1
|
Ong LT, Sia CH. Interactions between antidiabetes medications and heart-brain axis. Curr Opin Endocrinol Diabetes Obes 2025; 32:34-43. [PMID: 39639832 DOI: 10.1097/med.0000000000000896] [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: 12/07/2024]
Abstract
PURPOSE OF REVIEW The heart - brain axis (HBA) is the physiological interactions between the cardiovascular and nervous systems through autonomic nerves, hormones, and cytokines. Patients diagnosed with diabetes mellitus have an increased risk of the cardiovascular and neurological diseases. However, recent evidence demonstrated that different antidiabetic drugs may delay cognitive impairment and improve cardiovascular outcomes. This review examines the impact of antidiabetic drugs on the HBA in patients with diabetes. RECENT FINDINGS Metformin improves the cardiovascular and cognitive outcomes through adenosine 5'-monophosphate-activated protein kinase activation. Sodium-glucose cotransporter-2 inhibitors reduce inflammation, oxidative stress by inhibiting the NLRP3 inflammasome thereby reducing the incidence of heart failure and formation of beta-amyloid and neurofibrillary tangles in the brain. Dipeptidyl peptidase-4 inhibitors exhibit neuroprotective effects in Alzheimer's disease by reducing amyloid-beta and tau pathology and inflammation but may exacerbate heart failure risk due to increased sympathetic activity and prolonged β-adrenergic stimulation. Glucagon-like peptide-1 receptor agonists exhibit neuroprotective effects in Alzheimer's and Parkinson's diseases by reducing neuroinflammation, but may increase sympathetic activity, potentially elevating heart rate and blood pressure, despite their cardioprotective benefits. SUMMARY Antidiabetes medications have the potential to improve cardiovascular and cognitive outcomes; however, additional studies are required to substantiate these effects.
Collapse
Affiliation(s)
- Leong Tung Ong
- Department of Cardiology, National University Heart Centre, Singapore
| | | |
Collapse
|
2
|
Ríos JA, Bórquez JC, Godoy JA, Zolezzi JM, Furrianca MC, Inestrosa NC. Emerging role of Metformin in Alzheimer's disease: A translational view. Ageing Res Rev 2024; 100:102439. [PMID: 39074563 DOI: 10.1016/j.arr.2024.102439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 07/31/2024]
Abstract
Alzheimer's disease (AD) constitutes a major public-health issue of our time. Regrettably, despite our considerable understanding of the pathophysiological aspects of this disease, current interventions lead to poor outcomes. Furthermore, experimentally promising compounds have continuously failed when translated to clinical trials. Along with increased population ageing, Type 2 Diabetes Mellitus (T2DM) has become an extremely common condition, mainly due to unbalanced dietary habits. Substantial epidemiological evidence correlates T2DM with cognitive impairment as well. Considering that brain insulin resistance, mitochondrial dysfunction, oxidative stress, and amyloidogenesis are common phenomena, further approaching the common features among these pathological conditions. Metformin constitutes the first-choice drug to preclude insulin resistance in T2DM clinical management. Experimental evidence suggests that its functions might include neuroprotective effects, in addition to its hypoglycemic activity. This review aims to summarize and discuss current knowledge of experimental data on metformin on this path towards translational medicine. Finally, we discuss the controversial data of responses to metformin in vitro, and in vivo, animal models and human studies.
Collapse
Affiliation(s)
- Juvenal A Ríos
- Facultad de Medicina y Ciencia, Escuela de Medicina, Universidad San Sebastián, Santiago, Chile
| | - Juan Carlos Bórquez
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile; Facultad de Ciencias de la Salud, Universidad de Magallanes, Punta Arenas, Chile
| | - Juan A Godoy
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan M Zolezzi
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | | | - Nibaldo C Inestrosa
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
3
|
Feng L, Gao L. The role of neurovascular coupling dysfunction in cognitive decline of diabetes patients. Front Neurosci 2024; 18:1375908. [PMID: 38576869 PMCID: PMC10991808 DOI: 10.3389/fnins.2024.1375908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Neurovascular coupling (NVC) is an important mechanism to ensure adequate blood supply to active neurons in the brain. NVC damage can lead to chronic impairment of neuronal function. Diabetes is characterized by high blood sugar and is considered an important risk factor for cognitive impairment. In this review, we provide fMRI evidence of NVC damage in diabetic patients with cognitive decline. Combined with the exploration of the major mechanisms and signaling pathways of NVC, we discuss the effects of chronic hyperglycemia on the cellular structure of NVC signaling, including key receptors, ion channels, and intercellular connections. Studying these diabetes-related changes in cell structure will help us understand the underlying causes behind diabetes-induced NVC damage and early cognitive decline, ultimately helping to identify the most effective drug targets for treatment.
Collapse
Affiliation(s)
| | - Ling Gao
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
4
|
Rosell-Díaz M, Fernández-Real JM. Metformin, Cognitive Function, and Changes in the Gut Microbiome. Endocr Rev 2024; 45:210-226. [PMID: 37603460 PMCID: PMC10911951 DOI: 10.1210/endrev/bnad029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/03/2023] [Accepted: 08/16/2023] [Indexed: 08/23/2023]
Abstract
The decline in cognitive function and the prevalence of neurodegenerative disorders are among the most serious threats to health in old age. The prevalence of dementia has reached 50 million people worldwide and has become a major public health problem. The causes of age-related cognitive impairment are multiple, complex, and difficult to determine. However, type 2 diabetes (T2D) is linked to an enhanced risk of cognitive impairment and dementia. Human studies have shown that patients with T2D exhibit dysbiosis of the gut microbiota. This dysbiosis may contribute to the development of insulin resistance and increased plasma lipopolysaccharide concentrations. Metformin medication mimics some of the benefits of calorie restriction and physical activity, such as greater insulin sensitivity and decreased cholesterol levels, and hence may also have a positive impact on aging in humans. According to recent human investigations, metformin might partially restore gut dysbiosis related to T2D. Likewise, some studies showed that metformin reduced the risk of dementia and improved cognition, although not all studies are concordant. Therefore, this review focused on those human studies describing the effects of metformin on the gut microbiome (specifically the changes in taxonomy, function, and circulating metabolomics), the changes in cognitive function, and their possible bidirectional implications.
Collapse
Affiliation(s)
- Marisel Rosell-Díaz
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- CIBERobn Fisiopatología de la Obesidad y Nutrición, 28029 Madrid, Spain
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- CIBERobn Fisiopatología de la Obesidad y Nutrición, 28029 Madrid, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
| |
Collapse
|
5
|
Liu J, Zhang M, Deng D, Zhu X. The function, mechanisms, and clinical applications of metformin: potential drug, unlimited potentials. Arch Pharm Res 2023; 46:389-407. [PMID: 36964307 DOI: 10.1007/s12272-023-01445-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 03/08/2023] [Indexed: 03/26/2023]
Abstract
Metformin has been used clinically for more than 60 years. As time goes by, more and more miraculous effects of metformin beyond the clinic have been discovered and discussed. In addition to the clinically approved hypoglycemic effect, it also has a positive metabolic regulation effect on the human body that cannot be ignored. Such as anti-cancer, anti-aging, brain repair, cardiovascular protection, gastrointestinal regulation, hair growth and inhibition of thyroid nodules, and other nonclinical effects. Metformin affects almost the entire body in the situation taking it over a long period, and the preventive effects of metformin in addition to treating diabetes are also beginning to be recommended in some guidelines. This review is mainly composed of four parts: the development history of metformin, the progress of clinical efficacy, the nonclinical efficacy of metformin, and the consideration and prospect of its application.
Collapse
Affiliation(s)
- Jianhong Liu
- Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou Medical College, Hangzhou, China
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Ming Zhang
- Department of Physical Medicine and Rehabilitation, Zibo Central Hospital, Zibo, China
| | - Dan Deng
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Xiao Zhu
- Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou Medical College, Hangzhou, China.
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China.
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China.
| |
Collapse
|
6
|
Mechanism of metformin regulation in central nervous system: Progression and future perspectives. Biomed Pharmacother 2022; 156:113686. [DOI: 10.1016/j.biopha.2022.113686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/20/2022] Open
|
7
|
Gillotin S, Sahni V, Lepko T, Hanspal MA, Swartz JE, Alexopoulou Z, Marshall FH. Targeting impaired adult hippocampal neurogenesis in ageing by leveraging intrinsic mechanisms regulating Neural Stem Cell activity. Ageing Res Rev 2021; 71:101447. [PMID: 34403830 DOI: 10.1016/j.arr.2021.101447] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/14/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023]
Abstract
Deficits in adult neurogenesis may contribute to the aetiology of many neurodevelopmental, psychiatric and neurodegenerative diseases. Genetic ablation of neurogenesis provides proof of concept that adult neurogenesis is required to sustain complex and dynamic cognitive functions, such as learning and memory, mostly by providing a high degree of plasticity to neuronal circuits. In addition, adult neurogenesis is reactive to external stimuli and the environment making it particularly susceptible to impairment and consequently contributing to comorbidity. In the human brain, the dentate gyrus of the hippocampus is the main active source of neural stem cells that generate granule neurons throughout life. The regulation and preservation of the pool of neural stem cells is central to ensure continuous and healthy adult hippocampal neurogenesis (AHN). Recent advances in genetic and metabolic profiling alongside development of more predictive animal models have contributed to the development of new concepts and the emergence of molecular mechanisms that could pave the way to the implementation of new therapeutic strategies to treat neurological diseases. In this review, we discuss emerging molecular mechanisms underlying AHN that could be embraced in drug discovery to generate novel concepts and targets to treat diseases of ageing including neurodegeneration. To support this, we review cellular and molecular mechanisms that have recently been identified to assess how AHN is sustained throughout life and how AHN is associated with diseases. We also provide an outlook on strategies for developing correlated biomarkers that may accelerate the translation of pre-clinical and clinical data and review clinical trials for which modulation of AHN is part of the therapeutic strategy.
Collapse
|
8
|
Abdi M, Pasbakhsh P, Shabani M, Nekoonam S, Sadeghi A, Fathi F, Abouzaripour M, Mohamed W, Zibara K, Kashani IR, Zendedel A. Metformin Therapy Attenuates Pro-inflammatory Microglia by Inhibiting NF-κB in Cuprizone Demyelinating Mouse Model of Multiple Sclerosis. Neurotox Res 2021; 39:1732-1746. [PMID: 34570348 DOI: 10.1007/s12640-021-00417-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/30/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is a chronic disorder characterized by reactive gliosis, inflammation, and demyelination. Microglia plays a crucial role in the pathogenesis of MS and has the dynamic plasticity to polarize between pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes. Metformin, a glucose-lowering drug, attenuates inflammatory responses by activating adenosine monophosphate protein kinase (AMPK) which suppresses nuclear factor kappa B (NF-κB). In this study, we indirectly investigated whether metformin therapy would regulate microglia activity in the cuprizone (CPZ)-induced demyelination mouse model of MS via measuring the markers associated with pro- and anti-inflammatory microglia. Evaluation of myelin by luxol fast blue staining revealed that metformin treatment (CPZ + Met) diminished demyelination, in comparison to CPZ mice. In addition, metformin therapy significantly alleviated reactive microgliosis and astrogliosis in the corpus callosum, as measured by Iba-1 and GFAP staining. Moreover, metformin treatment significantly downregulated the expression of pro-inflammatory associated genes (iNOS, H2-Aa, and TNF-α) in the corpus callosum, whereas expression of anti-inflammatory markers (Arg1, Mrc1, and IL10) was not promoted, compared to CPZ mice. Furthermore, protein levels of iNOS (pro-inflammatory marker) were significantly decreased in the metformin group, while those of Trem2 (anti-inflammatory marker) were increased. In addition, metformin significantly increased AMPK activation in CPZ mice. Finally, metformin administration significantly reduced the activation level of NF-κB in CPZ mice. In summary, our data revealed that metformin attenuated pro-inflammatory microglia markers through suppressing NF-κB activity. The positive effects of metformin on microglia and remyelination suggest that it could be used as a promising candidate to lessen the incidence of inflammatory neurodegenerative diseases such as MS.
Collapse
Affiliation(s)
- Mahdad Abdi
- Department of Anatomy, school of medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomy, school of medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Maryam Shabani
- Department of Clinical Biochemistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Saied Nekoonam
- Department of Anatomy, school of medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Asie Sadeghi
- Department of Clinical Biochemistry, Faculty of medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Fardin Fathi
- Cellular and Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | | | - Wael Mohamed
- Basic Medical Science Department, International Islamic University Malaysia, Pahang, Malaysia.,Clinical Pharmacology Department, Menoufia Medical School, Menoufia University, Shebin El Kom, Egypt
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon.
| | - Iraj Ragerdi Kashani
- Department of Anatomy, school of medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Adib Zendedel
- Institute of Neuroanatomy, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
9
|
Qin X, Zhang X, Li P, Wang M, Yan L, Bao Z, Liu Q. Association Between Diabetes Medications and the Risk of Parkinson's Disease: A Systematic Review and Meta-Analysis. Front Neurol 2021; 12:678649. [PMID: 34349721 PMCID: PMC8326375 DOI: 10.3389/fneur.2021.678649] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/15/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Diabetes mellitus (DM) increases the risk of Parkinson's disease (PD). However, whether DM medications play a part on that increased PD risk is unclear. We designed this meta-analysis to assess the influence of different oral DM medications on the PD risk in patients with DM. Methods: We searched PubMed, Embase, and CENTRAL databases for relevant studies up until January 2021. We pooled adjusted outcomes to assess the PD risk in patients using different DM medications including sulfonylurea, metformin, glitazones (GTZ), dipeptidyl peptidase-4 inhibitors (DPP4i), and glucagon-like peptide-1 agonists (GLP1a). Results: We included 10 studies in our analysis. Our results indicate a lack of significant association between the PD risk and the use of sulfonylureas (three studies; HR, 1.26; 95% CI, 0.95 to 1.66; I2, 70%; p = 0.11), DPP4i (three studies; HR, 0.69; 95% CI, 0.35 to 1.38; I2, 88%; p = 0.30), metformin (five studies; HR, 1.23; 95% CI, 0.98 to 1.78; I2, 84%; p = 0.13), and GTZ (six studies; HR, 0.88; 95% CI, 0.66 to 1.16; I2, 92%; p = 0.35). After exclusion of a single study in the GTZ analysis, our results indicate a significantly reduced PD risk with GTZ use (HR, 0.78; 95% CI, 0.65 to 0.93; I2, 59%; p = 0.06). Similarly, after the exclusion of a single study, our results indicate a significantly increased PD risk with the use of metformin (HR, 1.50; 95% CI, 1.11 to 2.02; I2, 80%; p = 0.008). We also found a significantly reduced PD risk with the use of GLP1a (two studies; HR, 0.41; 95% CI, 0.19 to 0.87; I2, 0%; p = 0.02). Conclusion: The role of different DM medications on the PD risk remains unclear, and the quality of studies is low. While our analysis suggests a lack of association between the use of metformin, GTZ, DPP4i, and sulfonylureas and the PD risk, metformin (to a higher degree) and GTZ may still increase the risk. Limited data suggest a protective effect of GLP1a on the PD risk.
Collapse
Affiliation(s)
- Xiaocui Qin
- Department of Physiology, Zhaoqing Medical College, Zhaoqing, China
| | - Xia Zhang
- Department of Pathology and Physiology, Zhaoqing Medical College, Zhaoqing, China
| | - Pinyu Li
- Department of Pathology and Physiology, Zhaoqing Medical College, Zhaoqing, China
| | - Min Wang
- Department of Pathology and Physiology, Zhaoqing Medical College, Zhaoqing, China
| | - Li Yan
- Department of Pharmacology, Zhaoqing Medical College, Zhaoqing, China
| | - Zeqing Bao
- Department of Pharmacology, Zhaoqing Medical College, Zhaoqing, China
| | - Qili Liu
- Department of Physiology, Zhaoqing Medical College, Zhaoqing, China
| |
Collapse
|
10
|
Tang BL. Could metformin be therapeutically useful in Huntington's disease? Rev Neurosci 2020; 31:297-317. [PMID: 31751298 DOI: 10.1515/revneuro-2019-0072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022]
Abstract
Emerging evidence suggest that dimethylbiguanide (metformin), a first-line drug for type 2 diabetes mellitus, could be neuroprotective in a range of brain pathologies, which include neurodegenerative diseases and brain injury. However, there are also contraindications that associate metformin treatment with cognitive impairment as well as adverse outcomes in Alzheimer's disease and Parkinson's disease animal models. Recently, a beneficial effect of metformin in animal models of Huntington's disease (HD) has been strengthened by multiple reports. In this brief review, the findings associated with the effects of metformin in attenuating neurodegenerative diseases are discussed, focusing on HD-associated pathology and the potential underlying mechanisms highlighted by these studies. The mechanism of action of metformin is complex, and its therapeutic efficacy is therefore expected to be dependent on the disease context. The key metabolic pathways that are effectively affected by metformin, such as AMP-activated protein kinase activation, may be altered in the later decades of the human lifespan. In this regard, metformin may nonetheless be therapeutically useful for neurological diseases with early pathological onsets, such as HD.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, Singapore 117596, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Medical Drive, Singapore 119077, Singapore
| |
Collapse
|
11
|
|
12
|
Guo X, Shi Y, Du P, Wang J, Han Y, Sun B, Feng J. HMGB1/TLR4 promotes apoptosis and reduces autophagy of hippocampal neurons in diabetes combined with OSA. Life Sci 2019; 239:117020. [PMID: 31678553 DOI: 10.1016/j.lfs.2019.117020] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 10/11/2019] [Accepted: 10/26/2019] [Indexed: 12/27/2022]
Abstract
AIMS Obstructive sleep apnea (OSA) combined with type 2 diabetes (T2DM) may lead to cognitive dysfunction. We previously reported that cognitive impairment is exacerbated in KKAy mice exposed to intermittent hypoxia (IH), during which the DNA binding protein HMGB1 mediates hippocampal neuronal apoptosis by maintaining microglia-associated neuroinflammation, but the underlying mechanism remains largely unknown. MATERIALS AND METHODS We performed immunofluorescence, Western blotting, and immunohistochemistry experiments in mouse hippocampal tissues and HT22 cells. KKAy type 2 diabetes model mice and normal C57BL/6J mice were exposed to IH or intermittent normoxia. HT22 cells were cultured in high glucose medium and exposed to IH or intermittent normoxia. We transfected HMGB1 siRNA into HT22 cells and then treated them with high glucose combined with intermittent hypoxia. KEY FINDINGS In conclusion, IH aggravated apoptosis and autophagy defects in T2DM mice, and increased the protein expression of HMGB1 and TLR4. This was also confirmed in HG + IH-treated hippocampal HT22 cells. HMGB1 siRNA can significantly reduce the protein expression of HMGB1 and TLR4, reverse neuronal apoptosis and enhance autophagy. SIGNIFICANCE We believe that HMGB1 is a key factor in the regulation of hippocampal neuronal apoptosis and autophagy defects in T2DM combined with OSA. Targeting HMGB1/TLR4 signaling as a novel approach may delay or prevent the increased apoptosis and decreased autophagy induced by T2DM combined with OSA, and may ultimately improve cognitive dysfunction.
Collapse
Affiliation(s)
- Xiangyu Guo
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052, Tianjin, China
| | - Yu Shi
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052, Tianjin, China
| | - Ping Du
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052, Tianjin, China
| | - Jiahui Wang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052, Tianjin, China
| | - Yelei Han
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052, Tianjin, China
| | - Bei Sun
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin, 300134, China.
| | - Jing Feng
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052, Tianjin, China.
| |
Collapse
|
13
|
Del Ser T, Zea MA, Valentí M, Olazarán J, López-Álvarez J, Rebollo-Vázquez A, Ávila-Villanueva M, Frades B, Medina M, Fernández-Blázquez MA. Effects of commonly prescribed drugs on cognition and mild cognitive impairment in healthy elderly people. J Psychopharmacol 2019; 33:965-974. [PMID: 31241413 DOI: 10.1177/0269881119857206] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Chronic drug intake has been associated with negative and positive cognitive effects in elderly people, although subjacent conditions may be confounding factors. AIM To study the effects on cognitive performance of commonly prescribed medications in a cohort of cognitively normal older adults. METHODS Medication intake was recorded during two years in 1087 individuals 70-85 years old, without neurological or psychiatric conditions. The influence of every drug, drug family and therapeutic group on six cognitive scores and on the conversion to mild cognitive impairment over two years was ascertained by cross-sectional and longitudinal analyses controlling for demographic and clinical variables. RESULTS Small effects of several drugs on information processing were found in cross-sectional analyses but only confirmed for a positive effect of vitamin D in case-control analyses. Longitudinal analyses showed no drug effects on the cognitive slopes. Several hypotensive drugs reduced, whereas bromazepam and glucose lowering drugs increased, the conversion rate to mild cognitive impairment with very small effects (R2=0.3-1%). CONCLUSIONS Cognitively healthy elderly individuals show minimal negative effects on information processing associated with chronic intake of some drugs probably related to the subjacent condition. Some drugs slightly affect the rate of conversion to mild cognitive impairment. Positive effects of vitamin D, chondroitin, atorvastatin and antihypertensive drugs, and negative effects of antidepressants and benzodiazepines, should be further explored in studies with longer follow-up.
Collapse
Affiliation(s)
- Teodoro Del Ser
- 1 Alzheimer's Disease Investigation Research Unit, CIEN Foundation, Carlos III Institute of Health, Queen Sofia Foundation, Alzheimer Research Centre, Madrid, Spain
| | - María-Ascensión Zea
- 1 Alzheimer's Disease Investigation Research Unit, CIEN Foundation, Carlos III Institute of Health, Queen Sofia Foundation, Alzheimer Research Centre, Madrid, Spain
| | - Meritxell Valentí
- 1 Alzheimer's Disease Investigation Research Unit, CIEN Foundation, Carlos III Institute of Health, Queen Sofia Foundation, Alzheimer Research Centre, Madrid, Spain
| | - Javier Olazarán
- 1 Alzheimer's Disease Investigation Research Unit, CIEN Foundation, Carlos III Institute of Health, Queen Sofia Foundation, Alzheimer Research Centre, Madrid, Spain.,2 Service of Neurology, University Hospital Gregorio Marañón, Madrid, Spain
| | - Jorge López-Álvarez
- 1 Alzheimer's Disease Investigation Research Unit, CIEN Foundation, Carlos III Institute of Health, Queen Sofia Foundation, Alzheimer Research Centre, Madrid, Spain.,3 Service of Psychiatry, University Hospital 12 de Octubre, Madrid, Spain
| | - Ana Rebollo-Vázquez
- 1 Alzheimer's Disease Investigation Research Unit, CIEN Foundation, Carlos III Institute of Health, Queen Sofia Foundation, Alzheimer Research Centre, Madrid, Spain
| | - Marina Ávila-Villanueva
- 1 Alzheimer's Disease Investigation Research Unit, CIEN Foundation, Carlos III Institute of Health, Queen Sofia Foundation, Alzheimer Research Centre, Madrid, Spain
| | - Belén Frades
- 1 Alzheimer's Disease Investigation Research Unit, CIEN Foundation, Carlos III Institute of Health, Queen Sofia Foundation, Alzheimer Research Centre, Madrid, Spain
| | - Miguel Medina
- 1 Alzheimer's Disease Investigation Research Unit, CIEN Foundation, Carlos III Institute of Health, Queen Sofia Foundation, Alzheimer Research Centre, Madrid, Spain.,4 Centro de Investigación Biomédica en Red sobre Enfermedades Degenerativas (CIBERNED), Carlos III Institute of Health, Madrid, Spain
| | - Miguel A Fernández-Blázquez
- 1 Alzheimer's Disease Investigation Research Unit, CIEN Foundation, Carlos III Institute of Health, Queen Sofia Foundation, Alzheimer Research Centre, Madrid, Spain
| |
Collapse
|
14
|
Shi Q, Liu S, Fonseca VA, Thethi TK, Shi L. Effect of metformin on neurodegenerative disease among elderly adult US veterans with type 2 diabetes mellitus. BMJ Open 2019; 9:e024954. [PMID: 31366635 PMCID: PMC6677947 DOI: 10.1136/bmjopen-2018-024954] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE This study aimed to evaluate the association between metformin treatment and the risk of neurodegenerative disease (ND) among elderly adults with type 2 diabetes mellitus (T2DM). DESIGN/SETTING/PARTICIPANTS This retrospective longitudinal cohort study examined the effects of the length of metformin exposure on ND among elderly US veterans with T2DM and insulin treatment using the Veterans Affairs electronic medical record database. PRIMARY AND SECONDARY OUTCOME MEASURES The primary clinical outcome was defined as diagnosis of ND including dementia, Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and mild cognitive impairment during the follow-up period. The secondary clinical outcomes were separately measured by AD, PD, HD, dementia and mild cognitive impairment. RESULT Adjusted by propensity score weight, a total of 5528 patients (mean age, 63.2±10.9 years; male, 98%; white, 60%) with a median follow-up of 5.2 years were selected. Those with ND or other mental disorders at baseline or who were on insulin for less than two-thirds of the study period were excluded. The incidence rate of ND was 11.48 per 1000 person-years among patients with metformin treatment, compared with 25.45 per 1000 person-years for those without metformin. Compared with no metformin use, 2-4 years and >4 years of metformin exposure were significantly associated with lower risk of ND (adjusted HR (aHR)=0.62, 95% CI 0.45 to 0.85; aHR=0.19, 95% CI 0.12 to 0.31, respectively), while metformin exposure in the first 2 years showed no significant influence. CONCLUSION We conclude that long-term metformin therapy (>2 years) was associated with lower incidence of ND among elderly veterans with T2DM. We need to conduct a study with more representative population and more robust method for causal inferences. Further investigation into the mechanism involved is needed along with randomised trials to confirm a potential neuroprotective effect of metformin.
Collapse
Affiliation(s)
- Qian Shi
- Global Health Management and Policy, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Shuqian Liu
- Global Health Management and Policy, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Vivian A Fonseca
- Section of Endocrinology, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Endocrinology, Southeast Louisiana Veterans Health Care System, New Orleans, Louisiana, USA
| | - Tina K Thethi
- Section of Endocrinology, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Endocrinology, Southeast Louisiana Veterans Health Care System, New Orleans, Louisiana, USA
| | - Lizheng Shi
- Global Health Management and Policy, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
15
|
Halaris A, Leonard BE. Unraveling the complex interplay of immunometabolic systems that contribute to the neuroprogression of psychiatric disorders. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.npbr.2019.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
16
|
Mamo JC, Lam V, Brook E, Mooranian A, Al-Salami H, Fimognari N, Nesbit M, Takechi R. Probucol prevents blood-brain barrier dysfunction and cognitive decline in mice maintained on pro-diabetic diet. Diab Vasc Dis Res 2019; 16:87-97. [PMID: 30156119 DOI: 10.1177/1479164118795274] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
An emerging body of evidence consistently suggests that compromised blood-brain barrier integrity may be causally associated with cognitive decline induced by type-2 diabetes. Our previous studies demonstrated that selected anti-inflammatory/anti-oxidative agents can preserve the integrity of blood-brain barrier and prevent neuroinflammation in mouse models of dysfunctional blood-brain barrier. Therefore, we have tested whether the previously proven blood-brain barrier protective agent, probucol, can prevent blood-brain barrier breakdown and cognitive decline in a dietary-induced murine model of diabetic insulin resistance. After 6-month chronic ingestion of a diet high in fat and fructose, the mice became insulin resistant. The high-fat and high-fructose-fed mice showed significant cognitive decline assessed by Morris water maze, concomitant with significant elevations in cortical and hippocampal glial acidic fibrillary protein and Fluoro Jade-C staining, indicating heightened neuroinflammation and neurodegeneration, respectively. The integrity of blood-brain barrier in high-fat and high-fructose-fed mice was substantially compromised, and this showed a significant association with heightened neurodegeneration. Co-provision of probucol with high-fat and high-fructose diet completely prevented the cognitive decline and blood-brain barrier dysfunction. Similarly, metformin was able to restore the cognitive function in high-fat and high-fructose-fed mice, while its blood-brain barrier protective effects were modest. These data suggest that probucol may prevent cognitive decline induced by insulin resistance by preserving the integrity of blood-brain barrier, whereas metformin's neuroprotective effects may be mediated through a separate pathway.
Collapse
Affiliation(s)
- John Cl Mamo
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 2 School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Virginie Lam
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 2 School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Emily Brook
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 3 School of Biomedical Sciences, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Armin Mooranian
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 4 School of Pharmacy, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Hani Al-Salami
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 4 School of Pharmacy, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Nicholas Fimognari
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 2 School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Michael Nesbit
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 2 School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Ryusuke Takechi
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 2 School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| |
Collapse
|
17
|
Rad SK, Arya A, Karimian H, Madhavan P, Rizwan F, Koshy S, Prabhu G. Mechanism involved in insulin resistance via accumulation of β-amyloid and neurofibrillary tangles: link between type 2 diabetes and Alzheimer's disease. Drug Des Devel Ther 2018; 12:3999-4021. [PMID: 30538427 PMCID: PMC6255119 DOI: 10.2147/dddt.s173970] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The pathophysiological link between type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) has been suggested in several reports. Few findings suggest that T2DM has strong link in the development process of AD, and the complete mechanism is yet to be revealed. Formation of amyloid plaques (APs) and neurofibrillary tangles (NFTs) are two central hallmarks in the AD. APs are the dense composites of β-amyloid protein (Aβ) which accumulates around the nerve cells. Moreover, NFTs are the twisted fibers containing hyperphosphorylated tau proteins present in certain residues of Aβ that build up inside the brain cells. Certain factors contribute to the aetiogenesis of AD by regulating insulin signaling pathway in the brain and accelerating the formation of neurotoxic Aβ and NFTs via various mechanisms, including GSK3β, JNK, CamKII, CDK5, CK1, MARK4, PLK2, Syk, DYRK1A, PPP, and P70S6K. Progression to AD could be influenced by insulin signaling pathway that is affected due to T2DM. Interestingly, NFTs and APs lead to the impairment of several crucial cascades, such as synaptogenesis, neurotrophy, and apoptosis, which are regulated by insulin, cholesterol, and glucose metabolism. The investigation of the molecular cascades through insulin functions in brain contributes to probe and perceive progressions of diabetes to AD. This review elaborates the molecular insights that would help to further understand the potential mechanisms linking T2DM and AD.
Collapse
Affiliation(s)
- Sima Kianpour Rad
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Aditya Arya
- Department of Pharmacology and Therapeutics, School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia,
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia,
- Malaysian Institute of Pharmaceuticals and Nutraceuticals (IPharm), Bukit Gambir, Gelugor, Pulau Pinang, Malaysia,
| | - Hamed Karimian
- Department of Pharmacology and Therapeutics, School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia,
| | - Priya Madhavan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Farzana Rizwan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Shajan Koshy
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Girish Prabhu
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| |
Collapse
|
18
|
Zhong KL, Chen F, Hong H, Ke X, Lv YG, Tang SS, Zhu YB. New views and possibilities of antidiabetic drugs in treating and/or preventing mild cognitive impairment and Alzheimer's Disease. Metab Brain Dis 2018; 33:1009-1018. [PMID: 29626315 DOI: 10.1007/s11011-018-0227-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 03/28/2018] [Indexed: 12/13/2022]
Abstract
Mounting evidence suggests that diabetes mellitus (DM) is associated with mild cognitive impairment (MCI), vascular dementia and Alzheimer's disease (AD). Biological, clinical and epidemiological data support a close link between DM and AD. Increasingly, studies have found that several antidiabetic agents can promote neurogenesis, and clinically ameliorate cognitive and memory impairments in different clinical settings. Data has shown that these antidiabetic drugs positively affect mitochondrial and synaptic function, neuroinflammation, and brain metabolism. Evidence to date strongly suggests that these antidiabetic drugs could be developed as disease-modifying therapies for MCI and AD in patients with and without diabetes.
Collapse
Affiliation(s)
- Kai Long Zhong
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Fang Chen
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Hao Hong
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Xuan Ke
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Yang Ge Lv
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Su Su Tang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Yu Bing Zhu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
19
|
Schmitz L, Kuglin R, Bae-Gartz I, Janoschek R, Appel S, Mesaros A, Jakovcevski I, Vohlen C, Handwerk M, Ensenauer R, Dötsch J, Hucklenbruch-Rother E. Hippocampal insulin resistance links maternal obesity with impaired neuronal plasticity in adult offspring. Psychoneuroendocrinology 2018; 89:46-52. [PMID: 29324300 DOI: 10.1016/j.psyneuen.2017.12.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/20/2017] [Accepted: 12/27/2017] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Maternal obesity and a disturbed metabolic environment during pregnancy and lactation have been shown to result in many long-term health consequences for the offspring. Among them, impairments in neurocognitive development and performance belong to the most dreaded ones. So far, very few mechanistic approaches have aimed to determine the responsible molecular events. METHODS In a mouse model of maternal diet-induced obesity and perinatal hyperinsulinemia, we assessed adult offspring's hippocampal insulin signaling as well as concurrent effects on markers of hippocampal neurogenesis, synaptic plasticity and function using western blotting and immunohistochemistry. In search for a potential link between neuronal insulin resistance and hippocampal plasticity, we additionally quantified protein expression of key molecules of synaptic plasticity in an in vitro model of acute neuronal insulin resistance. RESULTS Maternal obesity and perinatal hyperinsulinemia result in adult hippocampal insulin resistance with subsequently reduced hippocampal mTor signaling and altered expression of markers of neurogenesis (doublecortin), synaptic plasticity (FoxO1, pSynapsin) and function (vGlut, vGAT) in the offspring. The observed effects are independent of the offspring's adult metabolic phenotype and can be associated with multiple previously reported behavioral abnormalities. Additionally, we demonstrate that induction of insulin resistance in cultured hippocampal neurons reduces mTor signaling, doublecortin and vGAT protein expression. CONCLUSIONS Hippocampal insulin resistance might play a key role in mediating the long-term effects of maternal obesity and perinatal hyperinsulinemia on hippocampal plasticity and the offspring's neurocognitive outcome.
Collapse
Affiliation(s)
- Lisa Schmitz
- Department of Pediatrics and Adolescent Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50924, Germany
| | - Rebecca Kuglin
- Department of Pediatrics and Adolescent Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50924, Germany
| | - Inga Bae-Gartz
- Department of Pediatrics and Adolescent Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50924, Germany
| | - Ruth Janoschek
- Department of Pediatrics and Adolescent Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50924, Germany
| | - Sarah Appel
- Department of Pediatrics and Adolescent Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50924, Germany
| | - Andrea Mesaros
- Max Planck Institute for Biology of Ageing, Phenotyping Core Facility, Joseph-Stelzmann-Str. 9b, Cologne, 50931, Germany
| | - Igor Jakovcevski
- Institute for Molecular and Behavioral Neuroscience, Center for Molecular Medicine Cologne, Cologne, Germany; Experimental Neurophysiology, German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Christina Vohlen
- Department of Pediatrics and Adolescent Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50924, Germany
| | - Marion Handwerk
- Department of Pediatrics and Adolescent Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50924, Germany
| | - Regina Ensenauer
- Experimental Pediatrics and Metabolism, University Children's Hospital, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jörg Dötsch
- Department of Pediatrics and Adolescent Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50924, Germany
| | - Eva Hucklenbruch-Rother
- Department of Pediatrics and Adolescent Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50924, Germany.
| |
Collapse
|
20
|
Ahmed S, Mahmood Z, Javed A, Hashmi SN, Zerr I, Zafar S, Zahid S. Effect of Metformin on Adult Hippocampal Neurogenesis: Comparison with Donepezil and Links to Cognition. J Mol Neurosci 2017; 62:88-98. [PMID: 28378260 DOI: 10.1007/s12031-017-0915-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 03/29/2017] [Indexed: 01/02/2023]
Abstract
Recent studies have uncovered evidence suggesting that interference with hippocampal adult neurogenesis contributes to neurodegeneration in Alzheimer's disease (AD). Evidence supporting that AD is a metabolic disease with derangements in brain glucose utilization implies the use of anti-diabetics as an alternate therapeutic strategy. The present study drew comparison between the pro-neurogenic potential of metformin and donepezil in AlCl3-induced mouse model of neurodegeneration. Morris water maze task and subsequent immunohistochemical evaluation for NeuN was conducted. Expression of neurogenesis markers and hippocampal proteome analysis was determined by qRT-PCR and SDS-PAGE, respectively, followed by ESI-QTOFF MS/MS identification. The results demonstrated impaired spatial memory and differential expression of eight proteins in the AlCl3 group as compared to the controls. Interestingly, treatment with metformin normalized the proteome profile and expression levels of neurogenesis markers along with improvement in the spatial memory. Moreover, as compared to donepezil, metformin-treated mice exhibited an enhanced number of post-mitotic NeuN-positive neurons. It is suggested that underlying molecular mechanisms of metformin-mediated adult hippocampal neurogenesis may have implications in treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Sara Ahmed
- Neurobiology Research Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Zahra Mahmood
- Neurobiology Research Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Aneela Javed
- Neurobiology Research Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Shoaib Naiyer Hashmi
- Department of Histopathology, Armed Forces Institute of Pathology, Rawalpindi, Pakistan
| | - Inga Zerr
- Department of Neurology, Clinical Dementia Center and DZNE, Georg-August University, University Medical Center Goettingen (UMG), Robert-Koch-Str. 40, 37075, Goettingen, Germany
| | - Saima Zafar
- Department of Neurology, Clinical Dementia Center and DZNE, Georg-August University, University Medical Center Goettingen (UMG), Robert-Koch-Str. 40, 37075, Goettingen, Germany
| | - Saadia Zahid
- Neurobiology Research Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan.
| |
Collapse
|
21
|
Chiang MC, Cheng YC, Chen SJ, Yen CH, Huang RN. Metformin activation of AMPK-dependent pathways is neuroprotective in human neural stem cells against Amyloid-beta-induced mitochondrial dysfunction. Exp Cell Res 2016; 347:322-31. [PMID: 27554603 DOI: 10.1016/j.yexcr.2016.08.013] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 08/15/2016] [Accepted: 08/17/2016] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is the general consequence of dementia and is diagnostic neuropathology by the cumulation of amyloid-beta (Aβ) protein aggregates, which are thought to promote mitochondrial dysfunction processes leading to neurodegeneration. AMP-activated protein kinase (AMPK), a critical regulator of energy homeostasis and a major player in lipid and glucose metabolism, is potentially implied in the mitochondrial deficiency of AD. Metformin, one of the widespread used anti- metabolic disease drugs, use its actions in part by stimulation of AMPK. While the mechanisms of AD are well established, the neuronal roles for AMPK in AD are still not well understood. In the present study, human neural stem cells (hNSCs) exposed to Aβ had significantly reduced cell viability, which correlated with decreased AMPK, neuroprotective genes (Bcl-2 and CREB) and mitochondria associated genes (PGC1α, NRF-1 and Tfam) expressions, as well as increased activation of caspase 3/9 activity and cytosolic cytochrome c. Co-treatment with metformin distinct abolished the Aβ-caused actions in hNSCs. Metformin also significantly rescued hNSCs from Aβ-mediated mitochondrial deficiency (lower D-loop level, mitochondrial mass, maximal respiratory function, COX activity, and mitochondrial membrane potential). Importantly, co-treatment with metformin significantly restored fragmented mitochondria to almost normal morphology in the hNSCs with Aβ. These findings extend our understanding of the central role of AMPK in Aβ-related neuronal impairment. Thus, a better understanding of AMPK might assist in both the recognition of its critical effects and the implementation of new therapeutic strategies in the treatment of AD.
Collapse
Affiliation(s)
- Ming-Chang Chiang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| | - Yi-Chuan Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shiang-Jiuun Chen
- Department of Life Science and Institute of Ecology and Evolutionary Biology, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Chia-Hui Yen
- Department of International Business, Ming Chuan University, Taipei 111, Taiwan
| | - Rong-Nan Huang
- Department of Entomology and Research Center for Plant-Medicine, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
22
|
Palleria C, Leporini C, Maida F, Succurro E, De Sarro G, Arturi F, Russo E. Potential effects of current drug therapies on cognitive impairment in patients with type 2 diabetes. Front Neuroendocrinol 2016; 42:76-92. [PMID: 27521218 DOI: 10.1016/j.yfrne.2016.07.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 07/13/2016] [Accepted: 07/22/2016] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes mellitus is a complex metabolic disease that can cause serious damage to various organs. Among the best-known complications, an important role is played by cognitive impairment. Impairment of cognitive functioning has been reported both in type 1 and 2 diabetes mellitus. While this comorbidity has long been known, no major advances have been achieved in clinical research; it is clear that appropriate control of blood glucose levels represents the best current (although unsatisfactory) approach in the prevention of cognitive impairment. We have focused our attention on the possible effect on the brain of antidiabetic drugs, despite their effects on blood glucose levels, giving a brief rationale on the mechanisms (e.g. GLP-1, BDNF, ghrelin) that might be involved. Indeed, GLP-1 agonists are currently clinically studied in other neurodegenerative diseases (i.e. Parkinson's and Alzheimer's disease); furthermore, also other antidiabetic drugs have proven efficacy in preclinical studies. Overall, promising results are already available and finding new intervention strategies represents a current need in this field of research.
Collapse
Affiliation(s)
- Caterina Palleria
- Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, Italy
| | - Christian Leporini
- Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, Italy
| | - Francesca Maida
- Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, Italy
| | - Elena Succurro
- Department of Medical and Surgical Sciences, Internal Medicine Unit of "Mater Domini", University Hospital, University "Magna Graecia" of Catanzaro, Policlinico "Mater Domini", Campus Universitario, Viale Europa, 88100 Catanzaro, Italy
| | - Giovambattista De Sarro
- Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, Italy
| | - Franco Arturi
- Department of Medical and Surgical Sciences, Internal Medicine Unit of "Mater Domini", University Hospital, University "Magna Graecia" of Catanzaro, Policlinico "Mater Domini", Campus Universitario, Viale Europa, 88100 Catanzaro, Italy
| | - Emilio Russo
- Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, Italy.
| |
Collapse
|
23
|
Sheen YJ, Sheu WHH. Association between hypoglycemia and dementia in patients with type 2 diabetes. Diabetes Res Clin Pract 2016; 116:279-87. [PMID: 27321346 DOI: 10.1016/j.diabres.2016.04.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 03/17/2016] [Accepted: 04/14/2016] [Indexed: 12/27/2022]
Abstract
In addition to increased risks of macrovascular and microvascular complications, patients with type 2 diabetes mellitus (T2DM) usually also are at increased risk for cognitive impairment and dementia. Hypoglycemia, a common consequence of diabetes treatment, is considered an independent risk factor for dementia in patients with T2DM. Hypoglycemia and dementia are clinically underestimated and are related to poor outcomes; thus, they may compromise the life expectancy of patients with T2DM. Epidemiological evidence of hypoglycemia-associated cognitive decline and dementia is highly varied. Acute, severe hypoglycemic episodes induce chronic subclinical brain damage, cognitive decline, and subsequent dementia. However, the effects of recurrent moderate hypoglycemia on cognitive decline and dementia remain largely uninvestigated. Poor glycemic control (including fluctuation of hemoglobin A1C [HbA1c] and glucose values) and the viscous circle of bidirectional associations between dementia and hypoglycemia may be clinically relevant. The possible pathophysiological hypotheses include post-hypoglycemic neuronal damage, inflammatory processes, coagulation defects, endothelial abnormalities, and synaptic dysfunction of hippocampal neurons during hypoglycemia episodes. This article reviews previous findings, provides insight into the detection of groups at high risk of hypoglycemia-associated dementia, and proposes specific strategies to minimize the potential burdens associated with hypoglycemia-related neurocognitive disorders in patients with T2DM.
Collapse
Affiliation(s)
- Yi-Jing Sheen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Hospital, Ministry of Health and Welfare, No. 199 Section 1, Sanmin Road, Taichung 403, Taiwan
| | - Wayne H H Sheu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, No. 1650, Section 4, Taiwan Boulevard, Taichung 407, Taiwan; School of Medicine, National Defense Medical Center, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Medical Technology, National Chung-Hsing University, Taichung, Taiwan.
| |
Collapse
|
24
|
Chen F, Dong RR, Zhong KL, Ghosh A, Tang SS, Long Y, Hu M, Miao MX, Liao JM, Sun HB, Kong LY, Hong H. Antidiabetic drugs restore abnormal transport of amyloid-β across the blood-brain barrier and memory impairment in db/db mice. Neuropharmacology 2015. [PMID: 26211973 DOI: 10.1016/j.neuropharm.2015.07.023] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Previous studies have shown significant changes in amyloid-β (Aβ) transport across the blood-brain barrier (BBB) under diabetic conditions with hypoinsulinemia, which is involved in diabetes-associated cognitive impairment. Present study employed db/db mice with hyperinsulinemia to investigate changes in Aβ transport across the BBB, hippocampal synaptic plasticity, and restorative effects of antidiabetic drugs. Our results showed that db/db mice exhibited similar changes in Aβ transport across the BBB to that of insulin-deficient mice. Chronic treatment of db/db mice with antidiabetic drugs such as metformin, glibenclamide and insulin glargine significantly decreased Aβ influx across the BBB determined by intra-arterial infusion of (125)I-Aβ(1-40), and expression of the receptor for advanced glycation end products (RAGE) participating in Aβ influx. Insulin glargine, but not, metformin or glibenclamide increased Aβ efflux across the BBB determined by stereotaxic intra-cerebral infusion of (125)I-Aβ(1-40), and expression of the low-density lipoprotein receptor related protein 1 (LRP1) participating in Aβ efflux. Moreover, treatment with these drugs significantly decreased hippocampal Aβ(1-40) or Aβ(1-42) and inhibited neuronal apoptosis. The drugs also ameliorated memory impairment confirmed by improved performance on behavioral tasks. However, insulin glargine or glibenclamide, but not metformin, restored hippocampal synaptic plasticity characterized by enhancing in vivo long-term potentiation (LTP). Further study found that these three drugs significantly restrained NF-κB, but only insulin glargine enhanced peroxisome proliferator-activated receptor γ (PPARγ) activity at the BBB in db/db mice. Our data indicate that the antidiabetic drugs can partially restore abnormal Aβ transport across the BBB and memory impairment under diabetic context.
Collapse
Affiliation(s)
- Fang Chen
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Rong Rong Dong
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Kai Long Zhong
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Arijit Ghosh
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Su Su Tang
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Yan Long
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Mei Hu
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Ming Xing Miao
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Jian Min Liao
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Hong Bing Sun
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Ling Yi Kong
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Hong
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
25
|
Rizvi SMD, Shaikh S, Waseem SMA, Shakil S, Abuzenadah AM, Biswas D, Tabrez S, Ashraf GM, Kamal MA. Role of anti-diabetic drugs as therapeutic agents in Alzheimer's disease. EXCLI JOURNAL 2015; 14:684-96. [PMID: 27152105 PMCID: PMC4849108 DOI: 10.17179/excli2015-252] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/05/2015] [Indexed: 12/16/2022]
Abstract
Recent data have suggested a strong possible link between Type 2 Diabetes Mellitus and Alzheimer's disease (AD), although exact mechanisms linking the two are still a matter of research and debate. Interestingly, both are diseases with high incidence and prevalence in later years of life. The link appears so strong that some scientists use Alzheimer's and Type 3 Diabetes interchangeably. In depth study of recent data suggests that the anti diabetic drugs not only have possible role in treatment of Alzheimer's but may also arrest the declining cognitive functions associated with it. The present review gives an insight into the possible links, existing therapeutics and clinical trials of anti diabetic drugs in patients suffering from AD primarily or as co-morbidity. It may be concluded that the possible beneficial effects and usefulness of the current anti diabetic drugs in AD cannot be neglected and further research is required to achieve positive results. Currently, several drug trials are in progress to give conclusive evidence based data.
Collapse
Affiliation(s)
| | | | - Shah Mohammad Abbas Waseem
- Department of Physiology, Integral Institute of Medical Sciences & Research, Integral University, Lucknow, India
| | - Shazi Shakil
- Center of Innovation in Personalized Medicine, Faculty of Applied Medical Sciences,King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adel M. Abuzenadah
- Center of Innovation in Personalized Medicine, Faculty of Applied Medical Sciences,King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md. Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Enzymoic, 7 Peterlee Pl, Hebersham, NSW 2770, Australia
| |
Collapse
|
26
|
Moreira PI. Metformin in the diabetic brain: friend or foe? ANNALS OF TRANSLATIONAL MEDICINE 2014; 2:54. [PMID: 25333029 DOI: 10.3978/j.issn.2305-5839.2014.06.10] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 06/16/2014] [Indexed: 12/20/2022]
Affiliation(s)
- Paula I Moreira
- 1 CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal ; 2 Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
27
|
Thioredoxin-mimetic peptide CB3 lowers MAPKinase activity in the Zucker rat brain. Redox Biol 2014; 2:447-56. [PMID: 24624334 PMCID: PMC3949098 DOI: 10.1016/j.redox.2013.12.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 12/20/2013] [Indexed: 02/07/2023] Open
Abstract
Diabetes is a high risk factor for dementia. High glucose may be a risk factor for dementia even among persons without diabetes, and in transgenic animals it has been shown to cause a potentiation of indices that are pre-symptomatic of Alzheimer's disease. To further elucidate the underlying mechanisms linking inflammatory events elicited in the brain during oxidative stress and diabetes, we monitored the activation of mitogen-activated kinsase (MAPKs), c-jun NH2-terminal kinase (JNK), p38 MAP kinases (p38MAPK), and extracellular activating kinsae1/2 (ERK1/2) and the anti-inflammatory effects of the thioredoxin mimetic (TxM) peptides, Ac-Cys-Pro-Cys-amide (CB3) and Ac-Cys-Gly-Pro-Cys-amide (CB4) in the brain of male leptin-receptor-deficient Zucker diabetic fatty (ZDF) rats and human neuroblastoma SH-SY5Y cells. Daily i.p. injection of CB3 to ZDF rats inhibited the phosphorylation of JNK and p38MAPK, and prevented the expression of thioredoxin-interacting-protein (TXNIP/TBP-2) in ZDF rat brain. Although plasma glucose/insulin remained high, CB3 also increased the phosphorylation of AMP-ribose activating kinase (AMPK) and inhibited p70S6K kinase in the brain. Both CB3 and CB4 reversed apoptosis induced by inhibiting thioredoxin reductase as monitored by decreasing caspase 3 cleavage and PARP dissociation in SH-SY5Y cells. The decrease in JNK and p38MAPK activity in the absence of a change in plasma glucose implies a decrease in oxidative or neuroinflammatory stress in the ZDF rat brain. CB3 not only attenuated MAPK phosphorylation and activated AMPK in the brain, but it also diminished apoptotic markers, most likely acting via the MAPK–AMPK–mTOR pathway. These results were correlated with CB3 and CB4 inhibiting inflammation progression and protection from oxidative stress induced apoptosis in human neuronal cells. We suggest that by attenuating neuro-inflammatory processes in the brain Trx1 mimetic peptides could become beneficial for preventing neurological disorders associated with diabetes. Thioredoxin mimeitics peptides (TXM) lower apoptosis in the brain of ZDF rat. TxM peptides prevent TXNIP/TBP-2 expression in the brain of ZDF rat. TxM peptides could become beneficial for preventing diabetes associated neurological disorders.
Collapse
|
28
|
Yoo DY, Kim W, Nam SM, Yoo KY, Lee CH, Choi JH, Won MH, Hwang IK, Yoon YS. Reduced cell proliferation and neuroblast differentiation in the dentate gyrus of high fat diet-fed mice are ameliorated by metformin and glimepiride treatment. Neurochem Res 2011; 36:2401-8. [PMID: 21818657 DOI: 10.1007/s11064-011-0566-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Revised: 07/26/2011] [Accepted: 07/27/2011] [Indexed: 12/12/2022]
Abstract
We investigated the effects of a high-fat diet (HFD) and the subsequent treatment of metformin (met) and glimepiride (glim), which are widely prescribed for type 2 diabetes, on cell proliferation and neuroblast differentiation using Ki67 and doublecortin (DCX) immunohistochemistry, respectively. Animals were fed low-fat diet (LFD) or HFD for 8 weeks. After 5 weeks of the HFD treatment, met alone or met + glim was administered orally once a day for 3 weeks. Body weight and food intake were much higher in the HFD + vehicle-treated group than the LFD-treated group. The administration of met or met + glim to the HFD-treated group resulted in a decrease in weight gain and food intake. Ki67-immunoreactive ((+)) nuclei, DCX(+) neuroblasts and brain-derived neurotrophic factor (BDNF) protein levels were markedly decreased in the dentate gyrus (DG) of the HFD + vehicle-treated group compared to the LFD-treated group. The administration of met or met + glim to the HFD-treated group prevented the reduction of Ki67(+) nuclei, DCX(+) neuroblasts, BDNF levels in the DG. The intraventricular injection of K252a (a BDNF receptor blocker) to the HFD-treated group treated met or met + glim distinctively lowered the reduction of cell proliferation and neuroblast differentiation induced by HFD. These results suggest that a HFD significantly reduces cell proliferation and neuroblast differentiation by reducing BDNF levels and these effects are ameliorated by treatment with met or met + glim.
Collapse
Affiliation(s)
- Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 151-742, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Rafalski VA, Brunet A. Energy metabolism in adult neural stem cell fate. Prog Neurobiol 2010; 93:182-203. [PMID: 21056618 DOI: 10.1016/j.pneurobio.2010.10.007] [Citation(s) in RCA: 218] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 10/20/2010] [Accepted: 10/28/2010] [Indexed: 12/26/2022]
Abstract
The adult mammalian brain contains a population of neural stem cells that can give rise to neurons, astrocytes, and oligodendrocytes and are thought to be involved in certain forms of memory, behavior, and brain injury repair. Neural stem cell properties, such as self-renewal and multipotency, are modulated by both cell-intrinsic and cell-extrinsic factors. Emerging evidence suggests that energy metabolism is an important regulator of neural stem cell function. Molecules and signaling pathways that sense and influence energy metabolism, including insulin/insulin-like growth factor I (IGF-1)-FoxO and insulin/IGF-1-mTOR signaling, AMP-activated protein kinase (AMPK), SIRT1, and hypoxia-inducible factors, are now implicated in neural stem cell biology. Furthermore, these signaling modules are likely to cooperate with other pathways involved in stem cell maintenance and differentiation. This review summarizes the current understanding of how cellular and systemic energy metabolism regulate neural stem cell fate. The known consequences of dietary restriction, exercise, aging, and pathologies with deregulated energy metabolism for neural stem cells and their differentiated progeny will also be discussed. A better understanding of how neural stem cells are influenced by changes in energy availability will help unravel the complex nature of neural stem cell biology in both the normal and diseased state.
Collapse
|