
|
Khan S, Pati S, Singh S, Akhtar M, Khare P, Khan S, Shafi S, Najmi AK. Targeting hypercoagulation to alleviate Alzheimer's disease progression in metabolic syndrome. Int J Obes (Lond) 2022; 46:245-254. [PMID: 34686782 DOI: 10.1038/s41366-021-00977-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 09/17/2021] [Accepted: 09/27/2021] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Metabolic Syndrome (MetS) constitutes an important risk factor for Alzheimer's disease (AD); however, the mechanism linking these two disorders has not been completely elucidated. Hence, hypercoagulation may account for the missing hallmark connecting MetS and AD. The present review proposes how hemostatic imbalance triggered in MetS advances in the context of AD. MetS causes interruption of insulin signaling and inflammation, inciting insulin resistance in the brain. Subsequently, neuroinflammation and brain endothelial dysfunction are prompted that further intensify the exorbitant infiltration of circulating lipids and platelet aggregation, thereby causing hypercoagulable state, impairing fibrinolysis and eventually inducing prothrombic state in the brain leading to neurodegeneration. OBJECTIVE This study aims to understand the role of hypercoagulation in triggering the progression of neurodegeneration in MetS. It also offers a few interventions to prevent the progression of AD in MetS targeting hypercoagulation. METHODS Literature studies based on MetS related neurodegeneration, the impact of coagulation on aggravating obesity and AD via the mechanisms of BBB disruption, neuroinflammation, and hypofibrinolysis. CONCLUSION The present paper proposes the hypothesis that hypercoagulation might amplify MetS associated insulin resistance, neuroinflammation, BBB disruption, and amyloid beta accumulation which eventually leads to AD.
Collapse
Affiliation(s)
- Sana Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Soumya Pati
- Translational Neurobiology Laboratory. Host Pathogen Interactions & Disease Modeling Group, Dept. of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, Pin-201314, UP, India
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Piush Khare
- Wave Pharma Regulatory Services Limited, New Delhi, India
| | - Saba Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Sadat Shafi
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
28
|
Rizzo MR, Di Meo I, Polito R, Auriemma MC, Gambardella A, di Mauro G, Capuano A, Paolisso G. Cognitive impairment and type 2 diabetes mellitus: Focus of SGLT2 inhibitors treatment. Pharmacol Res 2022; 176:106062. [PMID: 35017046 DOI: 10.1016/j.phrs.2022.106062] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 02/09/2023]
Abstract
Gliflozins are a novel class of oral anti-diabetic drugs, acting as inhibitors of sodium-glucose co-transporters (SGLTs) through the proximal convoluted tubules (PCT) and intestinal epithelium. The sodium-glucose co-transporters 2 (SGLT2) are mainly expressed in S1 and S2 segments of the proximal convoluted tubule in the kidneys. Clinical guidelines recommend their use especially in Type 2 Diabetes mellitus (T2DM) patients with vascular complications and/or heart failure highlighting the importance of sodium-glucose co-transporter 2 inhibitors (SGLT2i) pleiotropic effects. Interestingly, cognitive decline is a widely recognized complication of T2DM and, in addition, to clarify its pathophysiology, there is an urgent need to understand how and if diabetes therapies can control diabetes-related cognitive dysfunction. At the time, although SGLT2 proteins are present in the Central Nervous System (CNS), the SGLT2i effects on cognitive impairments remain partly unknown. In pre-clinical studies, SGLT2i ameliorates cognitive dysfunction in obese and T2DM mice, reducing oxidative stress, neuroinflammation and improving neuronal plasticity and mitochondrial brain pathway. In addition, SGLT2i could bring back mTOR to a physiological state of activation, stopping neurodegenerative diseases' onset or progression. Instead, clinical studies on T2DM-related cognitive dysfunction treated by SGLT2i are much more limited. For these reasons, further studies are needed to better elucidate if SGLT2i therapy can affect T2DM-related cognitive decline. In this scenario, this review aims to summarize the state of knowledge on the role of SGLT2i in T2DM-related cognitive dysfunction and stimulate new clinical trials.
Collapse
Affiliation(s)
- Maria Rosaria Rizzo
- Department of Advanced Medical and Surgical Sciences - University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Irene Di Meo
- Department of Advanced Medical and Surgical Sciences - University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Rita Polito
- Department of Advanced Medical and Surgical Sciences - University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Maria Chiara Auriemma
- Department of Advanced Medical and Surgical Sciences - University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Antonio Gambardella
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Gabriella di Mauro
- Department of Experimental Medicine - Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine - Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences - University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| |
Collapse
|