1
|
Pazarlar BA, Egilmez CB, Oyar EÖ, Mikkelsen JD. Increased Expression of Synaptic Vesicle Glycoprotein 2A (SV2A) in the Brain of Chronic Diabetic Rats. Synapse 2025; 79:e70018. [PMID: 40317510 PMCID: PMC12048859 DOI: 10.1002/syn.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/07/2025]
Abstract
AIM/HYPOTHESIS Diabetes mellitus has been reported to be a risk factor for cognitive dysfunction, depression, stroke, and seizures. Diabetic pathology is believed to interfere with synaptic plasticity. Synaptic vesicle glycoprotein 2A (SV2A) is a presynaptic vesicular protein and a popular synaptic density imaging marker. We investigated the effect of chronic hyperglycemia on the expression of SV2A in the cerebral cortex and hippocampus of rats and compared it to other presynaptic markers, such as GAP43, Synaptotagmin-1, and SNAP25. METHODS A single dose of streptozotocin (STZ, 45 mg/kg, i.p.) was administered to adult male rats, resulting in sustained hyperglycemia and reduced plasma insulin levels. Controls were injected with saline, and another STZ group was treated with insulin. Fasting blood glucose (FBG) and fasting plasma insulin (FPI) levels were monitored throughout the observation period, and the level of SV2A was determined by radioligand, [3H]UCB-J, binding capacity using in-vitro autoradiography and by ELISA. Similarly, the tissue concentration of other synaptic proteins GAP43, SNAP25, and SYN1 was measured using ELISA. Quantitative RT-qPCR was performed to measure Sv2a, Sv2b, and Sv2c transcripts. Finally, hippocampal and cortical glutamate levels were measured in all tissues. RESULTS [3H]UCB-J binding, SV2A (pg/mg protein) and Sv2a mRNA levels were significantly higher in hyperglycemic rats. The SV2A concentration detected by ELISA and [3H]UCB-J binding showed, as expected, a positive correlation with each other. The same positive and significant correlation was seen between SV2A, FBG, and glutamate l levels across animals (p ≤ 0.001). Notably, there was no difference and no linearity between FBG and other presynaptic markers such as GAP43, Synaptotagmin-1, and SNAP25. CONCLUSIONS Unlike other synaptic markers (e.g., SNAP25, SYN-1), SV2A levels rise independently of synaptic density, correlating with elevated glutamate and metabolic activity. These findings raise doubt about SV2A's role as a pure synaptic density marker.
Collapse
Affiliation(s)
- Burcu Azak Pazarlar
- Department of NeuroscienceUniversity of CopenhagenCopenhagenDenmark
- Neurobiology Research UnitUniversity Hospital Copenhagen, RigshospitaletCopenhagenDenmark
- Physiology Department, Faculty of MedicineIzmir Katip Celebi UniversityIzmirTurkey
| | | | - Eser Öz Oyar
- Physiology Department, Faculty of MedicineIzmir Katip Celebi UniversityIzmirTurkey
| | - Jens D. Mikkelsen
- Department of NeuroscienceUniversity of CopenhagenCopenhagenDenmark
- Neurobiology Research UnitUniversity Hospital Copenhagen, RigshospitaletCopenhagenDenmark
| |
Collapse
|
2
|
Zhou J, Sun X, Wang K, Shen M, Yu J, Yao Q, Hong H, Tang C, Wang Q. What Information do Systemic Pathological Changes Bring to the Diagnosis and Treatment of Alzheimer's Disease? Neurosci Bull 2025:10.1007/s12264-025-01399-z. [PMID: 40257662 DOI: 10.1007/s12264-025-01399-z] [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: 08/21/2024] [Accepted: 02/21/2025] [Indexed: 04/22/2025] Open
Abstract
Alzheimer's disease (AD) is regarded as a neurodegenerative disease, and it has been proposed that AD may be a systemic disease. Studies have reported associations between non-neurological diseases and AD. The correlations between AD pathology and systemic (non-neurological) pathological changes are intricate, and the mechanisms underlying these correlations and their causality are unclear. In this article, we review the association between AD and disorders of other systems. In addition, we summarize the possible mechanisms associated with AD and disorders of other systems, mainly from the perspective of AD pathology. Regarding the relationship between AD and systemic pathological changes, we aim to provide a new outlook on the early warning signs and treatment of AD, such as establishing a diagnostic and screening system based on more accessible peripheral samples.
Collapse
Affiliation(s)
- Jinyue Zhou
- Health Science Center, The First Affiliated Hospital, Ningbo University, Ningbo, 315010, China
| | - Xiaoli Sun
- Department of Chemistry, Lishui University, Lishui, 32300, China
| | - Keren Wang
- Health Science Center, School of Public Health, Ningbo University, Ningbo, 315211, China
| | - Min Shen
- Reference Laboratory, Medical System Biotechnology Co., Ltd, Ningbo, 315104, China
| | - Jingbo Yu
- Health Science Center, The First Affiliated Hospital, Ningbo University, Ningbo, 315010, China
| | - Qi Yao
- Health Science Center, The First Affiliated Hospital, Ningbo University, Ningbo, 315010, China
| | - Hang Hong
- Health Science Center, School of Public Health, Ningbo University, Ningbo, 315211, China.
| | - Chunlan Tang
- Health Science Center, School of Public Health, Ningbo University, Ningbo, 315211, China.
| | - Qinwen Wang
- Health Science Center, The First Affiliated Hospital, Ningbo University, Ningbo, 315010, China.
| |
Collapse
|
3
|
Kullmann S, Wagner L, Hauffe R, Kühnel A, Sandforth L, Veit R, Dannecker C, Machann J, Fritsche A, Stefan N, Preissl H, Kroemer NB, Heni M, Kleinridders A, Birkenfeld AL. A short-term, high-caloric diet has prolonged effects on brain insulin action in men. Nat Metab 2025; 7:469-477. [PMID: 39984682 PMCID: PMC11946887 DOI: 10.1038/s42255-025-01226-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 01/30/2025] [Indexed: 02/23/2025]
Abstract
Brain insulin responsiveness is linked to long-term weight gain and unhealthy body fat distribution. Here we show that short-term overeating with calorie-rich sweet and fatty foods triggers liver fat accumulation and disrupted brain insulin action that outlasted the time-frame of its consumption in healthy weight men. Hence, brain response to insulin can adapt to short-term changes in diet before weight gain and may facilitate the development of obesity and associated diseases.
Collapse
Affiliation(s)
- Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany.
| | - Lore Wagner
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Robert Hauffe
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Nutritional Science, Department of Molecular and Experimental Nutritional Medicine, University of Potsdam, Nuthetal, Germany
- German Institute of Human Nutrition, Junior Research Group Central Regulation of Metabolism, Nuthetal, Germany
| | - Anne Kühnel
- Section of Medical Psychology, Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Leontine Sandforth
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Ralf Veit
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Corinna Dannecker
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jürgen Machann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Section on Experimental Radiology, Department of Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Nobert Stefan
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Hubert Preissl
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Nils B Kroemer
- Section of Medical Psychology, Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Bonn, Bonn, Germany
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany
| | - Martin Heni
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - André Kleinridders
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Nutritional Science, Department of Molecular and Experimental Nutritional Medicine, University of Potsdam, Nuthetal, Germany
- German Institute of Human Nutrition, Junior Research Group Central Regulation of Metabolism, Nuthetal, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
4
|
Zhang X, Huang R, Li J, Yang M, Zhang D, Liu C, Fan K. Abnormal insulin metabolism and decreased levels of mindfulness in type 2 diabetes mellitus. Diabetol Metab Syndr 2025; 17:32. [PMID: 39849632 PMCID: PMC11755820 DOI: 10.1186/s13098-025-01594-y] [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: 11/04/2024] [Accepted: 01/13/2025] [Indexed: 01/25/2025] Open
Abstract
OBJECTIVE Disorders of insulin metabolism are strongly associated with a variety of psychological problems. The aim of this study was to investigate the differences in mindfulness levels among type 2 diabetes mellitus (T2DM) patients categorized based on their insulin resistance and β-cell function. METHODS A total of 157 T2DM patients were included in this study and divided into four groups according to their levels of insulin resistance and β-cell function. The Five Facet Mindfulness Questionnaire (FFMQ) was employed to assess the mindfulness levels of the patients. Linear regression models were utilized to investigate the relationships between various T2DM categories and mindfulness levels and dimensions. Furthermore, subgroup analyses of key variables were conducted, and mediation analysis was performed to evaluate the sources of differences. RESULTS Significant differences were observed among the four groups in terms of total mindfulness scores and in the dimensions of "Describing," "Non-judging of Inner Experience," and "Acting with Awareness" (P < 0.05). Compared to the control group (low HOMA-IR/high HOMA-β), the high HOMA-IR/low HOMA-β group exhibited markedly lower scores in "Non-judging of Inner Experience" (P = 0.02) and "Acting with Awareness" (P < 0.001). The low HOMA-IR/low HOMA-β group demonstrated weaker performance in "Non-judging of Inner Experience" (P = 0.005) and "Describing" (P = 0.002). CONCLUSION Significant differences in mindfulness levels were found to exist among T2DM patients with varying degrees of insulin resistance and β-cell function. Early-stage diabetes patients, particularly those with lower β-cell function or higher insulin resistance levels, may require additional psychological intervention support to enhance their mindfulness and overall well-being.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Rui Huang
- Department of Endocrinology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiaxin Li
- Department of Endocrinology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Mingyue Yang
- Department of Endocrinology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Daowen Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Cancan Liu
- Department of Endocrinology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kuanlu Fan
- Department of Endocrinology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
5
|
Padhy DS, Aggarwal P, Velayutham R, Banerjee S. Aerobic exercise and metformin attenuate the cognitive impairment in an experimental model of type 2 diabetes mellitus: focus on neuroinflammation and adult hippocampal neurogenesis. Metab Brain Dis 2025; 40:92. [PMID: 39775196 DOI: 10.1007/s11011-024-01489-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder that increases the prevalence of cognitive impairment in the geriatric population. Aerobic exercise is an excellent non-pharmacological therapeutic strategy to prevent Alzheimer's disease, the most common form of dementia. The exact molecular mechanism of aerobic exercise (Exe) as an intervention to counter cognitive decline is far from clear. Metformin is a first-line agent against T2DM with neuroprotective properties. The present study assessed the role of treadmill exercise in combination with a low dose of metformin (Met; 70 mg/kg) in cognitive impairment and its associated molecular mechanism in T2DM rats. The experimental model of T2DM-associated cognitive decline was created by administration of a high-fat diet (HFD) with a low dose of streptozotocin (STZ; 35 mg/kg). Neurobehavioral assessments were performed to evaluate spatial recognition and fear-conditioned memory across the groups: control, HFD + STZ, HFD + STZ + Exe, and HFD + STZ + Exe + Met. In addition, we performed immunohistochemistry and western blotting on the rat hippocampal tissue from the above groups for protein expression studies. T2DM rats showed a significant cognitive decline compared to the control group, which improved in the long-term exercise and metformin co-administered animals. The level of neuroinflammation was significantly elevated in the hippocampal tissue of T2DM rats compared to the control and lowered after exercise and metformin treatment. T2DM reduced mature neurons and neurogenesis while increasing astrogliosis and microgliosis, ameliorated by exercise and metformin treatment. Moreover, T2DM impaired hippocampal neurogenesis by reducing the canonical Wnt/β-catenin pathway, which got upregulated in exercise and metformin-co-administered rats. Long-term aerobic exercise with metformin treatment ameliorated neuroinflammation and promoted adult hippocampal neurogenesis via upregulating the canonical Wnt/β-catenin pathway in T2DM rats.
Collapse
Affiliation(s)
- Dibya Sundar Padhy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Kolkata, Kolkata, West Bengal, 700054, India
| | - Punita Aggarwal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Kolkata, Kolkata, West Bengal, 700054, India
| | - Ravichandiran Velayutham
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER)- Kolkata, Kolkata, West Bengal, 700054, India.
| | - Sugato Banerjee
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Kolkata, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
6
|
Guo T, Zou Q, Wang Q, Zhang Y, Zhong X, Lin H, Gong W, Wang Y, Xie K, Wu K, Chen F, Chen W. Association of TyG Index and TG/HDL-C Ratio with Trajectories of Depressive Symptoms: Evidence from China Health and Retirement Longitudinal Study. Nutrients 2024; 16:4300. [PMID: 39770920 PMCID: PMC11676214 DOI: 10.3390/nu16244300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVES To explore whether the triglyceride-glucose (TyG) index and the triglyceride to high-density lipoprotein cholesterol (TG/HDL-C) ratio are associated with the trajectories of depressive symptoms. METHODS In this longitudinal study, 4215 participants aged 45 years and older were recruited from the China Health and Retirement Longitudinal Study from 2011 to 2018. The trajectories of depressive symptoms, measured by the 10-item Center for Epidemiologic Studies Depression Scale (CESD-10), were identified using group-based trajectory modeling. Multinomial logistic models and restricted cubic spline analysis were used to investigate the relationships between the TyG index and the TG/HDL-C ratio and the trajectories of depressive symptoms. Stratified analyses were conducted based on sex, age, place of residence, and body mass index (BMI). RESULTS Five distinct trajectories of depressive symptoms characterized by stable low, stable moderate, decreasing, increasing, and stable high were identified during a follow-up of 7 years. The associations of the TyG index and the TG/HDL-C ratio with trajectories of depressive symptoms are not entirely consistent. After adjusting for covariates, a higher TyG index at baseline was associated with lower odds of being on the decreasing trajectory of depressive symptoms (ORad = 0.61, 95% CI: 0.40-0.92) compared to the stable low trajectory, and restricted cubic spline analysis revealed a negative linear relationship between the TyG index and the likelihood of a decreasing trajectory of depressive symptoms. However, the relationship between the TG/HDL-C ratio and the decreasing trajectory of depressive symptoms was no longer statistically significant when all confounders were controlled (ORad = 0.72, 95% CI: 0.50-1.04). Additionally, this negative association between the TyG index and decreasing trajectory of depressive symptoms was observed among 45-64-year-old individuals, female participants, those living in rural areas, and those with a normal BMI. LIMITATIONS This study was conducted in a middle-aged and elderly population in China, and extrapolation to other regions and populations requires further confirmation. CONCLUSIONS Compared to the TG/HDL-C ratio, the TyG index may be a better predictor for trajectories of depressive symptoms in middle-aged and older adults. Considering that the pathology of depression progresses long term, our findings may have utility for identifying available and reliable markers for the development of depression.
Collapse
Affiliation(s)
- Tingting Guo
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, 74 Zhongshan Second Rd, Guangzhou 510080, China; (T.G.); (Q.Z.); (Q.W.); (Y.Z.); (X.Z.); (H.L.); (W.G.); (Y.W.); (K.X.); (K.W.)
| | - Qing Zou
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, 74 Zhongshan Second Rd, Guangzhou 510080, China; (T.G.); (Q.Z.); (Q.W.); (Y.Z.); (X.Z.); (H.L.); (W.G.); (Y.W.); (K.X.); (K.W.)
| | - Qi Wang
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, 74 Zhongshan Second Rd, Guangzhou 510080, China; (T.G.); (Q.Z.); (Q.W.); (Y.Z.); (X.Z.); (H.L.); (W.G.); (Y.W.); (K.X.); (K.W.)
| | - Yi Zhang
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, 74 Zhongshan Second Rd, Guangzhou 510080, China; (T.G.); (Q.Z.); (Q.W.); (Y.Z.); (X.Z.); (H.L.); (W.G.); (Y.W.); (K.X.); (K.W.)
| | - Xinyuan Zhong
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, 74 Zhongshan Second Rd, Guangzhou 510080, China; (T.G.); (Q.Z.); (Q.W.); (Y.Z.); (X.Z.); (H.L.); (W.G.); (Y.W.); (K.X.); (K.W.)
| | - Hantong Lin
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, 74 Zhongshan Second Rd, Guangzhou 510080, China; (T.G.); (Q.Z.); (Q.W.); (Y.Z.); (X.Z.); (H.L.); (W.G.); (Y.W.); (K.X.); (K.W.)
| | - Wenxuan Gong
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, 74 Zhongshan Second Rd, Guangzhou 510080, China; (T.G.); (Q.Z.); (Q.W.); (Y.Z.); (X.Z.); (H.L.); (W.G.); (Y.W.); (K.X.); (K.W.)
| | - Yingbo Wang
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, 74 Zhongshan Second Rd, Guangzhou 510080, China; (T.G.); (Q.Z.); (Q.W.); (Y.Z.); (X.Z.); (H.L.); (W.G.); (Y.W.); (K.X.); (K.W.)
| | - Kun Xie
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, 74 Zhongshan Second Rd, Guangzhou 510080, China; (T.G.); (Q.Z.); (Q.W.); (Y.Z.); (X.Z.); (H.L.); (W.G.); (Y.W.); (K.X.); (K.W.)
| | - Kunpeng Wu
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, 74 Zhongshan Second Rd, Guangzhou 510080, China; (T.G.); (Q.Z.); (Q.W.); (Y.Z.); (X.Z.); (H.L.); (W.G.); (Y.W.); (K.X.); (K.W.)
| | - Feng Chen
- Department of Clinical Research, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025 Shennan Zhong Rd, Shenzhen 518033, China;
| | - Wen Chen
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, 74 Zhongshan Second Rd, Guangzhou 510080, China; (T.G.); (Q.Z.); (Q.W.); (Y.Z.); (X.Z.); (H.L.); (W.G.); (Y.W.); (K.X.); (K.W.)
- Center for Migrant Health Policy, Sun Yat-sen University, 74 Zhongshan Second Rd, Guangzhou 510080, China
| |
Collapse
|
7
|
Sanz-Martos AB, Roca M, Plaza A, Merino B, Ruiz-Gayo M, Olmo ND. Long-term saturated fat-enriched diets impair hippocampal learning and memory processes in a sex-dependent manner. Neuropharmacology 2024; 259:110108. [PMID: 39128582 DOI: 10.1016/j.neuropharm.2024.110108] [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: 05/28/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Consumption of saturated fat-enriched diets during adolescence has been closely associated with the reduction of hippocampal synaptic plasticity and the impairment of cognitive function. Nevertheless, the effect of long-term intake of these foods has not yet been studied. In the present study, we have investigated the effect of a treatment, lasting for 40 weeks, with a diet enriched in saturated fat (SOLF) on i) spatial learning and memory, ii) hippocampal synaptic transmission and plasticity, and iii) hippocampal gene expression levels in aged male and female mice. Our findings reveal that SOLF has a detrimental impact on spatial memory and synaptic plasticity mechanisms, such as long-term potentiation (LTP), and downregulates Gria1 expression specifically in males. In females, SOLF downregulates the gene expression of Gria1/2/3 and Grin1/2A/2B glutamate receptor subunits as well as some proinflammatory interleukins. These findings highlight the importance of considering sex-specific factors when assessing the long-term effects of high-fat diets on cognition and brain plasticity.
Collapse
Affiliation(s)
- Ana Belén Sanz-Martos
- Department of Psychobiology, School of Psychology, UNED, C/ Juan del Rosal 10, 28040, Madrid, Spain.
| | - María Roca
- Department of Psychobiology, School of Psychology, UNED, C/ Juan del Rosal 10, 28040, Madrid, Spain
| | - Adrián Plaza
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad CEU-San Pablo, CEU Universities, 28668, Madrid, Spain
| | - Beatriz Merino
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad CEU-San Pablo, CEU Universities, 28668, Madrid, Spain
| | - Mariano Ruiz-Gayo
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad CEU-San Pablo, CEU Universities, 28668, Madrid, Spain
| | - Nuria Del Olmo
- Department of Psychobiology, School of Psychology, UNED, C/ Juan del Rosal 10, 28040, Madrid, Spain
| |
Collapse
|
8
|
Selenius JS, Silveira PP, von Bonsdorff M, Lahti J, Koistinen H, Koistinen R, Seppälä M, Eriksson JG, Wasenius NS. Biologically Informed Polygenic Scores for Brain Insulin Receptor Network Are Associated with Cardiometabolic Risk Markers and Diabetes in Women. Diabetes Metab J 2024; 48:960-970. [PMID: 38527457 PMCID: PMC11449818 DOI: 10.4093/dmj.2023.0039] [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: 02/10/2023] [Accepted: 11/25/2023] [Indexed: 03/27/2024] Open
Abstract
BACKGRUOUND To investigate associations between variations in the co-expression-based brain insulin receptor polygenic score and cardiometabolic risk factors and diabetes mellitus. METHODS This cross-sectional study included 1,573 participants from the Helsinki Birth Cohort Study. Biologically informed expression-based polygenic risk scores for the insulin receptor gene network were calculated for the hippocampal (hePRS-IR) and the mesocorticolimbic (mePRS-IR) regions. Cardiometabolic markers included body composition, waist circumference, circulating lipids, insulin-like growth factor 1 (IGF-1), and insulin-like growth factor-binding protein 1 and 3 (IGFBP-1 and -3). Glucose and insulin levels were measured during a standardized 2-hour 75 g oral glucose tolerance test and impaired glucose regulation status was defined by the World Health Organization 2019 criteria. Analyzes were adjusted for population stratification, age, smoking, alcohol consumption, socioeconomic status, chronic diseases, birth weight, and leisure-time physical activity. RESULTS Multinomial logistic regression indicated that one standard deviation increase in hePRS-IR was associated with increased risk of diabetes mellitus in all participants (adjusted relative risk ratio, 1.17; 95% confidence interval, 1.01 to 1.35). In women, higher hePRS-IR was associated with greater waist circumference and higher body fat percentage, levels of glucose, insulin, total cholesterol, low-density lipoprotein cholesterol, triglycerides, apolipoprotein B, insulin, and IGFBP-1 (all P≤0.02). The mePRS-IR was associated with decreased IGF-1 level in women (P=0.02). No associations were detected in men and studied outcomes. CONCLUSION hePRS-IR is associated with sex-specific differences in cardiometabolic risk factor profiles including impaired glucose regulation, abnormal metabolic markers, and unfavorable body composition in women.
Collapse
Affiliation(s)
- Jannica S. Selenius
- Folkhälsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Patricia P. Silveira
- Department of Psychiatry, Faculty of Medicine, McGill University, Verdun, QC, Canada
- Ludmer Center for Neuroinformatic and Mental Health, Douglas Mental Health University Institute, McGill University, Verdun, QC, Canada
| | - Mikaela von Bonsdorff
- Folkhälsan Research Center, Helsinki, Finland
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Jari Lahti
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
- Turku Institute for Advanced Studies, University of Turku, Turku, Finland
| | - Hannu Koistinen
- Department of Clinical Chemistry and Haematology, Helsinki University Hospital, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riitta Koistinen
- Department of Clinical Chemistry and Haematology, Helsinki University Hospital, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Markku Seppälä
- Department of Clinical Chemistry and Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Johan G. Eriksson
- Folkhälsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Department of Obstetrics & Gynecology and Human Potential Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Niko S. Wasenius
- Folkhälsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| |
Collapse
|
9
|
Rhea EM, Leclerc M, Yassine HN, Capuano AW, Tong H, Petyuk VA, Macauley SL, Fioramonti X, Carmichael O, Calon F, Arvanitakis Z. State of the Science on Brain Insulin Resistance and Cognitive Decline Due to Alzheimer's Disease. Aging Dis 2024; 15:1688-1725. [PMID: 37611907 PMCID: PMC11272209 DOI: 10.14336/ad.2023.0814] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/14/2023] [Indexed: 08/25/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is common and increasing in prevalence worldwide, with devastating public health consequences. While peripheral insulin resistance is a key feature of most forms of T2DM and has been investigated for over a century, research on brain insulin resistance (BIR) has more recently been developed, including in the context of T2DM and non-diabetes states. Recent data support the presence of BIR in the aging brain, even in non-diabetes states, and found that BIR may be a feature in Alzheimer's disease (AD) and contributes to cognitive impairment. Further, therapies used to treat T2DM are now being investigated in the context of AD treatment and prevention, including insulin. In this review, we offer a definition of BIR, and present evidence for BIR in AD; we discuss the expression, function, and activation of the insulin receptor (INSR) in the brain; how BIR could develop; tools to study BIR; how BIR correlates with current AD hallmarks; and regional/cellular involvement of BIR. We close with a discussion on resilience to both BIR and AD, how current tools can be improved to better understand BIR, and future avenues for research. Overall, this review and position paper highlights BIR as a plausible therapeutic target for the prevention of cognitive decline and dementia due to AD.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA.
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Manon Leclerc
- Faculty of Pharmacy, Laval University, Quebec, Quebec, Canada.
- Neuroscience Axis, CHU de Québec Research Center - Laval University, Quebec, Quebec, Canada.
| | - Hussein N Yassine
- Departments of Neurology and Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Ana W Capuano
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Han Tong
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA.
| | - Shannon L Macauley
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA.
| | - Xavier Fioramonti
- International Associated Laboratory OptiNutriBrain, Bordeaux, France and Quebec, Canada.
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France.
| | - Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | - Frederic Calon
- Faculty of Pharmacy, Laval University, Quebec, Quebec, Canada.
- Neuroscience Axis, CHU de Québec Research Center - Laval University, Quebec, Quebec, Canada.
- International Associated Laboratory OptiNutriBrain, Bordeaux, France and Quebec, Canada.
| | - Zoe Arvanitakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
10
|
Cai M, Wan J, Cai K, Li S, Du X, Song H, Sun W, Hu J. The mitochondrial quality control system: a new target for exercise therapeutic intervention in the treatment of brain insulin resistance-induced neurodegeneration in obesity. Int J Obes (Lond) 2024; 48:749-763. [PMID: 38379083 DOI: 10.1038/s41366-024-01490-x] [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] [Received: 04/04/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/22/2024]
Abstract
Obesity is a major global health concern because of its strong association with metabolic and neurodegenerative diseases such as diabetes, dementia, and Alzheimer's disease. Unfortunately, brain insulin resistance in obesity is likely to lead to neuroplasticity deficits. Since the evidence shows that insulin resistance in brain regions abundant in insulin receptors significantly alters mitochondrial efficiency and function, strategies targeting the mitochondrial quality control system may be of therapeutic and practical value in obesity-induced cognitive decline. Exercise is considered as a powerful stimulant of mitochondria that improves insulin sensitivity and enhances neuroplasticity. It has great potential as a non-pharmacological intervention against the onset and progression of obesity associated neurodegeneration. Here, we integrate the current knowledge of the mechanisms of neurodegenration in obesity and focus on brain insulin resistance to explain the relationship between the impairment of neuronal plasticity and mitochondrial dysfunction. This knowledge was synthesised to explore the exercise paradigm as a feasible intervention for obese neurodegenration in terms of improving brain insulin signals and regulating the mitochondrial quality control system.
Collapse
Affiliation(s)
- Ming Cai
- Jinshan District Central Hospital affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201599, China
| | - Jian Wan
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China
| | - Keren Cai
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Shuyao Li
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Xinlin Du
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Haihan Song
- Central Lab, Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China
| | - Wanju Sun
- Central Lab, Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China.
| | - Jingyun Hu
- Central Lab, Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China.
| |
Collapse
|
11
|
Baer SB, Dorn AD, Osborne DM. Sex differences in response to obesity and caloric restriction on cognition and hippocampal measures of autophagic-lysosomal transcripts and signaling pathways. BMC Neurosci 2024; 25:1. [PMID: 38166559 PMCID: PMC10759648 DOI: 10.1186/s12868-023-00840-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 12/18/2023] [Indexed: 01/04/2024] Open
Abstract
BACKGROUND Obesity rates in the U.S. continue to increase, with nearly 50% of the population being either obese or morbidly obese. Obesity, along with female sex, are leading risk factors for sporadic Alzheimer's Disease (AD) necessitating the need to better understand how these variables impact cellular function independent of age or genetic mutations. Animal and clinical studies both indicate that autophagy-lysosomal pathway (ALP) dysfunction is among the earliest known cellular systems to become perturbed in AD, preceding cognitive decline, yet little is known about how obesity and sex affects these cellular functions in the hippocampus, a brain region uniquely susceptible to the negative effects of obesity. We hypothesized that obesity would negatively affect key markers of ALP in the hippocampus, effects would vary based on sex, and that caloric restriction would counteract obesity effects. METHODS Female and male mice were placed on an obesogenic diet for 10 months, at which point half were switched to caloric restriction for three months, followed by cognitive testing in the Morris watermaze. Hippocampus was analyzed by western blot and qPCR. RESULTS Cognitive function in female mice responded differently to caloric restriction based on whether they were on a normal or obesogenic diet; male cognition was only mildly affected by caloric restriction and not obesity. Significant male-specific changes occurred in cellular markers of autophagy, including obesity increasing pAkt, Slc38a9, and Atg12, while caloric restriction reduced pRPS6 and increased Atg7. In contrast females experienced changes due to diet/caloric restriction predominately in lysosomal markers including increased TFE3, FLCN, FNIP2, and pAMPK. CONCLUSIONS Results support that hippocampal ALP is a target of obesity and that sex shapes molecular responses, while providing insight into how dietary manipulations affect learning and memory based on sex.
Collapse
Affiliation(s)
- Sadie B Baer
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR, USA
| | - Adrianah D Dorn
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR, USA
| | | |
Collapse
|
12
|
Spoelder M, Bright Y, Morrison MC, van Kempen V, de Groodt L, Begalli M, Schuijt N, Kruiger E, Bulthuis R, Gross G, Kleemann R, van Diepen JA, Homberg JR. Cognitive Performance during the Development of Diabetes in the Zucker Diabetic Fatty Rat. Cells 2023; 12:2463. [PMID: 37887307 PMCID: PMC10605915 DOI: 10.3390/cells12202463] [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: 08/25/2023] [Revised: 09/30/2023] [Accepted: 10/05/2023] [Indexed: 10/28/2023] Open
Abstract
Increased insulin levels may support the development of neural circuits involved in cognition, while chronic mild inflammation may also result in cognitive impairment. This study aimed to gain more insight into whether cognition is already impacted during adolescence in a genetic rat model for obesity and type 2 diabetes. Visual discrimination learning throughout adolescence and the level of motivation during early adulthood were investigated in Zucker Diabetic Fatty (ZDF) obese and ZDF lean rats using operant touchscreens. Blood glucose, insulin, and lipids were longitudinally analyzed. Histological analyses were performed in the liver, white adipose tissues, and the prefrontal cortex. Prior to the experiments with the genetic ZDF research model, all experimental assays were performed in two groups of outbred Long Evans rats to investigate the effect of different feeding circumstances. Adolescent ZDF obese rats outperformed ZDF lean rats on visual discrimination performance. During the longitudinal cognitive testing period, insulin levels sharply increased over weeks in ZDF obese rats and were significantly enhanced from 6 weeks of age onwards. Early signs of liver steatosis and enlarged adipocytes in white adipose tissue were observed in early adult ZDF obese rats. Histological analyses in early adulthood showed no group differences in the number of prefrontal cortex neurons and microglia, nor PSD95 and SIRT1 mRNA expression levels. Together, our data show that adolescent ZDF obese rats even display enhanced cognition despite their early diabetic profile.
Collapse
Affiliation(s)
- Marcia Spoelder
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Yami Bright
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Martine C. Morrison
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 CE Leiden, The Netherlands
| | - Veerle van Kempen
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Lilian de Groodt
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Malvina Begalli
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Nikita Schuijt
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Eva Kruiger
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| | - Ronald Bulthuis
- Metris B.V., Kruisweg 829c, 2132 NG Hoofddorp, The Netherlands
| | - Gabriele Gross
- Medical and Scientific Affairs, Reckitt|Mead Johnson Nutrition Institute, Middenkampweg 2, 6545 CJ Nijmegen, The Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 CE Leiden, The Netherlands
| | - Janna A. van Diepen
- Medical and Scientific Affairs, Reckitt|Mead Johnson Nutrition Institute, Middenkampweg 2, 6545 CJ Nijmegen, The Netherlands
| | - Judith R. Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (Y.B.)
| |
Collapse
|
13
|
Raut S, Bhalerao A, Powers M, Gonzalez M, Mancuso S, Cucullo L. Hypometabolism, Alzheimer's Disease, and Possible Therapeutic Targets: An Overview. Cells 2023; 12:2019. [PMID: 37626828 PMCID: PMC10453773 DOI: 10.3390/cells12162019] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/19/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
The brain is a highly dynamic organ that requires a constant energy source to function normally. This energy is mostly supplied by glucose, a simple sugar that serves as the brain's principal fuel source. Glucose transport across the blood-brain barrier (BBB) is primarily controlled via sodium-independent facilitated glucose transport, such as by glucose transporter 1 (GLUT1) and 3 (GLUT3). However, other glucose transporters, including GLUT4 and the sodium-dependent transporters SGLT1 and SGLT6, have been reported in vitro and in vivo. When the BBB endothelial layer is crossed, neurons and astrocytes can absorb the glucose using their GLUT1 and GLUT3 transporters. Glucose then enters the glycolytic pathway and is metabolized into adenosine triphosphate (ATP), which supplies the energy to support cellular functions. The transport and metabolism of glucose in the brain are impacted by several medical conditions, which can cause neurological and neuropsychiatric symptoms. Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, traumatic brain injury (TBI), schizophrenia, etc., are a few of the most prevalent disorders, characterized by a decline in brain metabolism or hypometabolism early in the course of the disease. Indeed, AD is considered a metabolic disorder related to decreased brain glucose metabolism, involving brain insulin resistance and age-dependent mitochondrial dysfunction. Although the conventional view is that reduced cerebral metabolism is an effect of neuronal loss and consequent brain atrophy, a growing body of evidence points to the opposite, where hypometabolism is prodromal or at least precedes the onset of brain atrophy and the manifestation of clinical symptoms. The underlying processes responsible for these glucose transport and metabolic abnormalities are complicated and remain poorly understood. This review article provides a comprehensive overview of the current understanding of hypometabolism in AD and potential therapeutic targets.
Collapse
Affiliation(s)
- Snehal Raut
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Aditya Bhalerao
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Michael Powers
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA;
| | - Minelly Gonzalez
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Salvatore Mancuso
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| |
Collapse
|
14
|
Gulyaeva NV. Glucocorticoids Orchestrate Adult Hippocampal Plasticity: Growth Points and Translational Aspects. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:565-589. [PMID: 37331704 DOI: 10.1134/s0006297923050012] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/10/2023] [Accepted: 04/10/2023] [Indexed: 06/20/2023]
Abstract
The review analyzes modern concepts about the control of various mechanisms of the hippocampal neuroplasticity in adult mammals and humans by glucocorticoids. Glucocorticoid hormones ensure the coordinated functioning of key components and mechanisms of hippocampal plasticity: neurogenesis, glutamatergic neurotransmission, microglia and astrocytes, systems of neurotrophic factors, neuroinflammation, proteases, metabolic hormones, neurosteroids. Regulatory mechanisms are diverse; along with the direct action of glucocorticoids through their receptors, there are conciliated glucocorticoid-dependent effects, as well as numerous interactions between various systems and components. Despite the fact that many connections in this complex regulatory scheme have not yet been established, the study of the factors and mechanisms considered in the work forms growth points in the field of glucocorticoid-regulated processes in the brain and primarily in the hippocampus. These studies are fundamentally important for the translation into the clinic and the potential treatment/prevention of common diseases of the emotional and cognitive spheres and respective comorbid conditions.
Collapse
Affiliation(s)
- Natalia V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia.
- Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, Moscow, 115419, Russia
| |
Collapse
|
15
|
Hamzé R, Delangre E, Tolu S, Moreau M, Janel N, Bailbé D, Movassat J. Type 2 Diabetes Mellitus and Alzheimer's Disease: Shared Molecular Mechanisms and Potential Common Therapeutic Targets. Int J Mol Sci 2022; 23:ijms232315287. [PMID: 36499613 PMCID: PMC9739879 DOI: 10.3390/ijms232315287] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
The global prevalence of diabetes mellitus and Alzheimer's disease is increasing alarmingly with the aging of the population. Numerous epidemiological data suggest that there is a strong association between type 2 diabetes and an increased risk of dementia. These diseases are both degenerative and progressive and share common risk factors. The amyloid cascade plays a key role in the pathophysiology of Alzheimer's disease. The accumulation of amyloid beta peptides gradually leads to the hyperphosphorylation of tau proteins, which then form neurofibrillary tangles, resulting in neurodegeneration and cerebral atrophy. In Alzheimer's disease, apart from these processes, the alteration of glucose metabolism and insulin signaling in the brain seems to induce early neuronal loss and the impairment of synaptic plasticity, years before the clinical manifestation of the disease. The large amount of evidence on the existence of insulin resistance in the brain during Alzheimer's disease has led to the description of this disease as "type 3 diabetes". Available animal models have been valuable in the understanding of the relationships between type 2 diabetes and Alzheimer's disease, but to date, the mechanistical links are poorly understood. In this non-exhaustive review, we describe the main molecular mechanisms that may link these two diseases, with an emphasis on impaired insulin and IGF-1 signaling. We also focus on GSK3β and DYRK1A, markers of Alzheimer's disease, which are also closely associated with pancreatic β-cell dysfunction and type 2 diabetes, and thus may represent common therapeutic targets for both diseases.
Collapse
Affiliation(s)
- Rim Hamzé
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Etienne Delangre
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Stefania Tolu
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Manon Moreau
- Team Degenerative Process, Stress and Aging, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Nathalie Janel
- Team Degenerative Process, Stress and Aging, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Danielle Bailbé
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Jamileh Movassat
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
- Correspondence: ; Tel.: +33-1-57-27-77-82; Fax: +33-1-57-27-77-91
| |
Collapse
|
16
|
Li S, Deng X, Zhang Y. The Triglyceride-Glucose Index Is Associated with Longitudinal Cognitive Decline in a Middle-Aged to Elderly Population: A Cohort Study. J Clin Med 2022; 11:jcm11237153. [PMID: 36498726 PMCID: PMC9737091 DOI: 10.3390/jcm11237153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/24/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND To examine the effect of the triglyceride-glucose (TyG) index on longitudinal cognitive decline in a healthy middle-aged-to-elderly population. METHODS We conducted a population-based longitudinal study. A total of 1774 participants without cognitive impairment were enrolled in the 4-year follow-up. They were divided into four groups according to the quartile of the TyG index. Multivariable-adjusted Cox proportional hazard models were performed to examine the association between the TyG index and cognitive decline. Discrimination tests were used to evaluate the incremental predictive value of the TyG index beyond conventional risk factors. RESULTS During the follow-up, compared with those in the bottom quartile group, participants in the top TyG quartile group presented a 51% increase in the risk of cognitive decline (OR 1.51 (95% CI: 1.06-2.14)). As shown by discrimination tests, adding the TyG index into the conventional model resulted in a slight improvement in predicting the risk of cognitive decline (NRI 16.00% (p = 0.004)). CONCLUSION This study demonstrated that increasing values of the TyG index were positively associated with the risk of cognitive decline. Monitoring the TyG index may help in the early identification of individuals at high risk of cognitive deterioration.
Collapse
Affiliation(s)
- Siqi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Xuan Deng
- Clinical Research Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yumei Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Department of Rehabilitation Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Correspondence: ; Tel.: +86-10-59975531
| |
Collapse
|
17
|
DHA/EPA supplementation decreases anxiety-like behaviour, but it does not ameliorate metabolic profile in obese male rats. Br J Nutr 2022; 128:964-974. [PMID: 34605386 DOI: 10.1017/s0007114521003998] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Obesity is a major public health problem that predisposes to several diseases and higher mortality in patients with COVID-19. Obesity also generates neuroinflammation, which predisposes to the development of neuropsychiatric diseases. Since there is a lack of effective treatments for obesity, the search for new strategies to reverse its consequences is urgent. In this perspective, the anti-inflammatory properties of omega-3 polyunsaturated fatty acids such as DHA/EPA might reduce the harmful effects of obesity. Here, we used the cafeteria diet (CAF) model to induce obesity in Wistar rats. Animals received ultra-processed food for 20 weeks, and DHA/EPA supplementation (500 mg/kg per d) was performed between the 16th and the 20th week. At the end of the experiment, it was evaluated: body weight, visceral fat deposition, plasma glucose, insulin and triglycerides, and it was also measured the levels of inflammatory cytokines TNF-α and IL-6 in plasma and liver, and TNF-α in the prefrontal cortex. The elevated plus maze test was performed to analyse anxiety-like behaviour. Our results demonstrated that DHA/EPA could not reverse weight and fat gain and did not modify plasma dosages. However, there was a decrease in IL-6 in the liver (DHA/EPA effect: P = 0.023) and TNF-α in the brain (CAF compared with CAF + DHA/EPA, P < 0.05). Also, there was a decrease in the anxiety index in CAF + DHA/EPA compared with the CAF group (P < 0.01). Thus, DHA/EPA supplementation is helpful to reverse the consequences of obesity in the brain.
Collapse
|
18
|
Ayala-Guerrero L, García-delaTorre P, Sánchez-García S, Guzmán-Ramos K. Serum Levels of Glial Fibrillary Acidic Protein Association with Cognitive Impairment and Type 2 Diabetes. Arch Med Res 2022; 53:501-507. [DOI: 10.1016/j.arcmed.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/12/2022] [Accepted: 06/14/2022] [Indexed: 12/15/2022]
|
19
|
Burzynski H, Macht V, Woodruff J, Crawford J, Erichsen J, Piroli G, Grillo C, Fadel J, Reagan L. Pyridostigmine bromide elicits progressive and chronic impairments in the cholinergic anti-inflammatory pathway in the prefrontal cortex and hippocampus of male rats. Neurobiol Stress 2022; 18:100446. [PMID: 35573808 PMCID: PMC9095881 DOI: 10.1016/j.ynstr.2022.100446] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/18/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
Gulf War Illness (GWI) is a multi-symptom illness that continues to affect over 250,000 American Gulf War veterans. The causes of GWI remain equivocal; however, prophylactic use of the acetylcholinesterase inhibitor pyridostigmine bromide (PB), and the stress of combat have been identified as two potential causative factors. Both PB and stress alter acetylcholine (ACh), which mediates both cognition and anti-inflammatory responses. As inflammation has been proposed to contribute to the cognitive deficits and immune dysregulation in GWI, the goal of this study was to determine the long-term effects of PB and stress on the cholinergic anti-inflammatory pathway in the central nervous system and periphery. We used our previously established rat model of GWI and in vivo microdialysis to assess cholinergic neurochemistry in the prefrontal cortex (PFC) and hippocampus following a mild immune challenge (lipopolysaccharide; LPS). We then examined LPS-induced changes in inflammatory markers in PFC and hippocampal homogenates. We found that PB treatment produces a long-lasting potentiation of the cholinergic response to LPS in both the PFC and hippocampus. Interestingly, this prolonged effect of PB treatment enhancing cholinergic responses to LPS was accompanied by paradoxical increases in the release of pro-inflammatory cytokines in these brain regions. Collectively, these findings provide evidence that neuroinflammation resulting from dysregulation of the cholinergic anti-inflammatory pathway is a mechanistic mediator in the progression of the neurochemical and neurocognitive deficits in GWI and more broadly suggest that dysregulation of this pathway may contribute to neuroinflammatory processes in stress-related neurological disorders. Inflammation is thought to contribute to the progressive nature of GWI pathology. PB potentiates the central cholinergic response to LPS over time in model of GWI. PB progressively exacerbates the neuroinflammatory response to LPS. GWI may result from the dysregulation of the cholinergic anti-inflammatory pathway.
Collapse
|
20
|
Erichsen JM, Fadel JR, Reagan LP. Peripheral versus central insulin and leptin resistance: Role in metabolic disorders, cognition, and neuropsychiatric diseases. Neuropharmacology 2022; 203:108877. [PMID: 34762922 PMCID: PMC8642294 DOI: 10.1016/j.neuropharm.2021.108877] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/14/2021] [Accepted: 11/04/2021] [Indexed: 02/06/2023]
Abstract
Insulin and leptin are classically regarded as peptide hormones that play key roles in metabolism. In actuality, they serve several functions in both the periphery and central nervous system (CNS). Likewise, insulin and leptin resistance can occur both peripherally and centrally. Metabolic disorders such as diabetes and obesity share several key features including insulin and leptin resistance. While the peripheral effects of these disorders are well-known (i.e. cardiovascular disease, hypertension, stroke, dyslipidemia, etc.), the CNS complications of leptin and insulin resistance have come into sharper focus. Both preclinical and clinical findings have indicated that insulin and leptin resistance are associated with cognitive deficits and neuropsychiatric diseases such as depression. Importantly, these studies also suggest that these deficits in neuroplasticity can be reversed by restoration of insulin and leptin sensitivity. In view of these observations, this review will describe, in detail, the peripheral and central functions of insulin and leptin and explain the role of insulin and leptin resistance in various metabolic disorders, cognition, and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Jennifer M Erichsen
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29208, USA.
| | - Jim R Fadel
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29208, USA
| | - Lawrence P Reagan
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29208, USA; Columbia VA Health Care System, Columbia, SC, 29208, USA
| |
Collapse
|
21
|
Erickson KI, Donofry SD, Sewell KR, Brown BM, Stillman CM. Cognitive Aging and the Promise of Physical Activity. Annu Rev Clin Psychol 2022; 18:417-442. [PMID: 35044793 DOI: 10.1146/annurev-clinpsy-072720-014213] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Is the field of cognitive aging irretrievably concerned with decline and deficits, or is it shifting to emphasize the hope of preservation and enhancement of cognitive function in late life? A fragment of an answer comes from research attempting to understand the reasons for individual variability in the extent and rate of cognitive decline. This body of work has created a sense of optimism based on evidence that there are some health behaviors that amplify cognitive performance or mitigate the rate of age-related cognitive decline. In this context, we discuss the role of physical activity on neurocognitive function in late adulthood and summarize how it can be conceptualized as a constructive approach both for the maintenance of cognitive function and as a therapeutic for enhancing or optimizing cognitive function in late life. In this way, physical activity research can be used to shape perceptions of cognitive aging. Expected final online publication date for the Annual Review of Clinical Psychology, Volume 18 is May 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Kirk I Erickson
- Department of Psychology and Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; .,Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia.,PROFITH "PROmoting FITness and Health through physical activity" Research Group, Department of Physical Education and Sports, Faculty of Sport Sciences, University of Granada, Granada, Spain
| | - Shannon D Donofry
- Department of Psychology and Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; .,Psychiatry and Behavioral Health Institute, Allegheny Health Network, Pittsburgh, Pennsylvania
| | - Kelsey R Sewell
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Belinda M Brown
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Chelsea M Stillman
- Department of Psychology and Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
| |
Collapse
|
22
|
Parent MB, Higgs S, Cheke LG, Kanoski SE. Memory and eating: A bidirectional relationship implicated in obesity. Neurosci Biobehav Rev 2022; 132:110-129. [PMID: 34813827 PMCID: PMC8816841 DOI: 10.1016/j.neubiorev.2021.10.051] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 10/17/2021] [Accepted: 10/28/2021] [Indexed: 01/03/2023]
Abstract
This paper reviews evidence demonstrating a bidirectional relationship between memory and eating in humans and rodents. In humans, amnesia is associated with impaired processing of hunger and satiety cues, disrupted memory of recent meals, and overconsumption. In healthy participants, meal-related memory limits subsequent ingestive behavior and obesity is associated with impaired memory and disturbances in the hippocampus. Evidence from rodents suggests that dorsal hippocampal neural activity contributes to the ability of meal-related memory to control future intake, that endocrine and neuropeptide systems act in the ventral hippocampus to provide cues regarding energy status and regulate learned aspects of eating, and that consumption of hypercaloric diets and obesity disrupt these processes. Collectively, this evidence indicates that diet-induced obesity may be caused and/or maintained, at least in part, by a vicious cycle wherein excess intake disrupts hippocampal functioning, which further increases intake. This perspective may advance our understanding of how the brain controls eating, the neural mechanisms that contribute to eating-related disorders, and identify how to treat diet-induced obesity.
Collapse
Affiliation(s)
- Marise B Parent
- Neuroscience Institute & Department of Psychology, Georgia State University, Box 5030, Atlanta, GA 30303-5030, United States.
| | - Suzanne Higgs
- School of Psychology, University of Birmingham, Edgbaston, Birmingham, BI5 2TT, United Kingdom.
| | - Lucy G Cheke
- Department of Psychology, University of Cambridge, Downing Street, Cambridge, CB2 3EB, United Kingdom.
| | - Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, 90089-0371, United States.
| |
Collapse
|
23
|
Hoirisch-Clapauch S. Mechanisms affecting brain remodeling in depression: do all roads lead to impaired fibrinolysis? Mol Psychiatry 2022; 27:525-533. [PMID: 34404914 DOI: 10.1038/s41380-021-01264-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/28/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023]
Abstract
Fibrinolysis occurs when plasminogen activators, such as tissue plasminogen activator (tPA), convert plasminogen to plasmin, which dissolves the fibrin clot. The proteolytic activity of tPA and plasmin is not restricted to fibrin degradation. In the extravascular space, these two proteases modify a variety of substrates other than fibrin, playing a crucial role in physiological and pathological tissue remodeling. In the brain, for example, tPA and plasmin mediate the conversion of brain-derived neurotrophic factor precursor (proBDNF) to mature brain-derived neurotrophic factor precursor (BDNF). Thus, the fibrinolytic system influences processes reported to be dysfunctional in depression, including neurogenesis, synaptic plasticity, and reward processing. The hypothesis that decreased fibrinolytic activity is an important element in the pathogenesis of depression is supported by the association between depression and increased levels of plasminogen activator inhibitor (PAI)-1, the main inhibitor of tPA. Also, various biochemical markers of depression induce PAI-1 synthesis, including hypercortisolism, hyperinsulinemia, hyperleptinemia, increased levels of cytokines, and hyperhomocysteinemia. Moreover, hypofibrinolysis provides a link between depression and emotional eating, binge eating, vegetarianism, and veganism. This paper discusses the role of reduced fibrinolytic activity in the bidirectional interplay between depression and its somatic manifestations and complications. It also reviews evidence that abnormal fibrinolysis links heterogeneous conditions associated with treatment-resistant depression. Understanding the role of hypofibrinolysis in depression may open new avenues for its treatment.
Collapse
|
24
|
Cente M, Zorad S, Smolek T, Fialova L, Paulenka Ivanovova N, Krskova K, Balazova L, Skrabana R, Filipcik P. Plasma Leptin Reflects Progression of Neurofibrillary Pathology in Animal Model of Tauopathy. Cell Mol Neurobiol 2022; 42:125-136. [PMID: 32997211 PMCID: PMC11441179 DOI: 10.1007/s10571-020-00972-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/23/2020] [Indexed: 10/23/2022]
Abstract
The close relationship between Alzheimer's disease (AD) and obesity was recognized many years ago. However, complete understanding of the pathological mechanisms underlying the interactions between degeneration of CNS and fat metabolism is still missing. The leptin a key adipokine of white adipose tissue has been suggested as one of the major mediators linking the obesity and AD. Here we investigated the association between peripheral levels of leptin, general metabolic status and stage of the pathogenesis in rat transgenic model of AD. We demonstrate significantly decreased levels of plasma leptin in animals with experimentally induced progressive neurofibrillary pathology, which represents only 62.3% (P = 0.0015) of those observed in normal wild type control animals. More detailed analysis showed a strong and statistically significant inverse correlation between the load of neurofibrillary pathology and peripheral levels of leptin (r = - 0.7248, P = 0.0177). We also observed a loss of body weight during development of neurodegeneration (about 14% less than control animals, P = 0.0004) and decrease in several metabolic parameters such as glucose, insulin, triglycerides and VLDL in plasma of the transgenic animals. Our data suggest that plasma leptin could serve as a convenient peripheral biomarker for tauopathies and Alzheimer's disease. Decrease in gene expression of leptin in fat tissue and its plasma level was found as one of the consequences of experimentally induced neurodegeneration. Our data may help to design rational diagnostic and therapeutic strategies for patients suffering from Alzheimer's disease or other forms of tauopathy.
Collapse
Affiliation(s)
- Martin Cente
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10, Bratislava, Slovakia
- Axon Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Stefan Zorad
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tomas Smolek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10, Bratislava, Slovakia
- Axon Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Lubica Fialova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10, Bratislava, Slovakia
- Axon Neuroscience R&D Services SE, Bratislava, Slovakia
| | | | - Katarina Krskova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lucia Balazova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Rostislav Skrabana
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10, Bratislava, Slovakia
- Axon Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Peter Filipcik
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10, Bratislava, Slovakia.
- Axon Neuroscience R&D Services SE, Bratislava, Slovakia.
| |
Collapse
|
25
|
Yang S, Boudier-Revéret M, Kwon S, Lee MY, Chang MC. Effect of Diabetes on Post-stroke Recovery: A Systematic Narrative Review. Front Neurol 2021; 12:747878. [PMID: 34970205 PMCID: PMC8712454 DOI: 10.3389/fneur.2021.747878] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/19/2021] [Indexed: 01/11/2023] Open
Abstract
Background: Patients with stroke often have comorbid diabetes. Considering its detrimental effects on brain function, diabetes may increase the risk of poor recovery. Methods: The aim of this review was to investigate the effect of diabetes on post-stroke recovery by a systematic review. Several specific aspects of post-stroke recovery, including activities of daily living (ADL), motor, cognitive, and quality of life (QOL) recovery, were examined. We searched the PubMed, SCOPUS, Embase, and Cochrane Library databases for relevant studies on the effect of diabetes on post-stroke recovery, published until May 26, 2021. A total of 52,051 potentially relevant articles were identified. After reading the titles and abstracts and assessing their eligibility based on full-text articles, 34 publications were included in this review. Results: Of 29 studies that assessed ADL recovery after stroke, 22 studies suggested that diabetes had a negative effect on recovery of ADL after stroke. Regarding motor recovery, only one out of four studies showed that diabetes had some effect on motor recovery after stroke. Of the two studies on cognitive recovery, one reported that diabetes was an independent predictor of poor cognitive recovery after stroke. Three studies on QOL reported that a poor QOL after stroke was associated with the presence of diabetes. Conclusions: The current review suggests that the post-stroke recovery of ADL seems to be poorer in patients with diabetes than patients without diabetes. Further, there are insufficient data to conclude the effect of diabetes on motor and cognitive recovery, but it may have some influence on the quality of life after stroke. Systematic Review Registration: doi: 10.37766/inplasy2021.11.0032, identifier: INPLASY2021110032.
Collapse
Affiliation(s)
- Seoyon Yang
- Department of Rehabilitation Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University School of Medicine, Seoul, South Korea
| | - Mathieu Boudier-Revéret
- Department of Physical Medicine and Rehabilitation, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - SuYeon Kwon
- Department of Rehabilitation Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University School of Medicine, Seoul, South Korea
| | - Min Yong Lee
- Department of Dermatology, Ewha Womans University Seoul Hospital, Ewha Womans University School of Medicine, Seoul, South Korea
| | - Min Cheol Chang
- Department of Rehabilitation Medicine, College of Medicine, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
26
|
Nasca C, Barnhill O, DeAngelis P, Watson K, Lin J, Beasley J, Young SP, Myoraku A, Dobbin J, Bigio B, McEwen B, Rasgon N. Multidimensional predictors of antidepressant responses: Integrating mitochondrial, genetic, metabolic and environmental factors with clinical outcomes. Neurobiol Stress 2021; 15:100407. [PMID: 34815985 PMCID: PMC8592929 DOI: 10.1016/j.ynstr.2021.100407] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 09/17/2021] [Accepted: 10/04/2021] [Indexed: 12/02/2022] Open
Abstract
Major depressive disorder (MDD) is a primary psychiatric illness worldwide; there is a dearth of new mechanistic models for the development of better therapeutic strategies. Although we continue to discover individual biological factors, a major challenge is the identification of integrated, multidimensional traits underlying the complex heterogeneity of depression and treatment outcomes. Here, we set out to ascertain the emergence of the novel mitochondrial mediator of epigenetic function acetyl-L-carnitine (LAC) in relation to previously described individual predictors of antidepressant responses to the insulin-sensitizing agent pioglitazone. Herein, we report that i) subjects with MDD and shorter leukocyte telomere length (LTL) show decreased levels of LAC, increased BMI, and a history of specific types of childhood trauma; and that ii) these multidimensional factors spanning mitochondrial metabolism, cellular aging, metabolic function, and childhood trauma provide more detailed signatures to predict longitudinal changes in depression severity in response to pioglitazone than individual factors. The findings of multidimensional signatures involved in the pathophysiology of depression and their role in predicting treatment outcomes provide a starting point for the development of a mechanistic framework linking biological networks and environmental factors to clinical outcomes in pursuit of personalized medicine strategies to effectively treat MDD.
Collapse
Affiliation(s)
- Carla Nasca
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, USA
- Department of Psychiatry, New York University, Grossman School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University Neuroscience Institute, New York University, New York, NY, USA
- Corresponding author. Department of Psychiatry, New York University, Grossman School of Medicine, New York, NY, USA.
| | - Olivia Barnhill
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, USA
| | - Paolo DeAngelis
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, USA
| | - Kathleen Watson
- Center for Neuroscience in Women's Health, Stanford University, Palo Alto, USA
| | - Jue Lin
- Department of Psychiatry, University of California, San Francisco, San Francisco, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, USA
| | - James Beasley
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Sarah P. Young
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Biochemical Genetics Laboratory, Duke University Health System, Durham, USA
| | - Alison Myoraku
- Center for Neuroscience in Women's Health, Stanford University, Palo Alto, USA
| | - Josh Dobbin
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, USA
| | - Benedetta Bigio
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, USA
- Biostatistics and Experimental Research Design, Center for Clinical and Translational Science, Rockefeller University, New York, USA
| | - Bruce McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, USA
| | - Natalie Rasgon
- Center for Neuroscience in Women's Health, Stanford University, Palo Alto, USA
| |
Collapse
|
27
|
Meng J, Zhu Y, Ma H, Wang X, Zhao Q. The role of traditional Chinese medicine in the treatment of cognitive dysfunction in type 2 diabetes. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114464. [PMID: 34329715 DOI: 10.1016/j.jep.2021.114464] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/04/2021] [Accepted: 07/24/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Diabetic cognitive dysfunction (DCD) is mainly one of the complications of type 2 diabetes mellitus (T2DM) with complex and obscure pathogenesis. Extensive evidence has demonstrated the effectiveness and safety of traditional Chinese medicine (TCM) for DCD management. AIM OF THE STUDY This review attempted to systematically summarize the possible pathogenesis of DCD and the current Chinese medicine on the treatment of DCD. MATERIALS AND METHODS We acquired information of TCM on DCD treatment from PubMed, Web of Science, Science Direct and CNKI databases. We then dissected the potential mechanisms of currently reported TCMs and their active ingredients for the treatment of DCD by discussing the deficiencies and giving further recommendations. RESULTS Most TCMs and their active ingredients could improve DCD through alleviating insulin resistance, microvascular dysfunction, abnormal gut microbiota composition, inflammation, and the damages of the blood-brain barrier, cerebrovascular and neurons under hyperglycemia conditions. CONCLUSIONS TCM is effective in the treatment of DCD with few adverse reactions. A large number of in vivo and in vitro, and clinical trials are still needed to further reveal the potential quality markers of TCM on DCD treatment.
Collapse
Affiliation(s)
- Jinni Meng
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, Ningxia, China
| | - Yafei Zhu
- College of Basic Medicine, Ningxia Medical University, Ningxia, China
| | - Huixia Ma
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, Ningxia, China
| | - Xiaobo Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Qipeng Zhao
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, Ningxia, China; Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Ningxia, China.
| |
Collapse
|
28
|
Reagan L, Cowan H, Woodruff J, Piroli G, Erichsen J, Evans A, Burzynski H, Maxwell N, Loyo-Rosado F, Macht V, Grillo C. Hippocampal-specific insulin resistance elicits behavioral despair and hippocampal dendritic atrophy. Neurobiol Stress 2021; 15:100354. [PMID: 34258333 PMCID: PMC8252121 DOI: 10.1016/j.ynstr.2021.100354] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 01/02/2023] Open
Abstract
Insulin resistance is a major contributor to the neuroplasticity deficits observed in patients with metabolic disorders. However, the relative contribution of peripheral versus central insulin resistance in the development of neuroplasticity deficits remains equivocal. To distinguish between peripheral and central insulin resistance, we developed a lentiviral vector containing an antisense sequence selective for the insulin receptor (LV-IRAS). We previously demonstrated that intra-hippocampal injection of this vector impairs synaptic transmission and hippocampal-dependent learning and memory in the absence of peripheral insulin resistance. In view of the increased risk for the development of neuropsychiatric disorders in patients with insulin resistance, the current study examined depressive and anxiety-like behaviors, as well as hippocampal structural plasticity in rats with hippocampal-specific insulin resistance. Following hippocampal administration of either the LV-control virus or the LV-IRAS, anhedonia was evaluated by the sucrose preference test, despair behavior was assessed in the forced swim test, and anxiety-like behaviors were determined in the elevated plus maze. Hippocampal neuron morphology was studied by Golgi-Cox staining. Rats with hippocampal insulin resistance exhibited anxiety-like behaviors and behavioral despair without differences in anhedonia, suggesting that some but not all components of depressive-like behaviors were affected. Morphologically, hippocampal-specific insulin resistance elicited atrophy of the basal dendrites of CA3 pyramidal neurons and dentate gyrus granule neurons, and also reduced the expression of immature dentate gyrus granule neurons. In conclusion, hippocampal-specific insulin resistance elicits structural deficits that are accompanied by behavioral despair and anxiety-like behaviors, identifying hippocampal insulin resistance as a key factor in depressive illness.
Collapse
Affiliation(s)
- L.P. Reagan
- Columbia VA Health Care System, Columbia, SC, 29209, USA
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29209, USA
| | - H.B. Cowan
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29209, USA
| | - J.L. Woodruff
- Columbia VA Health Care System, Columbia, SC, 29209, USA
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29209, USA
| | - G.G. Piroli
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29209, USA
| | - J.M. Erichsen
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29209, USA
| | - A.N. Evans
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29209, USA
| | - H.E. Burzynski
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29209, USA
| | - N.D. Maxwell
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29209, USA
| | - F.Z. Loyo-Rosado
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29209, USA
| | - V.A. Macht
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29209, USA
| | - C.A. Grillo
- Columbia VA Health Care System, Columbia, SC, 29209, USA
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, 29209, USA
| |
Collapse
|
29
|
Macht V, Vetreno R, Elchert N, Crews F. Galantamine prevents and reverses neuroimmune induction and loss of adult hippocampal neurogenesis following adolescent alcohol exposure. J Neuroinflammation 2021; 18:212. [PMID: 34530858 PMCID: PMC8447570 DOI: 10.1186/s12974-021-02243-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/18/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Binge ethanol exposure during adolescence reduces hippocampal neurogenesis, a reduction which persists throughout adulthood despite abstinence. This loss of neurogenesis, indicated by reduced doublecortin+ immunoreactivity (DCX+IR), is paralleled by an increase in hippocampal proinflammatory signaling cascades. As galantamine, a cholinesterase inhibitor, has anti-inflammatory actions, we tested the hypothesis that galantamine would prevent (study 1) or restore (study 2) AIE induction of proinflammatory signals within the hippocampus as well as AIE-induced loss of hippocampal neurogenesis. METHODS Galantamine (4 mg/kg) or vehicle (saline) was administered to Wistar rats during adolescent intermittent ethanol (AIE; 5.0 g/kg ethanol, 2 days on/2 days off, postnatal day [P] 25-54) (study 1, prevention) or after AIE during abstinent maturation to adulthood (study 2, restoration). RESULTS Results indicate AIE reduced DCX+IR and induced cleaved caspase3 (Casp3) in DCX-expressing immature neurons. Excitingly, AIE induction of activated Casp3 in DCX-expressing neurons is both prevented and reversed by galantamine treatment, which also resulted in prevention and restoration of neurogenesis (DCX+IR). Similarly, galantamine prevented and/or reversed AIE induction of proinflammatory markers, including the chemokine (C-C motif) ligand 2 (CCL2), cyclooxygenase-2 (COX-2), and high mobility group box 1 (HMGB1) protein, suggesting that AIE induction of proinflammatory signaling mediates both cell death cascades and hippocampal neurogenesis. Interestingly, galantamine treatment increased Ki67+IR generally as well as increased pan-Trk expression specifically in AIE-treated rats but failed to reverse AIE induction of NADPH-oxidase (gp91phox). CONCLUSIONS Collectively, our studies suggest that (1) loss of neurogenesis after AIE is mediated by persistent induction of proinflammatory cascades which drive activation of cell death machinery in immature neurons, and (2) galantamine can prevent and restore AIE disruptions in the hippocampal environmental milieu to then prevent and restore AIE-mediated loss of neurogenesis.
Collapse
Affiliation(s)
- Victoria Macht
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 104 Manning Drive, Chapel Hill, NC, 27599, USA.
| | - Ryan Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 104 Manning Drive, Chapel Hill, NC, 27599, USA
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Natalie Elchert
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 104 Manning Drive, Chapel Hill, NC, 27599, USA
| | - Fulton Crews
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 104 Manning Drive, Chapel Hill, NC, 27599, USA
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
30
|
Hersey M, Woodruff JL, Maxwell N, Sadek AT, Bykalo MK, Bain I, Grillo CA, Piroli GG, Hashemi P, Reagan LP. High-fat diet induces neuroinflammation and reduces the serotonergic response to escitalopram in the hippocampus of obese rats. Brain Behav Immun 2021; 96:63-72. [PMID: 34010713 PMCID: PMC8319113 DOI: 10.1016/j.bbi.2021.05.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/22/2021] [Accepted: 05/14/2021] [Indexed: 12/27/2022] Open
Abstract
Clinical studies indicate that obese individuals have an increased risk of developing co-morbid depressive illness and that these patients have reduced responses to antidepressant therapy, including selective serotonin reuptake inhibitors (SSRIs). Obesity, a condition of chronic mild inflammation including obesity-induced neuroinflammation, is proposed to contribute to decreases in synaptic concentrations of neurotransmitters like serotonin (5HT) by decreasing 5HT synthesis in the dorsal raphe nucleus (DRN) and/or affecting 5HT reuptake in DRN target regions like the hippocampus. In view of these observations, the goal of the current study was to examine inflammatory markers and serotonergic dynamics in co-morbid obesity and depression. Biochemical and behavioral assays revealed that high-fat diet produced an obesity and depressive-like phenotype in one cohort of rats and that these changes were marked by increases in key pro-inflammatory cytokines in the hippocampus. In real time using fast scan cyclic voltammetry (FSCV), we observed no changes in basal levels of hippocampal 5HT; however responses to escitalopram were significantly impaired in the hippocampus of obese rats compared to diet resistant rats and control rats. Further studies revealed that these neurochemical observations could be explained by increases in serotonin transporter (SERT) expression in the hippocampus driven by elevated neuroinflammation. Collectively, these results demonstrate that obesity-induced increases in neuroinflammation adversely affect SERT expression in the hippocampus of obese rats, thereby providing a potential synaptic mechanism for reduced SSRI responsiveness in obese subjects with co-morbid depressive illness.
Collapse
Affiliation(s)
- Melinda Hersey
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA,Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina SOM, Columbia, SC, USA
| | - Jennifer L. Woodruff
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina SOM, Columbia, SC, USA,Columbia VA Health Care System, Columbia, SC, USA
| | - Nicholas Maxwell
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina SOM, Columbia, SC, USA
| | - Alia T. Sadek
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina SOM, Columbia, SC, USA
| | - Maria K. Bykalo
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina SOM, Columbia, SC, USA
| | - Ian Bain
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Claudia A. Grillo
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina SOM, Columbia, SC, USA,Columbia VA Health Care System, Columbia, SC, USA
| | - Gerardo G. Piroli
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina SOM, Columbia, SC, USA
| | - Parastoo Hashemi
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA,Department of Bioengineering, Imperial College, London, SW7 2AZ UK
| | - Lawrence P. Reagan
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina SOM, Columbia, SC, USA,Columbia VA Health Care System, Columbia, SC, USA,Corresponding author: Lawrence P. Reagan, Ph.D., Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Road, D40, Columbia, SC, USA 29208, Phone: 001 803 216 3515; Fax: 001 803 216 3538,
| |
Collapse
|
31
|
Rojas M, Chávez-Castillo M, Pirela D, Parra H, Nava M, Chacín M, Angarita L, Añez R, Salazar J, Ortiz R, Durán Agüero S, Gravini-Donado M, Bermúdez V, Díaz-Camargo E. Metabolic Syndrome: Is It Time to Add the Central Nervous System? Nutrients 2021; 13:nu13072254. [PMID: 34208833 PMCID: PMC8308252 DOI: 10.3390/nu13072254] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/05/2021] [Accepted: 06/09/2021] [Indexed: 12/28/2022] Open
Abstract
Metabolic syndrome (MS) is a set of cardio-metabolic risk factors that includes central obesity, hyperglycemia, hypertension, and dyslipidemias. The syndrome affects 25% of adults worldwide. The definition of MS has evolved over the last 80 years, with various classification systems and criteria, whose limitations and benefits are currently the subject of some controversy. Likewise, hypotheses regarding the etiology of MS add more confusion from clinical and epidemiological points of view. The leading suggestion for the pathophysiology of MS is insulin resistance (IR). IR can affect multiple tissues and organs, from the classic “triumvirate” (myocyte, adipocyte, and hepatocyte) to possible effects on organs considered more recently, such as the central nervous system (CNS). Mild cognitive impairment (MCI) and Alzheimer’s disease (AD) may be clinical expressions of CNS involvement. However, the association between MCI and MS is not understood. The bidirectional relationship that seems to exist between these factors raises the questions of which phenomenon occurs first and whether MCI can be a precursor of MS. This review explores shared pathophysiological mechanisms between MCI and MS and establishes a hypothesis of a possible MCI role in the development of IR and the appearance of MS.
Collapse
Affiliation(s)
- Milagros Rojas
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | | | - Daniela Pirela
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | - Heliana Parra
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | - Manuel Nava
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | - Maricarmen Chacín
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 08002, Colombia;
| | - Lissé Angarita
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andrés Bello, Sede Concepción 4260000, Chile;
| | - Roberto Añez
- Departamento de Endocrinología y Nutrición, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain;
| | - Juan Salazar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | - Rina Ortiz
- Posgrado, Carrera de Medicina, Universidad Católica de Cuenca, Cantón de Cuenca 010101, Ecuador;
| | - Samuel Durán Agüero
- Facultad de Ciencias Para el Cuidado de la Salud, Universidad San Sebastián, Los Leones 8420524, Chile;
| | - Marbel Gravini-Donado
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Barranquilla 080002, Colombia;
| | - Valmore Bermúdez
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540006, Colombia;
| | - Edgar Díaz-Camargo
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540006, Colombia;
- Correspondence:
| |
Collapse
|
32
|
Moustafa SR. The immune-opioid axis in prediabetes: predicting prediabetes with insulin resistance by plasma interleukin-10 and endomorphin-2 to kappa-opioid receptors ratio. Diabetol Metab Syndr 2021; 13:61. [PMID: 34099024 PMCID: PMC8185911 DOI: 10.1186/s13098-021-00677-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 05/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Prediabetes is characterized by a hemoglobin A1c of 5.7-6.4% and fasting blood glucose of 100-125 mg/dl. A high percentage of prediabetes subjects develop type 2 diabetes mellitus in the next years. The effects of opioid peptides and their receptors, in addition to immunological cytokines, on prediabetes are not well understood. Therefore, molecular, physiological, and clinical studies are required to link the opioid system, immune system, and insulin resistance (IR) in prediabetes. We hypothesize that opioid peptides (endomorphin-2 (EM2), and β-endorphin (βEP)), and their receptors (µ-opioid receptors (MOR) and κ-opioid receptors (KOR)), in addition to the inflammatory cytokines (IL-6) and anti-inflammatory cytokine (IL-10), affect IR parameters in patients with prediabetes. METHODS Sixty prediabetes patients with IR (prediabetes+IR) and sixty prediabetes patients without IR (prediabetes-IR), in addition to 58 controls, have participated in the study. IL-6, IL-10, EM2, βEP, MOR, and KOR were measured by the ELISA technique. RESULTS In general, most prediabetes subjects have dyslipidemia. The IL-6, IL-10, β-endorphin, MOR, and endomorphin-2 were higher in the prediabetes subgroups than the control group. The immune system was activated in the prediabetes in an IR-dependent manner. Prediabetes+IR can be predicted by the increased levels of IL-10, βEP, and EM2 and by the combination of IL-10 and EM2/KOR with good sensitivity and specificity. CONCLUSION Opioid peptides and their receptors were upregulated in patients with prediabetes, depending on the significance of IR and the immune cytokines. The intercorrelation between the immune system, EOS, and insulin in prediabetes was confirmed.
Collapse
Affiliation(s)
- Shatha Rouf Moustafa
- Clinical Analysis Department, College of Pharmacy, Hawler Medical University, Roya Towers C21, Erbil, Iraq.
| |
Collapse
|
33
|
Diaz A, Muñoz-Arenas G, Venegas B, Vázquez-Roque R, Flores G, Guevara J, Gonzalez-Vergara E, Treviño S. Metforminium Decavanadate (MetfDeca) Treatment Ameliorates Hippocampal Neurodegeneration and Recognition Memory in a Metabolic Syndrome Model. Neurochem Res 2021; 46:1151-1165. [PMID: 33559829 DOI: 10.1007/s11064-021-03250-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/02/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
The consumption of foods rich in carbohydrates, saturated fat, and sodium, accompanied by a sedentary routine, are factors that contribute to the progress of metabolic syndrome (MS). In this way, they cause the accumulation of body fat, hypertension, dyslipidemia, and hyperglycemia. Additionally, MS has been shown to cause oxidative stress, inflammation, and death of neurons in the hippocampus. Consequently, spatial and recognition memory is affected. It has recently been proposed that metformin decavanadate (MetfDeca) exerts insulin mimetic effects that enhance metabolism in MS animals; however, what effects it can cause on the hippocampal neurons of rats with MS are unknown. The objective of the work was to evaluate the effect of MetfDeca on hippocampal neurodegeneration and recognition memory in rats with MS. Administration of MetfDeca for 60 days in MS rats improved object recognition memory (NORt). In addition, MetfDeca reduced markers of oxidative stress and hippocampal neuroinflammation. Accompanied by an increase in the density and length of the dendritic spines of the hippocampus of rats with MS. We conclude that MetfDeca represents an important therapeutic agent to treat MS and induce neuronal and cognitive restoration mechanisms.
Collapse
Affiliation(s)
- Alfonso Diaz
- Faculty of Chemical Sciences, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Guadalupe Muñoz-Arenas
- Faculty of Chemical Sciences, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Berenice Venegas
- Faculty of Biological Sciences, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Rubén Vázquez-Roque
- Laboratory of Neuropsychiatry, Institute of Physiology, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Gonzalo Flores
- Laboratory of Neuropsychiatry, Institute of Physiology, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Jorge Guevara
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | | | - Samuel Treviño
- Faculty of Chemical Sciences, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico.
| |
Collapse
|
34
|
Hernández-Ramírez S, Osorio-Gómez D, Escobar ML, Rodríguez-Durán L, Velasco M, Bermúdez-Rattoni F, Hiriart M, Guzmán-Ramos KR. Catecholaminergic stimulation restores high-sucrose diet-induced hippocampal dysfunction. Psychoneuroendocrinology 2021; 127:105178. [PMID: 33706043 DOI: 10.1016/j.psyneuen.2021.105178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 02/20/2021] [Accepted: 02/24/2021] [Indexed: 11/15/2022]
Abstract
Increasing evidence suggests that long-term consumption of high-caloric diets increases the risk of developing cognitive dysfunctions. In the present study, we assessed the catecholaminergic activity in the hippocampus as a modulatory mechanism that is altered in rats exposed to six months of a high-sucrose diet (HSD). Male Wistar rats fed with this diet developed a metabolic disorder and showed impaired spatial memory in both water maze and object location memory (OLM) tasks. Intrahippocampal free-movement microdialysis showed a diminished dopaminergic and noradrenergic response to object exploration during OLM acquisition compared to rats fed with normal diet. In addition, electrophysiological results revealed an impaired long-term potentiation (LTP) of the perforant to dentate gyrus pathway in rats exposed to a HSD. Local administration of nomifensine, a catecholaminergic reuptake inhibitor, prior to OLM acquisition or LTP induction, improved long-term memory and electrophysiological responses, respectively. These results suggest that chronic exposure to HSD induces a hippocampal deterioration which impacts on cognitive and neural plasticity events negatively; these impairments can be ameliorated by increasing or restituting the affected catecholaminergic activity.
Collapse
Affiliation(s)
- Susana Hernández-Ramírez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Daniel Osorio-Gómez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Martha L Escobar
- División de Investigación y Estudios de Posgrado, Facultad de Psicología, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Luis Rodríguez-Durán
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Myrian Velasco
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Federico Bermúdez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Marcia Hiriart
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico.
| | - Kioko R Guzmán-Ramos
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Lerma, Av. de las Garzas No. 10, Col. El Panteón, Lerma de Villada, Estado de México, C.P. 52005, Mexico.
| |
Collapse
|
35
|
Santos-Bezerra DP, Cavaleiro AM, Santos AS, Suemoto CK, Pasqualucci CA, Jacob-Filho W, Leite REP, Passarelli M, Marie SKN, Machado UF, Correa-Giannella ML. Alcohol Use Disorder is Associated with Upregulation of MicroRNA-34a and MicroRNA-34c in Hippocampal Postmortem Tissue. Alcohol Clin Exp Res 2021; 45:64-68. [PMID: 33190281 DOI: 10.1111/acer.14505] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/04/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND To investigate epigenetic mechanisms potentially involved in the cognitive decline associated with chronic alcohol intake, we evaluated the expressions of three micro-RNAs (miR-34a, -34b, and -34c) highly expressed in the hippocampus and involved in neuronal physiology and pathology. MiR-34a participates in functioning and survival of mature neurons; miR-34b is associated with Alzheimer-like disorders; and miR-34c is implicated in the memory impairment of Alzheimer disease in rodents and humans. METHODS A total of 69 cases were selected from the Biobank for Aging Studies and categorized according to the absence (n = 50) or presence (n = 19) of alcohol use disorder (AUD). Cases presenting with neuropathological diagnoses of dementias were excluded. Total RNA was extracted from hippocampal paraffinized slices, complementary DNA was synthesized from miRs, and RT-qPCR was performed with TaqMan® assays. RESULTS Higher expressions of miR-34a and miR-34c, but not of miR-34b, were found in the group with AUD in comparison with the group without AUD after adjustment for potential confounders (age, sex, body mass index, presence of hypertension, diabetes mellitus, smoking, and physical inactivity). CONCLUSIONS Hippocampal upregulation of miR-34a and miR-34c may be involved in the cognitive decline associated with chronic alcohol consumption.
Collapse
Affiliation(s)
- Daniele P Santos-Bezerra
- From the Laboratorio de Carboidratos e Radioimunoensaio (LIM-18) do Hospital das Clinicas HCFMUSP, DPS-B, AMC, ASS, MLC-G Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Ana Mercedes Cavaleiro
- From the Laboratorio de Carboidratos e Radioimunoensaio (LIM-18) do Hospital das Clinicas HCFMUSP, DPS-B, AMC, ASS, MLC-G Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Aritania Sousa Santos
- From the Laboratorio de Carboidratos e Radioimunoensaio (LIM-18) do Hospital das Clinicas HCFMUSP, DPS-B, AMC, ASS, MLC-G Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Claudia Kimie Suemoto
- Division of Geriatrics (CKS, WJ-F, REPL), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | | | - Wilson Jacob-Filho
- Division of Geriatrics (CKS, WJ-F, REPL), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Renata Elaine Paraizo Leite
- Division of Geriatrics (CKS, WJ-F, REPL), University of Sao Paulo Medical School, Sao Paulo, Brazil.,Department of Pathology (CAP, REPL), University of Sao Paulo, Sao Paulo, Brazil
| | - Marisa Passarelli
- Laboratorio de Lipides (LIM-10) do HCFMUSP (MP), Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, Brazil.,Programa de Pos-Graduacao em Medicina (MP, MLC-G), Universidade Nove de Julho (UNINOVE), Sao Paulo, Brazil
| | | | - Ubiratan Fabres Machado
- Departmento de Fisiologia e Biofisica (UFM), Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Maria Lucia Correa-Giannella
- From the Laboratorio de Carboidratos e Radioimunoensaio (LIM-18) do Hospital das Clinicas HCFMUSP, DPS-B, AMC, ASS, MLC-G Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil.,Programa de Pos-Graduacao em Medicina (MP, MLC-G), Universidade Nove de Julho (UNINOVE), Sao Paulo, Brazil
| |
Collapse
|
36
|
Chen M, Huang N, Liu J, Huang J, Shi J, Jin F. AMPK: A bridge between diabetes mellitus and Alzheimer's disease. Behav Brain Res 2020; 400:113043. [PMID: 33307136 DOI: 10.1016/j.bbr.2020.113043] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023]
Abstract
The pathogenesis and etiology of diabetes mellitus (DM) and Alzheimer's disease (AD) share many common cellular and molecular themes. Recently, a growing body of research has shown that AMP-activated protein kinase (AMPK), a biomolecule that regulates energy balance and glucose and lipid metabolism, plays key roles in DM and AD. In this review, we summarize the relevant research on the roles of AMPK in DM and AD, including its functions in gluconeogenesis and insulin resistance (IR) and its relationships with amyloid β-protein (Aβ), Tau and AMPK activators. In DM, AMPK is involved in the regulation of glucose metabolism and IR. AMPK is closely related to gluconeogenesis, which can not only be activated by the upstream kinases liver kinase B1 (LKB1), transforming growth factor β-activated kinase 1 (TAK1), and Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ) but also regulate the downstream kinases glucose-6-phosphatase (G-6-Pase) and phosphoenolpyruvate carboxy kinase (PEPCK), thereby affecting gluconeogenesis and ameliorating DM. Moreover, AMPK can regulate glucose transporter 4 (GLUT4) and free fatty acids to improve IR. In AD, AMPK can ameliorate abnormal brain energy metabolism, not only by reduces Aβ deposition through β-secretase but also reduces tau hyperphosphorylation through sirtuin 1 (SIRT1) and protein phosphatase 2A (PP2A). Therefore, AMPK is a bridge between DM and AD.
Collapse
Affiliation(s)
- Meixiang Chen
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Nanqu Huang
- National Drug Clinical Trial Institution, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Ju Liu
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Juan Huang
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Feng Jin
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
37
|
Ettcheto M, Busquets O, Espinosa-Jiménez T, Verdaguer E, Auladell C, Camins A. A Chronological Review of Potential Disease-Modifying Therapeutic Strategies for Alzheimer's Disease. Curr Pharm Des 2020; 26:1286-1299. [PMID: 32066356 DOI: 10.2174/1381612826666200211121416] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 12/18/2019] [Indexed: 01/28/2023]
Abstract
Late-onset Alzheimer's disease (LOAD) is a neurodegenerative disorder that has become a worldwide health problem. This pathology has been classically characterized for its affectation on cognitive function and the presence of depositions of extracellular amyloid β-protein (Aβ) and intracellular neurofibrillary tangles (NFT) composed of hyperphosphorylated Tau protein. To this day, no effective treatment has been developed. Multiple strategies have been proposed over the years with the aim of finding new therapeutic approaches, such as the sequestration of Aβ in plasma or the administration of anti-inflammatory drugs. Also, given the significant role of the insulin receptor in the brain in the proper maintenance of cognitive function, drugs focused on the amelioration of insulin resistance have been proposed as potentially useful and effective in the treatment of AD. In the present review, taking into account the molecular complexity of the disease, it has been proposed that the most appropriate therapeutic strategy is a combinatory treatment of several drugs that will regulate a wide spectrum of the described altered pathological pathways.
Collapse
Affiliation(s)
- Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Faculty of Medicine and Life Sciences, University Rovira i Virgili, Reus, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Oriol Busquets
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Faculty of Medicine and Life Sciences, University Rovira i Virgili, Reus, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Triana Espinosa-Jiménez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Ester Verdaguer
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Carme Auladell
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| |
Collapse
|
38
|
Western diet-induced obesity disrupts the diurnal rhythmicity of hippocampal core clock gene expression in a mouse model. Brain Behav Immun 2020; 88:815-825. [PMID: 32454134 DOI: 10.1016/j.bbi.2020.05.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/17/2020] [Accepted: 05/20/2020] [Indexed: 12/22/2022] Open
Abstract
Western diet (WD) feeding disrupts core clock gene expression in peripheral tissues and contributes to WD-induced metabolic disease. The hippocampus, the mammalian center for memory, is also sensitive to WD feeding, but whether the WD disrupts its core clock is unknown. To this end, male mice were maintained on a WD for 16 weeks and diurnal metabolism, gene expression and memory were assessed. WD-induced obesity disrupted the diurnal rhythms of whole-body metabolism, markers of inflammation and hepatic gene expression, but did not disrupt diurnal expression of hypothalamic Bmal1, Npas2 and Per2. However, all measured core clock genes were disrupted in the hippocampus after WD feeding and the expression pattern of genes implicated in Alzheimer's disease and synaptic function were altered. Finally, WD feeding disrupted hippocampal memory in a task- and time-dependent fashion. Our results implicate WD-induced alterations in the rhythmicity of hippocampal gene expression in the etiology of diet-induced memory deficits.
Collapse
|
39
|
Teixeira AE, Rocha-Gomes A, Pereira dos Santos T, Amaral BLS, da Silva AA, Malagutti AR, Leite FRF, Stuckert-Seixas SR, Riul TR. Cafeteria diet administered from lactation to adulthood promotes a change in risperidone sensitivity on anxiety, locomotion, memory, and social interaction of Wistar rats. Physiol Behav 2020; 220:112874. [DOI: 10.1016/j.physbeh.2020.112874] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 01/24/2023]
|
40
|
Caputo V, Termine A, Strafella C, Giardina E, Cascella R. Shared (epi)genomic background connecting neurodegenerative diseases and type 2 diabetes. World J Diabetes 2020; 11:155-164. [PMID: 32477452 PMCID: PMC7243483 DOI: 10.4239/wjd.v11.i5.155] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/10/2020] [Accepted: 03/22/2020] [Indexed: 02/05/2023] Open
Abstract
The progressive aging of populations has resulted in an increased prevalence of chronic pathologies, especially of metabolic, neurodegenerative and movement disorders. In particular, type 2 diabetes (T2D), Alzheimer’s disease (AD) and Parkinson’s disease (PD) are among the most prevalent age-related, multifactorial pathologies that deserve particular attention, given their dramatic impact on patient quality of life, their economic and social burden as well the etiopathogenetic mechanisms, which may overlap in some cases. Indeed, the existence of common triggering factors reflects the contribution of mutual genetic, epigenetic and environmental features in the etiopathogenetic mechanisms underlying T2D and AD/PD. On this subject, this review will summarize the shared (epi)genomic features that characterize these complex pathologies. In particular, genetic variants and gene expression profiles associated with T2D and AD/PD will be discussed as possible contributors to determine the susceptibility and progression to these disorders. Moreover, potential shared epigenetic modifications and factors among T2D, AD and PD will also be illustrated. Overall, this review shows that findings from genomic studies still deserves further research to evaluate and identify genetic factors that directly contribute to the shared etiopathogenesis. Moreover, a common epigenetic background still needs to be investigated and characterized. The evidences discussed in this review underline the importance of integrating large-scale (epi)genomic data with additional molecular information and clinical and social background in order to finely dissect the complex etiopathogenic networks that build up the “disease interactome” characterizing T2D, AD and PD.
Collapse
Affiliation(s)
- Valerio Caputo
- Department of Biomedicine and Prevention, Tor Vergata University, Rome 00133, Italy
- Molecular Genetics Laboratory UILDM, Santa Lucia Foundation, Rome 00142, Italy
| | - Andrea Termine
- Molecular Genetics Laboratory UILDM, Santa Lucia Foundation, Rome 00142, Italy
- Experimental and Behavioral Neurophysiology Laboratory, Santa Lucia Foundation, Rome 00142, Italy
| | - Claudia Strafella
- Molecular Genetics Laboratory UILDM, Santa Lucia Foundation, Rome 00142, Italy
- Department of Biomedicine and Prevention, Tor Vergata University, Rome 00133, Italy
| | - Emiliano Giardina
- Molecular Genetics Laboratory UILDM, Santa Lucia Foundation, Rome 00142, Italy
- Department of Biomedicine and Prevention, Tor Vergata University, Rome 00133, Italy
| | - Raffaella Cascella
- Department of Biomedicine and Prevention, Tor Vergata University, Rome 00133, Italy
- Department of Biomedical Sciences, Catholic University Our Lady of Good Counsel, Tirana 1000, Albania
| |
Collapse
|
41
|
Park S, Zhang T, Wu X, Yi Qiu J. Ketone production by ketogenic diet and by intermittent fasting has different effects on the gut microbiota and disease progression in an Alzheimer's disease rat model. J Clin Biochem Nutr 2020; 67:188-198. [PMID: 33041517 PMCID: PMC7533860 DOI: 10.3164/jcbn.19-87] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/22/2019] [Indexed: 12/16/2022] Open
Abstract
The benefits of ketone production regimens remain controversial. Here, we hypothesized that the ketone-producing regimens modulated cognitive impairment, glucose metabolism, and inflammation while altering the gut microbiome. The hypothesis and the mechanism were explored in amyloid-β infused rats. Rats that received an amyloid-β(25–35) infusion into the hippocampus had either ketogenic diet (AD-KD), intermittent fasting (AD-IMF), 30 energy percent fat diet (AD-CON), or high carbohydrate (starch) diet (AD-CHO) for 8 weeks. AD-IMF and AD-CHO, but not AD-KD, lowered the hippocampal amyloid-β deposition compared to the AD-CON despite serum ketone concentrations being elevated in both AD-KD and AD-IMF. AD-IMF and AD-CHO, but not AD-KD, improved memory function in passive avoidance, Y maze, and water maze tests compared to the AD-CON. Hippocampal insulin signaling (pAkt→pGSK-3β) was potentiated and pTau was attenuated in AD-IMF and AD-CHO much more than AD-CON. AD-IMF and AD-CON had similar glucose tolerance results during OGTT, but AD-KD and AD-IMF exhibited glucose intolerance. AD-KD exacerbated gut dysbiosis by increasing Proteobacteria, and AD-CHO improved it by elevating Bacteriodetes. In conclusion, ketone production itself might not improve memory function, insulin resistance, neuroinflammation or the gut microbiome when induced by ketone-producing remedies. Intermittent fasting and a high carbohydrate diet containing high starch may be beneficial for people with dementia.
Collapse
Affiliation(s)
- Sunmin Park
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, 165 Sechul-Ri Baebang-Yup, Asan-si, Chungchungnam-Do, 336795, Korea
| | - Ting Zhang
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, 165 Sechul-Ri Baebang-Yup, Asan-si, Chungchungnam-Do, 336795, Korea
| | - Xuangao Wu
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, 165 Sechul-Ri Baebang-Yup, Asan-si, Chungchungnam-Do, 336795, Korea
| | - Jing Yi Qiu
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, 165 Sechul-Ri Baebang-Yup, Asan-si, Chungchungnam-Do, 336795, Korea
| |
Collapse
|
42
|
Gao W, Wang W, Zhang J, Deng P, Hu J, Yang J, Deng Z. Allicin ameliorates obesity comorbid depressive-like behaviors: involvement of the oxidative stress, mitochondrial function, autophagy, insulin resistance and NOX/Nrf2 imbalance in mice. Metab Brain Dis 2019; 34:1267-1280. [PMID: 31201726 DOI: 10.1007/s11011-019-00443-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 05/28/2019] [Indexed: 12/29/2022]
Abstract
The increased prevalence of obesity has been a major medical and public health problem in the past decades. In obese status, insulin resistance and sustained oxidative stress damage might give rise to behavioral deficits. The anti-obesity and anti-oxidant effects of allicin have been previously reported in peripheral tissues. In the present study, the functions and mechanisms of allicin involved in the prevention of high-fat diet (HFD)-induced depressive-like behaviors were investigated to better understand the pharmacological activities of allicin. Obese mice (five weeks of age) were treated with allicin (50, 100, and 200 mg/kg) by gavage for 15 weeks and behavioral test (sucrose preference, open field, and tail suspension) were performed. Furthermore, markers of oxidative stress, mitochondrial function, autophagy, and insulin resistance were measured in the hippocampal tissue. Finally, the levels of NADPH oxidase (NOX2, NOX4) and the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway were evaluated in the hippocampus. The body weight, metabolic disorders, and depressive-like behaviors in obese mice were ameliorated by allicin. The depressive-like behaviors presented in the obese mice were accompanied by remarkably excessive reactive oxygen species (ROS) production and oxidative stress, damaged mitochondrial function, imbalanced autophagy, and enhanced insulin resistance in the hippocampus. We found that allicin improved the above undesirable effects in the obese mice. Furthermore, allicin significantly decreased NOX2 and NOX4 levels and activated the Nrf2 pathway. Allicin attenuated depressive-like behaviors triggered by long-term HFD consumption by inhibiting ROS production and oxidative stress, improving mitochondrial function, regulating autophagy, and reducing insulin resistance in the hippocampus via optimization of NOX/Nrf2 imbalance.
Collapse
Affiliation(s)
- Wenqi Gao
- Department of Central Experimental Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, 443000, China
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University&Technology, Wuhan, Hubei, China
| | - Wei Wang
- Department of Central Experimental Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, 443000, China
| | - Jing Zhang
- Department of Central Experimental Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, 443000, China
| | - Pengyi Deng
- Department of Nuclear medicine, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, 443000, China
| | - Jun Hu
- Department of Central Experimental Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, 443000, China
| | - Jian Yang
- Department of Central Experimental Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, 443000, China.
| | - Zhifang Deng
- Department of Pharmacy, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, 443000, China.
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, China.
| |
Collapse
|
43
|
Brain signalling systems: A target for treating type I diabetes mellitus. Brain Res Bull 2019; 152:191-201. [PMID: 31325597 DOI: 10.1016/j.brainresbull.2019.07.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/08/2019] [Accepted: 07/15/2019] [Indexed: 01/26/2023]
Abstract
From early to later stages of Type I Diabetes Mellitus (TIDM), signalling molecules including brain indolamines and protein kinases are altered significantly, and that has been implicated in the Metabolic Disorders (MD) as well as impairment of retinal, renal, neuronal and cardiovascular systems. Considerable attention has been focused to the effects of diabetes on these signalling systems. However, the exact pathophysiological mechanisms of these signals are not completely understood in TIDM, but it is likely that hyperglycemia, acidosis, and insulin resistance play significant roles. Insulin maintains normal glycemic levels and it acts by binding to its receptor, so that it activates the receptor's tyrosine kinase activity, resulting in phosphorylation of several substrates. Those substrates provide binding/interaction sites for signalling molecules, including serine/threonine kinases and indolamines. For more than two decades, our research has been focused on the mechanisms of protein kinases, CaM Kinase and Serotonin transporter mediated alterations of indolamines in TIDM. In this review, we have also discussed how discrete areas of brain respond to insulin or some of the pharmacological agents that triggers or restores these signalling molecules, and it may be useful for the treatment of specific region wise changes/disorders of diabetic brain.
Collapse
|