1
|
Verma A, Goyal A. Beyond insulin: The Intriguing role of GLP-1 in Parkinson's disease. Eur J Pharmacol 2024; 982:176936. [PMID: 39182542 DOI: 10.1016/j.ejphar.2024.176936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/12/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
GLP-1 (Glucagon-like peptide 1) serves as both a peptide hormone and a growth factor, is released upon nutrient intake and contributes to insulin secretion stimulated by glucose levels. Also, GLP-1 is synthesized within several brain areas and plays a vital function in providing neuroprotection and reducing inflammation through the activation of the GLP-1 receptor. Parkinson's Disease (PD) is a neurodegenerative illness that worsens with time and is defined by considerable morbidity. Presently, there are few pharmaceutical choices available, and none of the existing therapies are capable of modifying the course of the disease. There is a suggestion that type 2 diabetes mellitus (T2DM) could increase the risk of PD, and the presence of both conditions concurrently might exacerbate PD symptoms and hasten neurodegeneration. GLP-1 receptor (GLP-1R) agonists exhibit numerous implications like enhancement of glucose-dependent insulin release and biosynthesis, suppression of glucagon secretion and gastric emptying. Also, some GLP-1R agonists have received clinical approval for the management of T2DM. Moreover, the use of GLP-1R agonists has demonstrated counter-inflammatory, neurotrophic, and neuroprotective actions in various preclinical models of neurodegenerative disorders. Considering the significant amount of evidence backing the potential of GLP-1R agonists to protect the nervous system across different research settings, this article delves into examining the hopeful prospect of GLP-1R agonists as a treatment option for PD. This review sheds light on combined neuroprotective benefits of GLP-1R agonists and the possible mechanisms driving the protective effects on the PD brain, through the collection of data from various preclinical and clinical investigations.
Collapse
Affiliation(s)
- Aanchal Verma
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| |
Collapse
|
2
|
Siddeeque N, Hussein MH, Abdelmaksoud A, Bishop J, Attia AS, Elshazli RM, Fawzy MS, Toraih EA. Neuroprotective effects of GLP-1 receptor agonists in neurodegenerative Disorders: A Large-Scale Propensity-Matched cohort study. Int Immunopharmacol 2024; 143:113537. [PMID: 39486172 DOI: 10.1016/j.intimp.2024.113537] [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: 09/04/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND GLP-1 receptor agonists, traditionally used for treating type 2 diabetes mellitus and obesity, have demonstrated anti-inflammatory properties. However, their potential neuroprotective effects in neurodegenerative disorders remain unclear. OBJECTIVE To evaluate the impact of GLP-1 receptor agonists on the risk of developing various neurodegenerative conditions in obese patients. METHODS This comprehensive retrospective cohort study analyzed data from 5,307,845 obese adult patients across 73 healthcare organizations in 17 countries. Propensity score matching was performed, resulting in 102,935 patients in each cohort. We compared the risk of developing neurodegenerative disorders between obese patients receiving GLP-1 receptor agonist therapy and those who were not. RESULTS Obese patients treated with GLP-1 receptor agonists showed significantly lower risks of developing Alzheimer's disease (RR = 0.627, 95 %CI = 0.481-0.817), Lewy body dementia (RR = 0.590, 95 %CI = 0.462-0.753), and vascular dementia (RR = 0.438, 95 %CI = 0.327-0.588). The risk reduction for Parkinson's disease was not statistically significant overall (RR = 0.784, 95 %CI = 0.580-1.058) but was significant for semaglutide users (RR = 0.574, 95 %CI = 0.369-0.893). Semaglutide consistently showed the most pronounced protective effects across all disorders. Additionally, a significant reduction in all-cause mortality was observed (HR = 0.525, 95 %CI = 0.493-0.558). CONCLUSION This study provides evidence that the effects of GLP-1 receptor agonists may extend beyond their known metabolic and cardioprotective benefits to include neuroprotection, associated with a decreased risk of developing various neurodegenerative disorders. These findings suggest the potential for expanding the therapeutic applications of GLP-1 receptor agonists to improve neurocognitive outcomes. Further research is warranted to elucidate the mechanisms underlying these neuroprotective effects and to explore their clinical applications in neurodegenerative disease prevention and treatment.
Collapse
Affiliation(s)
| | | | - Ahmed Abdelmaksoud
- Department of Internal Medicine, University of California, Riverside, CA 92521, USA
| | - Julia Bishop
- Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Abdallah S Attia
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Rami M Elshazli
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA; Biochemistry and Molecular Genetic Unit, Department of Basic Sciences, Faculty of Physical Therapy, Horus University - Egypt, New Damietta 34517, Egypt; Department of Biological Sciences, Faculty of Science, New Mansoura University, New Mansoura City 35742, Egypt
| | - Manal S Fawzy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia; Center for Health Research, Northern Border University, Arar 1321, Saudi Arabia
| | - Eman A Toraih
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA; Medical Genetics Unit, Department of Histology and Cell Biology, Suez Canal University, Ismailia 41522, Egypt.
| |
Collapse
|
3
|
Qi Y, Dong Y, Chen J, Xie S, Ma X, Yu X, Yu Y, Wang Y. Lactiplantibacillus plantarum SG5 inhibits neuroinflammation in MPTP-induced PD mice through GLP-1/PGC-1α pathway. Exp Neurol 2024; 383:115001. [PMID: 39406307 DOI: 10.1016/j.expneurol.2024.115001] [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: 08/04/2024] [Revised: 09/29/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Mounting evidence suggests that alterations in gut microbial composition play an active role in the pathogenesis of Parkinson's disease (PD). Probiotics are believed to modulate gut microbiota, potentially influencing PD development through the microbiota-gut-brain axis. However, the potential beneficial effects of Lactiplantibacillus plantarum SG5 (formerly known as Lactobacillus plantarum, abbreviated as L. plantarum) on PD and its underlying mechanisms remain unclear. In this study, we employed immunofluorescence, Western blotting, ELISA, and 16S rRNA gene sequencing to investigate the neuroprotective effects of L. plantarum SG5 against neuroinflammation in an MPTP-induced PD model and to explore the underlying mechanisms. Our results demonstrated that L. plantarum SG5 ameliorated MPTP-induced motor deficits, dopaminergic neuron loss, and elevated α-synuclein protein levels. Furthermore, SG5 inhibited MPTP-triggered overactivation of microglia and astrocytes in the substantia nigra (SN), attenuated disruption of both blood-brain and intestinal barriers, and suppressed the release of inflammatory factors in the colon and SN. Notably, SG5 modulated the composition and structure of the gut microbiota in mice. The MPTP-induced decrease in colonic GLP-1 secretion was reversed by SG5 treatment, accompanied by increased expression of GLP-1R and PGC-1α in the SN. Importantly, the GLP-1R antagonist Exendin 9-39 and PGC-1α inhibitor SR18292 attenuated the protective effects of SG5 in PD mice. In conclusion, we demonstrate a neuroprotective role of L. plantarum SG5 in the MPTP-induced PD mouse model, which likely involves modulation of the gut microbiota and, significantly, the GLP-1/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Yueyan Qi
- Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Hebei Key Laboratory of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
| | - Yuxuan Dong
- Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Hebei Key Laboratory of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
| | - Jinhu Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang 050051, China
| | - Siyou Xie
- Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Hebei Key Laboratory of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
| | - Xin Ma
- Thankcome Biotechnology (Su Zhou) Co., Suzhou, China
| | - Xueping Yu
- Thankcome Biotechnology (Su Zhou) Co., Suzhou, China
| | - Yang Yu
- Thankcome Biotechnology (Su Zhou) Co., Suzhou, China
| | - Yanqin Wang
- Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Hebei Key Laboratory of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China.
| |
Collapse
|
4
|
Yang S, Zhao X, Zhang Y, Tang Q, Li Y, Du Y, Yu P. Tirzepatide shows neuroprotective effects via regulating brain glucose metabolism in APP/PS1 mice. Peptides 2024; 179:171271. [PMID: 39002758 DOI: 10.1016/j.peptides.2024.171271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/22/2024] [Accepted: 07/09/2024] [Indexed: 07/15/2024]
Abstract
Tirzepatide (LY3298176), a GLP-1 and GIP receptor agonist, is fatty-acid-modified and 39-amino acid linear peptide, which ameliorates learning and memory impairment in diabetic rats. However, the specific molecular mechanism remains unknown. In the present study, we investigated the role of tirzepatide in the neuroprotective effects in Alzheimer's disease (AD) model mice. Tirzepatide was administrated intraperitoneal (i.p.) APP/PS1 mice for 8 weeks with at 10 nmol/kg once-weekly, it significantly decreased the levels of GLP-1R, and GFAP protein expression and amyloid plaques in the cortex, it also lowered neuronal apoptosis induced by amyloid-β (Aβ), but did not affect the anxiety and cognitive function in APP/PS1 mice. Moreover, tirzepatide reduced the blood glucose levels and increased the mRNA expression of GLP-1R, SACF1, ATF4, Glu2A, and Glu2B in the hypothalamus of APP/PS1 mice. Tirzepatide increased the mRNA expression of glucose transporter 1, hexokinase, glucose-6-phosphate dehydrogenase, and phosphofructokinase in the cortex. Lastly, tirzepatide improved the energetic metabolism by regulated reactive oxygen species production and mitochondrial membrane potential caused by Aβ, thereby decreasing mitochondrial function and ATP levels in astrocytes through GLP-1R. These results provide valuable insights into the mechanism of brain glucose metabolism and mitochondrial function of tirzepatide, presenting potential strategies for AD treatment.
Collapse
Affiliation(s)
- Shaobin Yang
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, China.
| | - Xiaoqian Zhao
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, China
| | - Yimeng Zhang
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, China
| | - Qi Tang
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, China
| | - Yanhong Li
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, China
| | - Yaqin Du
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, China
| | - Peng Yu
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, China
| |
Collapse
|
5
|
de Paiva IHR, da Silva RS, Mendonça IP, de Souza JRB, Peixoto CA. Semaglutide Attenuates Anxious and Depressive-Like Behaviors and Reverses the Cognitive Impairment in a Type 2 Diabetes Mellitus Mouse Model Via the Microbiota-Gut-Brain Axis. J Neuroimmune Pharmacol 2024; 19:36. [PMID: 39042202 DOI: 10.1007/s11481-024-10142-w] [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: 08/09/2023] [Accepted: 07/14/2024] [Indexed: 07/24/2024]
Abstract
Newly conducted research suggests that metabolic disorders, like diabetes and obesity, play a significant role as risk factors for psychiatric disorders. This connection presents a potential avenue for creating novel antidepressant medications by repurposing drugs originally developed to address antidiabetic conditions. Earlier investigations have shown that GLP-1 (Glucagon-like Peptide-1) analogs exhibit neuroprotective qualities in various models of neurological diseases, encompassing conditions such as Alzheimer's disease, Parkinson's disease, and stroke. Moreover, GLP-1 analogs have demonstrated the capability to enhance neurogenesis, a process recognized for its significance in memory formation and the cognitive and emotional aspects of information processing. Nonetheless, whether semaglutide holds efficacy as both an antidepressant and anxiolytic agent remains uncertain. To address this, our study focused on a mouse model of depression linked to type 2 diabetes induced by a High Fat Diet (HFD). In this model, we administered semaglutide (0.05 mg/Kg intraperitoneally) on a weekly basis to evaluate its potential as a therapeutic option for depression and anxiety. Diabetic mice had higher blood glucose, lipidic profile, and insulin resistance. Moreover, mice fed HFD showed higher serum interleukin (IL)-1β and lipopolysaccharide (LPS) associated with impaired humor and cognition. The analysis of behavioral responses revealed that the administration of semaglutide effectively mitigated depressive- and anxiety-like behaviors, concurrently demonstrating an enhancement in cognitive function. Additionally, semaglutide treatment protected synaptic plasticity and reversed the hippocampal neuroinflammation induced by HFD fed, improving activation of the insulin pathway, demonstrating the protective effects of semaglutide. We also found that semaglutide treatment decreased astrogliosis and microgliosis in the dentate gyrus region of the hippocampus. In addition, semaglutide prevented the DM2-induced impairments of pro-opiomelanocortin (POMC), and G-protein-coupled receptor 43 (GPR43) and simultaneously increased the NeuN + and Glucagon-like Peptide-1 receptor (GLP-1R+) neurons in the hippocampus. Our data also showed that semaglutide increased the serotonin (5-HT) and serotonin transporter (5-HTT) and glutamatergic receptors in the hippocampus. At last, semaglutide changed the gut microbiota profile (increasing Bacterioidetes, Bacteroides acidifaciens, and Blautia coccoides) and decreased leaky gut, improving the gut-brain axis. Taken together, semaglutide has the potential to act as a therapeutic tool for depression and anxiety.
Collapse
MESH Headings
- Animals
- Glucagon-Like Peptides/pharmacology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/psychology
- Diabetes Mellitus, Type 2/metabolism
- Mice
- Cognitive Dysfunction/drug therapy
- Cognitive Dysfunction/prevention & control
- Cognitive Dysfunction/etiology
- Cognitive Dysfunction/metabolism
- Depression/drug therapy
- Depression/psychology
- Depression/metabolism
- Male
- Anxiety/drug therapy
- Anxiety/psychology
- Anxiety/etiology
- Gastrointestinal Microbiome/drug effects
- Mice, Inbred C57BL
- Brain-Gut Axis/drug effects
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/psychology
- Diabetes Mellitus, Experimental/metabolism
- Disease Models, Animal
- Antidepressive Agents/pharmacology
- Antidepressive Agents/therapeutic use
Collapse
Affiliation(s)
- Igor Henrique Rodrigues de Paiva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Av. Moraes Rego s/n, Recife CEP, PE, 50670-420, Brazil.
- Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, PE, Brazil.
| | - Rodrigo Soares da Silva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Av. Moraes Rego s/n, Recife CEP, PE, 50670-420, Brazil
- Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | - Ingrid Prata Mendonça
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Av. Moraes Rego s/n, Recife CEP, PE, 50670-420, Brazil
- Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | | | - Christina Alves Peixoto
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Av. Moraes Rego s/n, Recife CEP, PE, 50670-420, Brazil.
- Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Recife, Brazil.
| |
Collapse
|
6
|
Lemche E, Killick R, Mitchell J, Caton PW, Choudhary P, Howard JK. Molecular mechanisms linking type 2 diabetes mellitus and late-onset Alzheimer's disease: A systematic review and qualitative meta-analysis. Neurobiol Dis 2024; 196:106485. [PMID: 38643861 DOI: 10.1016/j.nbd.2024.106485] [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: 06/30/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/23/2024] Open
Abstract
Research evidence indicating common metabolic mechanisms through which type 2 diabetes mellitus (T2DM) increases risk of late-onset Alzheimer's dementia (LOAD) has accumulated over recent decades. The aim of this systematic review is to provide a comprehensive review of common mechanisms, which have hitherto been discussed in separate perspectives, and to assemble and evaluate candidate loci and epigenetic modifications contributing to polygenic risk linkages between T2DM and LOAD. For the systematic review on pathophysiological mechanisms, both human and animal studies up to December 2023 are included. For the qualitative meta-analysis of genomic bases, human association studies were examined; for epigenetic mechanisms, data from human studies and animal models were accepted. Papers describing pathophysiological studies were identified in databases, and further literature gathered from cited work. For genomic and epigenomic studies, literature mining was conducted by formalised search codes using Boolean operators in search engines, and augmented by GeneRif citations in Entrez Gene, and other sources (WikiGenes, etc.). For the systematic review of pathophysiological mechanisms, 923 publications were evaluated, and 138 gene loci extracted for testing candidate risk linkages. 3 57 publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight insulin signalling, inflammation and inflammasome pathways, proteolysis, gluconeogenesis and glycolysis, glycosylation, lipoprotein metabolism and oxidation, cell cycle regulation or survival, autophagic-lysosomal pathways, and energy. Documented findings suggest interplay between brain insulin resistance, neuroinflammation, insult compensatory mechanisms, and peripheral metabolic dysregulation in T2DM and LOAD linkage. The results allow for more streamlined longitudinal studies of T2DM-LOAD risk linkages.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom.
| | - Richard Killick
- Section of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom
| | - Jackie Mitchell
- Department of Basic and Clinical Neurosciences, Maurice Wohl CIinical Neurosciences Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, United Kingdom
| | - Paul W Caton
- Diabetes Research Group, School of Life Course Sciences, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL, United Kingdom
| | - Pratik Choudhary
- Diabetes Research Group, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, Hodgkin Building, Guy's Campus, King's College London, Great Maze Pond, London SE1 1UL, United Kingdom
| |
Collapse
|
7
|
Canário NS, Crisóstomo J, Moreno C, Duarte JV, Duarte IC, Ribeiro MJ, Caramelo B, Gomes LV, Matafome P, Oliveira FP, Castelo-Branco M. Functional reorganization of memory processing in the hippocampus is associated with neuroprotector GLP-1 levels in type 2 diabetes. Heliyon 2024; 10:e27412. [PMID: 38509913 PMCID: PMC10950584 DOI: 10.1016/j.heliyon.2024.e27412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/22/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Type 2 diabetes (T2D) often impairs memory functions, suggesting specific vulnerability of the hippocampus. In vivo neuroimaging studies relating encoding and retrieval of memory information with endogenous neuroprotection are lacking. The neuroprotector glucagon-like peptide (GLP-1) has a high receptor density in anterior/ventral hippocampus, as shown by animal models. Using an innovative event-related fMRI design in 34 participants we investigated patterns of hippocampal activity in T2D (n = 17) without mild cognitive impairment (MCI) versus healthy controls (n = 17) during an episodic memory task. We directly measured neurovascular coupling by estimating the hemodynamic response function using event-related analysis related to encoding and retrieval of episodic information in the hippocampus. We applied a mixed-effects general linear model analysis and a two-factor ANOVA to test for group differences. Significant between-group differences were found for memory encoding, showing evidence for functional reorganization: T2D patients showed an augmented activation in the posterior hippocampus while anterior activation was reduced. The latter was negatively correlated with both GLP-1 pre- and post-breakfast levels, in the absence of grey matter changes. These results suggest that patients with T2D without MCI have pre-symptomatic functional reorganization in brain regions underlying episodic memory, as a function of the concentration of the neuroprotective neuropeptide GLP-1.
Collapse
Affiliation(s)
- Nádia S. Canário
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health, Portugal
- Faculty of Medicine, University of Coimbra, Portugal
| | - Joana Crisóstomo
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health, Portugal
| | - Carolina Moreno
- Department of Endocrinology, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - João V. Duarte
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health, Portugal
- Faculty of Medicine, University of Coimbra, Portugal
| | - Isabel C. Duarte
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health, Portugal
| | - Mário J. Ribeiro
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health, Portugal
- The Faculty of Science and Technology, University of Coimbra, Portugal
| | - Beatriz Caramelo
- Faculty of Medicine, University of Coimbra, Portugal
- Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center of Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | - Leonor V. Gomes
- Department of Endocrinology, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - Paulo Matafome
- Faculty of Medicine, University of Coimbra, Portugal
- Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center of Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | | | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health, Portugal
- Faculty of Medicine, University of Coimbra, Portugal
| |
Collapse
|
8
|
Verma A, Chaudhary S, Solanki K, Goyal A, Yadav HN. Exendin-4: A potential therapeutic strategy for Alzheimer's disease and Parkinson's disease. Chem Biol Drug Des 2024; 103:e14426. [PMID: 38230775 DOI: 10.1111/cbdd.14426] [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: 10/06/2023] [Revised: 11/16/2023] [Accepted: 12/05/2023] [Indexed: 01/18/2024]
Abstract
Neurodegenerative disorders, which affect millions worldwide, are marked by a steady decline of neurons that are selectively susceptible. Due to the complex pathological processes underlying neurodegeneration, at present, there is no viable therapy available for neurodegenerative disorders. Consequently, the establishment of a novel therapeutic approach for such conditions is a clinical void that remains. The potential significance of various peptides as neuroprotective interventions for neurodegenerative disorders is gaining increasing attention. In the past few years, there has been growing scientific interest in glucagon-like peptide-1 receptor agonists due to their claimed neuroprotective effects. Exendin-4 is a glucagon-like peptide-1 receptor agonist that is known to possess anti-diabetic effects and does not degrade for hours, making it a superior candidate for such disorders. Moreover, exendin-4's neuroprotective effects have been reported in several preclinical studies. Exendin-4's diverse therapeutic targets suggest its potential therapeutic uses in neurodegenerative ailments like Alzheimer's disease and Parkinson's disease and have garnered an increasing amount of attention. Given the substantial body of evidence supporting the neuroprotective potential of exendin-4 in various research models, this article is dedicated to exploring the promising role of exendin-4 as a therapeutic agent for the treatment and management of Alzheimer's disease and Parkinson's disease. This review draws insights from the findings of numerous preclinical and clinical studies to highlight the collective neuroprotective advantages of exendin-4 and the potential mechanisms that underlie its neuroprotective effects.
Collapse
Affiliation(s)
- Aanchal Verma
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Shobhit Chaudhary
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Kunal Solanki
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | | |
Collapse
|
9
|
Nowell J, Blunt E, Gupta D, Edison P. Antidiabetic agents as a novel treatment for Alzheimer's and Parkinson's disease. Ageing Res Rev 2023; 89:101979. [PMID: 37328112 DOI: 10.1016/j.arr.2023.101979] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/18/2023]
Abstract
Therapeutic strategies for neurodegenerative disorders have commonly targeted individual aspects of the disease pathogenesis to little success. Neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD), are characterized by several pathological features. In AD and PD, there is an abnormal accumulation of toxic proteins, increased inflammation, decreased synaptic function, neuronal loss, increased astrocyte activation, and perhaps a state of insulin resistance. Epidemiological evidence has revealed a link between AD/PD and type 2 diabetes mellitus, with these disorders sharing some pathological commonalities. Such a link has opened up a promising avenue for repurposing antidiabetic agents in the treatment of neurodegenerative disorders. A successful therapeutic strategy for AD/PD would likely require a single or several agents which target the separate pathological processes in the disease. Targeting cerebral insulin signalling produces numerous neuroprotective effects in preclinical AD/PD brain models. Clinical trials have shown the promise of approved diabetic compounds in improving motor symptoms of PD and preventing neurodegenerative decline, with numerous further phase II trials and phase III trials underway in AD and PD populations. Alongside insulin signalling, targeting incretin receptors in the brain represents one of the most promising strategies for repurposing currently available agents for the treatment of AD/PD. Most notably, glucagon-like-peptide-1 (GLP-1) receptor agonists have displayed impressive clinical potential in preclinical and early clinical studies. In AD the GLP-1 receptor agonist, liraglutide, has been demonstrated to improve cerebral glucose metabolism and functional connectivity in small-scale pilot trials. Whilst in PD, the GLP-1 receptor agonist exenatide is effective in restoring motor function and cognition. Targeting brain incretin receptors reduces inflammation, inhibits apoptosis, prevents toxic protein aggregation, enhances long-term potentiation and autophagy as well as restores dysfunctional insulin signalling. Support is also increasing for the use of additional approved diabetic treatments, including intranasal insulin, metformin hydrochloride, peroxisome proliferator-activated nuclear receptor γ agonists, amylin analogs, and protein tyrosine phosphatase 1B inhibitors which are in the investigation for deployment in PD and AD treatment. As such, we provide a comprehensive review of several promising anti-diabetic agents for the treatment of AD and PD.
Collapse
Affiliation(s)
- Joseph Nowell
- Department of Brain Sciences, Imperial College London, London, UK
| | - Eleanor Blunt
- Department of Brain Sciences, Imperial College London, London, UK
| | - Dhruv Gupta
- Department of Brain Sciences, Imperial College London, London, UK
| | - Paul Edison
- Department of Brain Sciences, Imperial College London, London, UK; School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK.
| |
Collapse
|
10
|
Hardonova M, Siarnik P, Sivakova M, Sucha B, Penesova A, Radikova Z, Havranova A, Imrich R, Vlcek M, Zitnanova I, Krastev G, Kiacikova M, Kollar B, Turcani P. Endothelial Function in Patients with Multiple Sclerosis: The Role of GLP-1 Agonists, Lipoprotein Subfractions, and Redox Balance. Int J Mol Sci 2023; 24:11162. [PMID: 37446338 DOI: 10.3390/ijms241311162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
INTRODUCTION Epidemiological studies have suggested an increased vascular risk in patients with multiple sclerosis (MS). There is increasing evidence of the beneficial effects of GLP-1 agonists (GLP-1a) in preventing vascular complications and slowing the progression of neurodegeneration. Our objective was to explore the changes in the endothelial function of MS patients after 12 months of GLP-1a therapy. We also explored the role of lipoprotein subfractions and the antioxidant capacity of plasma. METHODS MS patients were enrolled in a prospective, unicentric study. GLP-1a (dulaglutide) was administered to 13 patients. The control population consisted of 12 subjects. Endothelial function was determined by peripheral arterial tonometry and expressed as reperfusion hyperemia index (RHI). Trolox equivalent antioxidant capacity (TEAC) was used to assess the total antioxidant capacity of the plasma. The levels of lipoprotein subfractions were evaluated. RESULTS The GLP-1a group did not have a significant change in their RHIs after 12 months (2.1 ± 0.6 vs. 2.1 ± 0.7; p = 0.807). However, a significant increase in their TEACs was observed (4.1 ± 1.4 vs. 5.2 ± 0.5 mmol/L, p = 0.010). On the contrary, the subjects in the control group had a significant worsening of their RHIs (2.1 ± 0.5 vs. 1.8 ± 0.6; p = 0.030), without significant changes in their TEACs. Except for a significant decrease in very-low-density lipoprotein (VLDL) (30.8 ± 10.2 vs. 22.6 ± 8.3 mg/dL, p = 0.043), no other significant changes in the variables were observed in the control group. VLDL levels (beta = -0.637, p = 0.001), the use of GLP-1a therapy (beta = 0.560, p = 0.003), and small LDL (beta = 0.339, p = 0.043) were the only significant variables in the model that predicted the follow-up RHI. CONCLUSION Our results suggest that the application of additional GLP-1a therapy may have atheroprotective and antioxidant effects in MS patients with high MS activity and thus may prospectively mitigate their vascular risk. However, the lipoprotein profile may also play an important role in the atherogenic risk of MS subjects.
Collapse
Affiliation(s)
- Miroslava Hardonova
- 1st Department of Neurology, Faculty of Medicine, Comenius University, 813 69 Bratislava, Slovakia
| | - Pavel Siarnik
- 1st Department of Neurology, Faculty of Medicine, Comenius University, 813 69 Bratislava, Slovakia
| | - Monika Sivakova
- 1st Department of Neurology, Faculty of Medicine, Comenius University, 813 69 Bratislava, Slovakia
| | - Bianka Sucha
- 1st Department of Neurology, Faculty of Medicine, Comenius University, 813 69 Bratislava, Slovakia
| | - Adela Penesova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Zofia Radikova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Andrea Havranova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Richard Imrich
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Miroslav Vlcek
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Ingrid Zitnanova
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Georgi Krastev
- Department of Neurology, Faculty Hospital, 917 75 Trnava, Slovakia
| | - Maria Kiacikova
- Department of Neurology, Faculty Hospital, 911 01 Trencin, Slovakia
| | - Branislav Kollar
- 1st Department of Neurology, Faculty of Medicine, Comenius University, 813 69 Bratislava, Slovakia
| | - Peter Turcani
- 1st Department of Neurology, Faculty of Medicine, Comenius University, 813 69 Bratislava, Slovakia
| |
Collapse
|
11
|
Cooper DH, Ramachandra R, Ceban F, Di Vincenzo JD, Rhee TG, Mansur RB, Teopiz KM, Gill H, Ho R, Cao B, Lui LMW, Jawad MY, Arsenault J, Le GH, Ramachandra D, Guo Z, McIntyre RS. Glucagon-like peptide 1 (GLP-1) receptor agonists as a protective factor for incident depression in patients with diabetes mellitus: A systematic review. J Psychiatr Res 2023; 164:80-89. [PMID: 37331261 DOI: 10.1016/j.jpsychires.2023.05.041] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/20/2023] [Accepted: 05/12/2023] [Indexed: 06/20/2023]
Abstract
Glucagon-like peptide 1 (GLP-1) receptor agonists are widely used for glycemic control in patients with diabetes mellitus (DM) and are primarily indicated for type 2 diabetes mellitus (T2DM). GLP-1 receptor agonists have also been shown to have neuroprotective and antidepressant properties. Replicated evidence suggests that individuals with DM are significantly more likely to develop depression. Herein, we aim to investigate whether GLP-1 receptor agonists can be used prophylactically on patients with DM to lower the risk of incident depression. We conducted a systematic search for English-language articles published on the PubMed/MEDLINE, Scopus, Embase, APA, PsycInfo, Ovid and Google Scholar databases from inception to June 6, 2022. Four retrospective observational studies were identified that evaluated the neuroprotective effects of GLP-1 receptor agonists on incident depression in patients with DM. We found mixed results with regards to lowering the risk of incident depression, with two studies demonstrating a significant reduction in risk and two studies showing no such effect. A single study found that dulaglutide may lower susceptibility to depression. Our results were limited by high interstudy heterogeneity, paucity of literature, and lack of controlled trials. While we did not find evidence of GLP-1 receptor agonists significantly lowering risk of incident depression in patients with DM, promising neuroprotective data presented in two of the included papers, specifically on dulaglutide where information is scarce, provide the impetus for further investigation. Future research should focus on better elucidating the neuroprotective potential of different classes and doses of GLP-1 receptor agonists using controlled trials.
Collapse
Affiliation(s)
- Daniel H Cooper
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada.
| | - Ranuk Ramachandra
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada.
| | - Felicia Ceban
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada.
| | - Joshua D Di Vincenzo
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada.
| | - Taeho Greg Rhee
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA; VA New England Mental Illness, Research, Education and Clinical Center (MIRECC), VA Connecticut Healthcare System, West Haven, CT, USA; Department of Public Health Sciences, School of Medicine, University of Connecticut, Farmington, CT, USA.
| | - Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, ON, Canada.
| | - Kayla M Teopiz
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada.
| | - Hartej Gill
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, ON, Canada.
| | - Roger Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, Singapore.
| | - Bing Cao
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, 400715, PR China.
| | - Leanna M W Lui
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, ON, Canada.
| | | | - Juliet Arsenault
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada.
| | - Gia Han Le
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada.
| | - Diluk Ramachandra
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada.
| | - Ziji Guo
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
12
|
Longo M, Di Meo I, Caruso P, Francesca Muscio M, Scappaticcio L, Maio A, Ida Maiorino M, Bellastella G, Signoriello G, Knop FK, Rosaria Rizzo M, Esposito K. Circulating levels of endothelial progenitor cells are associated with better cognitive function in older adults with glucagon-like peptide 1 receptor agonist-treated type 2 diabetes. Diabetes Res Clin Pract 2023; 200:110688. [PMID: 37116797 DOI: 10.1016/j.diabres.2023.110688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 04/30/2023]
Abstract
AIMS To evaluate cognitive function in subjects with type 2 diabetes (T2D) treated with glucagon-like peptide 1 receptor agonist (GLP-1RA) plus metformin or metformin alone and its association with endothelial progenitor cells (EPCs). METHODS Adults with T2D treated with GLP-1RA plus metformin (GLP-1RA + MET) or MET alone for at least 12 months were included. Montreal Cognitive Assessment test (MoCA), Mini-Mental State Examination (MMSE), Mini Nutritional Assessment (MNA) and disability tests were administered. Circulating levels of seven EPCs phenotypes were measured by flow cytometry. RESULTS A total of 154 elderly patients were included, of whom 78 in GLP-1RA + MET group and 76 in MET group. The GLP-1RA + MET group showed better cognitive function as indicated by a significant higher MoCA and MMSE scores, and higher levels of CD34+ CD133+, CD133+ KDR+, and CD34+ CD133+ KDR+ as compared with MET group. The number of CD34+ CD133+ KDR+ cells was an independent predictor of higher MoCA, MMSE and MNA scores. CONCLUSIONS People with T2D on GLP-1RA + MET treatment had better cognitive function and higher circulating levels of EPCs as compared with those on MET alone warranting further studies to understand the interrelationship between EPCs, GLP-RA treatment and cognitive health.
Collapse
Affiliation(s)
- Miriam Longo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Division of Endocrinology and Metabolic Diseases, AOU University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Irene Di Meo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Division of Geriatrics and Internal Medicine, AOU University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paola Caruso
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Division of Endocrinology and Metabolic Diseases, AOU University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Francesca Muscio
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Division of Geriatrics and Internal Medicine, AOU University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lorenzo Scappaticcio
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Division of Endocrinology and Metabolic Diseases, AOU University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonietta Maio
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Division of Endocrinology and Metabolic Diseases, AOU University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Ida Maiorino
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Division of Endocrinology and Metabolic Diseases, AOU University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe Bellastella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Division of Endocrinology and Metabolic Diseases, AOU University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe Signoriello
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark, Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria Rosaria Rizzo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Division of Geriatrics and Internal Medicine, AOU University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Katherine Esposito
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Division of Endocrinology and Metabolic Diseases, AOU University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
13
|
Lin MH, Cheng PC, Hsiao PJ, Chen SC, Hung CH, Kuo CH, Huang SK, Clair Chiou HY. The GLP-1 receptor agonist exenatide ameliorates neuroinflammation, locomotor activity, and anxiety-like behavior in mice with diet-induced obesity through the modulation of microglial M2 polarization and downregulation of SR-A4. Int Immunopharmacol 2023; 115:109653. [PMID: 36587502 DOI: 10.1016/j.intimp.2022.109653] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/16/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
Obesity is associated with multiple comorbidities, such as metabolic abnormalities and cognitive dysfunction. Moreover, accumulating evidence indicates that neurodegenerative disorders are associated with chronic neuroinflammation. GLP-1 receptor agonists (RAs) have been extensively studied as a treatment for type 2 diabetes. Emerging evidence has demonstrated a protective effect of GLP-1 RAs on neurodegenerative disease, which is independent of its glucose-lowering effects. In this study, we aimed to examine the effects of a long-acting GLP-1 RA, exenatide, on high-fat diet (HFD)-induced neuroinflammation and related brain function impairment. First, mice treated with exenatide exhibited significantly reduced HFD-increased body weight and blood glucose. In an open field test, exenatide treatment ameliorated the reduction in local motor activity and anxiety in HFD-fed mice. Moreover, HFD induced astrogliosis, microgliosis, and upregulation of IL-1β, IL-6 and TNF-α in hippocampus and cortex. Exenatide treatment reduced HFD-induced astrogliosis and IL-1β and TNF-α expressions. Moreover, exenatide increased phosphor-ERK and M2-type microglia marker arginase-1 expression in the hippocampus and cortex. In addition, we found that scavenger receptor-A4 protein expression was induced by HFD and was subsequently inhibited by exenatide. SR-A4 knockout reversed the locomotor activity impairment but not the anxiety behavior caused by HFD consumption. SR-A4 knockout also reduced HFD-induced neuroinflammation, as shown by the reduced expression of GFAP and IBA-1 compared with that in wild-type control mice. These results demonstrate that exenatide decreases HFD-increased neuroinflammation and promotes anti-inflammatory M2 differentiation. The inhibition of SR-A4 by exenatide exerts anti-inflammatory activity.
Collapse
Affiliation(s)
- Ming-Hong Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; M.Sc. Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Po-Ching Cheng
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Center for International Tropical Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Pi-Jung Hsiao
- Division of Endocrinology and Metabolism, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan.
| | - Szu-Chia Chen
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Chih-Hsing Hung
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Pediatrics, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Chao-Hung Kuo
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Shau-Ku Huang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli County 350, Taiwan; Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Hsin-Ying Clair Chiou
- Center of Teaching and Research, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan; Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan.
| |
Collapse
|
14
|
Woodfield A, Gonzales T, Helmerhorst E, Laws S, Newsholme P, Porter T, Verdile G. Current Insights on the Use of Insulin and the Potential Use of Insulin Mimetics in Targeting Insulin Signalling in Alzheimer's Disease. Int J Mol Sci 2022; 23:15811. [PMID: 36555450 PMCID: PMC9779379 DOI: 10.3390/ijms232415811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/10/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) and type 2 diabetes (T2D) are chronic diseases that share several pathological mechanisms, including insulin resistance and impaired insulin signalling. Their shared features have prompted the evaluation of the drugs used to manage diabetes for the treatment of AD. Insulin delivery itself has been utilized, with promising effects, in improving cognition and reducing AD related neuropathology. The most recent clinical trial involving intranasal insulin reported no slowing of cognitive decline; however, several factors may have impacted the trial outcomes. Long-acting and rapid-acting insulin analogues have also been evaluated within the context of AD with a lack of consistent outcomes. This narrative review provided insight into how targeting insulin signalling in the brain has potential as a therapeutic target for AD and provided a detailed update on the efficacy of insulin, its analogues and the outcomes of human clinical trials. We also discussed the current evidence that warrants the further investigation of the use of the mimetics of insulin for AD. These small molecules may provide a modifiable alternative to insulin, aiding in developing drugs that selectively target insulin signalling in the brain with the aim to attenuate cognitive dysfunction and AD pathologies.
Collapse
Affiliation(s)
- Amy Woodfield
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Australia
| | - Tatiana Gonzales
- Curtin Medical School, Curtin University, Bentley 6102, Australia
| | - Erik Helmerhorst
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Australia
| | - Simon Laws
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup 6027, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup 6027, Australia
| | - Philip Newsholme
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Australia
| | - Tenielle Porter
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup 6027, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup 6027, Australia
| | - Giuseppe Verdile
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup 6027, Australia
| |
Collapse
|
15
|
Reich N, Hölscher C. The neuroprotective effects of glucagon-like peptide 1 in Alzheimer's and Parkinson's disease: An in-depth review. Front Neurosci 2022; 16:970925. [PMID: 36117625 PMCID: PMC9475012 DOI: 10.3389/fnins.2022.970925] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/08/2022] [Indexed: 12/16/2022] Open
Abstract
Currently, there is no disease-modifying treatment available for Alzheimer's and Parkinson's disease (AD and PD) and that includes the highly controversial approval of the Aβ-targeting antibody aducanumab for the treatment of AD. Hence, there is still an unmet need for a neuroprotective drug treatment in both AD and PD. Type 2 diabetes is a risk factor for both AD and PD. Glucagon-like peptide 1 (GLP-1) is a peptide hormone and growth factor that has shown neuroprotective effects in preclinical studies, and the success of GLP-1 mimetics in phase II clinical trials in AD and PD has raised new hope. GLP-1 mimetics are currently on the market as treatments for type 2 diabetes. GLP-1 analogs are safe, well tolerated, resistant to desensitization and well characterized in the clinic. Herein, we review the existing evidence and illustrate the neuroprotective pathways that are induced following GLP-1R activation in neurons, microglia and astrocytes. The latter include synaptic protection, improvements in cognition, learning and motor function, amyloid pathology-ameliorating properties (Aβ, Tau, and α-synuclein), the suppression of Ca2+ deregulation and ER stress, potent anti-inflammatory effects, the blockage of oxidative stress, mitochondrial dysfunction and apoptosis pathways, enhancements in the neuronal insulin sensitivity and energy metabolism, functional improvements in autophagy and mitophagy, elevated BDNF and glial cell line-derived neurotrophic factor (GDNF) synthesis as well as neurogenesis. The many beneficial features of GLP-1R and GLP-1/GIPR dual agonists encourage the development of novel drug treatments for AD and PD.
Collapse
Affiliation(s)
- Niklas Reich
- Biomedical and Life Sciences Division, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| | - Christian Hölscher
- Neurology Department, Second Associated Hospital, Shanxi Medical University, Taiyuan, China
- Henan University of Chinese Medicine, Academy of Chinese Medical Science, Zhengzhou, China
| |
Collapse
|
16
|
Anti-Inflammatory Effects of GLP-1 Receptor Activation in the Brain in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23179583. [PMID: 36076972 PMCID: PMC9455625 DOI: 10.3390/ijms23179583] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/17/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
The glucagon-like peptide-1 (GLP-1) is a pleiotropic hormone well known for its incretin effect in the glucose-dependent stimulation of insulin secretion. However, GLP-1 is also produced in the brain and displays a critical role in neuroprotection and inflammation by activating the GLP-1 receptor signaling pathways. Several studies in vivo and in vitro using preclinical models of neurodegenerative diseases show that GLP-1R activation has anti-inflammatory properties. This review explores the molecular mechanistic action of GLP-1 RAS in relation to inflammation in the brain. These findings update our knowledge of the potential benefits of GLP-1RAS actions in reducing the inflammatory response. These molecules emerge as a potential therapeutic tool in treating neurodegenerative diseases and neuroinflammatory pathologies.
Collapse
|
17
|
Ölmestig J, Marlet IR, Vilsbøll T, Rungby J, Rostrup E, Lambertsen KL, Kruuse C. A single dose of exenatide had no effect on blood flow velocity in the middle cerebral artery in elderly healthy volunteers: Randomized, placebo-controlled, double-blind clinical trial. Front Aging Neurosci 2022; 14:899389. [PMID: 36636739 PMCID: PMC9831269 DOI: 10.3389/fnagi.2022.899389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/04/2022] [Indexed: 01/26/2023] Open
Abstract
Background and aims Glucagon-like peptide 1 (GLP-1) receptor agonists (GLP-1RA) are widely used for the treatment of type 2 diabetes, and recent studies indicate that they may be cardio- and neuroprotective. The safety and effect of a single dose of exenatide, a short-acting GLP-1RA, on cerebral and peripheral arterial function remain unknown. Methods In this randomized, double-blind pilot trial, we assigned elderly healthy volunteers without diabetes and no previous history of stroke to receive a single dose of subcutaneous exenatide (5 μg) or placebo. Primary outcome was immediate changes over time in blood flow velocity of the middle cerebral arteries (VMCA) assessed by repeated transcranial Doppler measurements. Secondary outcomes were changes in peripheral arterial function with finger plethysmography, ankle-brachial index (ABI), and inflammatory- and endothelial-specific biomarkers. Results Healthy volunteers (13 women and 17 men) were included: (mean ± standard deviation) age: 62 ± 8 years; body weight: 79.6 ± 12.7 kg; VMCA: 65.3 ± 10.7 cm/s; fasting plasma glucose: 5.5 ± 0.5 mmol/L; HbA1c: 33.9 ± 4.1 mmol/mol (5.3 ± 0.38%). No differences between exenatide and placebo group were seen regarding VMCA (p = 0.058), systolic ABI (p = 0.71), plethysmography (p = 0.45), tumor necrosis factor (p = 0.33), interleukin-6 (p = 0.11), interleukin-1β (p = 0.34), vascular cell adhesion molecule 1 (p = 0.73), intercellular adhesion molecule 1 (p = 0.74), or E-selectin (p = 0.31). No severe adverse events were observed. Conclusion A single dose of exenatide did not change cerebral blood flow velocity or peripheral vessel function in elderly healthy volunteers. The medication was safe to use in persons without diabetes allowing us to investigate this drug further in search of the neuroprotective mechanisms. Clinical Trial Registration https://clinicaltrials.gov, Identifier NCT02838589.
Collapse
Affiliation(s)
- Joakim Ölmestig
- Neurovascular Research Unit, Department of Neurology, Copenhagen University Hospital – Herlev and Gentofte, Copenhagen, Denmark,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ida R. Marlet
- Neurovascular Research Unit, Department of Neurology, Copenhagen University Hospital – Herlev and Gentofte, Copenhagen, Denmark
| | - Tina Vilsbøll
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark,Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | - Jørgen Rungby
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark,Department of Endocrinology, Copenhagen University Hospital – Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Egill Rostrup
- Center for Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital – Mental Health Center Glostrup, Copenhagen, Denmark
| | - Kate L. Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark,Department of Neurology, Odense University Hospital, Odense, Denmark,BRIDGE – Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Christina Kruuse
- Neurovascular Research Unit, Department of Neurology, Copenhagen University Hospital – Herlev and Gentofte, Copenhagen, Denmark,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark,*Correspondence: Christina Kruuse,
| |
Collapse
|
18
|
Grigoryan GA. Neuroinflammation and Reconsolidation of Memory. NEUROCHEM J+ 2022. [DOI: 10.1134/s1819712422020076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Zhu C, Hong T, Li H, Jiang S, Guo B, Wang L, Ding J, Gao C, Sun Y, Sun T, Wang F, Wang Y, Wan D. Glucagon-Like Peptide-1 Agonist Exendin-4 Facilitates the Extinction of Cocaine-Induced Condition Place Preference. Front Syst Neurosci 2022; 15:711750. [PMID: 35024034 PMCID: PMC8744468 DOI: 10.3389/fnsys.2021.711750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
Accumulating studies suggest that the glucagon-like peptide-1 receptor agonist exendin-4 (Ex4) and toll-like receptor 4 (TLR4) play a pivotal role in the maladaptive behavior of cocaine. However, few studies have assessed whether Ex4 can facilitate the extinction of drug-associated behavior and attenuate the reinstatement of cocaine-induced condition place preference (CPP) in mice. The main objective of the present study was to evaluate Ex4's ability to regulate the extinction and reinstatement of cocaine-induced CPP. C57BL/6 mice were conditioned to either cocaine (20 mg/kg) or an equivalent volume of saline to establish a cocaine-mediated CPP paradigm. To investigate the potential effects of Ex4 on extinction, animals received an intraperitoneal injection of Ex4 either immediately or 6 h after each extinction or only on the test day. The persistence of extinction was measured using the reinstatement paradigm evoked by 10 mg/kg of cocaine. To explore the possible impacts of Ex4 and neuroinflammation on cocaine, the expression levels of TLR4 within the hippocampus was detected using western blotting. As a result, we found that systemic administration of Ex4 immediately after each extinction training, instead of 6 h after each extinction and on the day of extinction test, was capable of facilitating extinction in the confined or non-confined CPP extinction paradigms and blocking the cocaine-primed reinstatement of cocaine-induced CPP. Additionally, we also observed that Ex4 was competent to alleviate TLR4 signaling that has been up-regulated by cocaine. Altogether, our findings indicated that the combination of Ex4 with daily extinction training was sufficient to facilitate extinction of the conditioned behavior, attenuate reinstatement of cocaine-induced CPP and inhibit TLR4 signaling. Thus, Ex4 deserves further investigation as a potential intervention for the treatment of cocaine use disorder.
Collapse
Affiliation(s)
- Changliang Zhu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Tao Hong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hailiang Li
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Shucai Jiang
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Baorui Guo
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Lei Wang
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Jiangwei Ding
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Caibin Gao
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Yu Sun
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Tao Sun
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Feng Wang
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China.,Department of Neurosurgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yangyang Wang
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Din Wan
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
20
|
Katsenos AP, Davri AS, Simos YV, Nikas IP, Bekiari C, Paschou SA, Peschos D, Konitsiotis S, Vezyraki P, Tsamis KI. New treatment approaches for Alzheimer's disease: preclinical studies and clinical trials centered on antidiabetic drugs. Expert Opin Investig Drugs 2022; 31:105-123. [PMID: 34941464 DOI: 10.1080/13543784.2022.2022122] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) represent two major chronic diseases that affect a large percentage of the population and share common pathogenetic mechanisms, including oxidative stress and inflammation. Considering their common mechanistic aspects, and given the current lack of effective therapies for AD, accumulating research has focused on the therapeutic potential of antidiabetic drugs in the treatment or prevention of AD. AREAS COVERED This review examines the latest preclinical and clinical evidence on the potential of antidiabetic drugs as candidates for AD treatment. Numerous approved drugs for T2DM, including insulin, metformin, glucagon-like peptide-1 receptor agonists (GLP-1 RA), and sodium glucose cotransporter 2 inhibitors (SGLT2i), are in the spotlight and may constitute novel approaches for AD treatment. EXPERT OPINION Among other pharmacologic agents, GLP-1 RA and SGLT2i have so far exhibited promising results as novel treatment approaches for AD, while current research has centered on deciphering their action on the central nervous system (CNS). Further investigation is crucial to reveal the most effective pharmacological agents and their optimal combinations, maximize their beneficial effects on neurons, and find ways to increase their distribution to the CNS.
Collapse
Affiliation(s)
- Andreas P Katsenos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece.,Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, Greece
| | - Athena S Davri
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Yannis V Simos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece.,Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, Greece
| | - Ilias P Nikas
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - Chryssa Bekiari
- Laboratory of Anatomy and Histology, school of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stavroula A Paschou
- Endocrine Unit and Diabetes Centre, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Peschos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece.,Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, Greece
| | | | - Patra Vezyraki
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Konstantinos I Tsamis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece.,Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, Greece.,Department of Neurology, University Hospital of Ioannina, Ioannina, Greece
| |
Collapse
|
21
|
Cernea S, Dima L, Correll CU, Manu P. Pharmacological Management of Glucose Dysregulation in Patients Treated with Second-Generation Antipsychotics. Drugs 2021; 80:1763-1781. [PMID: 32930957 DOI: 10.1007/s40265-020-01393-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Fasting hyperglycemia, impaired glucose tolerance, prediabetes, and diabetes are frequently present in patients treated with second-generation antipsychotics (SGAPs) for schizophrenia, bipolar disorder, and other severe mental illnesses. These drugs are known to produce weight gain, which may lead to insulin resistance, glucose intolerance, and metabolic syndrome, which constitute important risk factors for the emergence of diabetes. The aim of this review was to formulate therapeutic guidelines for the management of diabetes in patients treated with SGAPs, based on the association between SGAP-induced weight gain and glucose dysregulation. A systematic search in PubMed from inception to March 2020 for randomized controlled trials (RCTs) of diabetes or prediabetes in patients treated with SGAPs was performed. PubMed was also searched for the most recent clinical practice guidelines of interventions for co-morbid conditions associated with diabetes mellitus (DM) (arterial hypertension and dyslipidemia), lifestyle interventions and switching from high metabolic liability SGAPs to safer SGAPs. The search identified 14 RCTs in patients treated with SGAPs. Drug therapy using metformin as first-line therapy and glucagon-like peptide-1 receptor agonists (GLP-1 RAs) or perhaps sodium-glucose cotransporter-2 (SGLT2) inhibitors as add-on therapy, might be preferred in these patients as well, as they favorably influence glucose metabolism and body mass index, and provide cardio-renal benefits in general to the DM population, although for the SGLT-2 inhibitors there are no RCTs in this specific patient category so far. Metformin is also useful for treatment of prediabetes. Arterial hypertension should be treated with angiotensin-converting enzyme inhibitors or angiotensin-receptor blockers, and statins should be used for correction of dyslipidemia. The outcome of lifestyle-changing interventions has been disappointing. Switching from clozapine, olanzapine, or quetiapine to lower cardiometabolic-risk SGAPs, like aripiprazole, brexpiprazole, cariprazine, lurasidone, or ziprasidone, has been recommended.
Collapse
Affiliation(s)
- Simona Cernea
- Faculty of Medicine/Department M4/Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Târgu Mureș, Romania.,Diabetes, Nutrition and Metabolic Diseases Outpatient Unit, Emergency County Clinical Hospital, Târgu Mureş, Romania
| | - Lorena Dima
- Department of Fundamental Disciplines and Clinical Prevention, Faculty of Medicine, Universitatea Transilvania, Nicolae Balcescu Str 59, Brașov, 500019, Romania.
| | - Christoph U Correll
- Charite Universitaetsmedizin, Department of Child and Adolescent Psychiatry, Berlin, and Campus Virchow-Klinikum, Mittelallee 5A, Berlin, 13353, Germany.,Department of Psychiatry and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.,Department of Psychiatry and Molecular Medicine, Zucker Hillside Hospital, Northwell Health System, Glen Oaks, NY, USA
| | - Peter Manu
- Department of Psychiatry, Hofstra Northwell School of Medicine, Hempstead, NY, USA.,Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, NY, USA.,South Oaks Hospital, Northwell Health System, Amityville, NY, USA
| |
Collapse
|
22
|
Zhu C, Tao H, Rong S, Xiao L, Li X, Jiang S, Guo B, Wang L, Ding J, Gao C, Chang H, Sun T, Wang F. Glucagon-Like Peptide-1 Analog Exendin-4 Ameliorates Cocaine-Mediated Behavior by Inhibiting Toll-Like Receptor 4 Signaling in Mice. Front Pharmacol 2021; 12:694476. [PMID: 34349653 PMCID: PMC8327264 DOI: 10.3389/fphar.2021.694476] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
Exendin-4 (Ex4), a long-lasting glucagon-like peptide-1 analog, was reported to exert favourable actions on inhibiting cocaine-associated rewarding and reinforcing effects of drug in animal models of addiction. However, the therapeutic potential of different dose of GLP-1 receptor agonist Ex4 in different behavioral paradigms and the underlying pharmacological mechanisms of action are incompletely understood. Herein, we firstly investigated the effects of Ex4 on cocaine-induced condition place preference (CPP) as well as extinction and reinstatement in male C57BL/6J mice. Additionally, we sought to elucidate the underlying pharmacological mechanism of these actions of Ex4. The paradigm of cocaine-induced CPP was established using 20 mg/kg cocaine or saline alternately during conditioning, while the reinstatement paradigm was modeled using 10 mg/kg cocaine on the reinstatement day. Different dose of Ex4 was administrated intraperitoneally either during conditioning or during extinction state or only on the test day. To elucidate the molecular mechanism underlying the potential effects of Ex4 on maladaptive behaviors of cocaine, the TLR4-related inflammation within the hippocampus was observed by immunofluorescence staining, and the expression levels of toll-like receptor 4 (TLR4), tumor necrosis factor (TNF)-α, and interleukin (IL)-1β were detected by Western blotting. As a consequence, systemic administration of different dose of Ex4 was sufficient to inhibit the acquisition and expression of cocaine-induced CPP, facilitate the extinction of cocaine-associated reward and attenuate reinstatement of cocaine-induced behavior. Furthermore, Ex4 treatment diminished expression levels of TLR4, TNF-α, and IL-1β, which were up-regulated by cocaine exposure. Altogether, our results indicated that Ex4 effectively ameliorated cocaine-induced behaviors likely through neurobiological mechanisms partly attributable to the inhibition of TLR4, TNF-α and IL-1β in mice. Consequently, our findings improved our understanding of the efficacy of Ex4 for the amelioration of cocaine-induced behavior and suggested that Ex4 may be applied as a drug candidate for cocaine addiction.
Collapse
Affiliation(s)
- Changliang Zhu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Hong Tao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shikuo Rong
- Department of General Surgery, Chengdu Second Hospital, Chendu, China
| | - Lifei Xiao
- Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Xinxiao Li
- Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Shucai Jiang
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Baorui Guo
- Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Lei Wang
- Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Jiangwei Ding
- Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Caibing Gao
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Haigang Chang
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Tao Sun
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Feng Wang
- Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China.,Department of Neurosurgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
23
|
Turan I, Sayan Ozacmak H, Ozacmak VH, Ergenc M, Bayraktaroğlu T. The effects of glucagon-like peptide 1 receptor agonist (exenatide) on memory impairment, and anxiety- and depression-like behavior induced by REM sleep deprivation. Brain Res Bull 2021; 174:194-202. [PMID: 34146656 DOI: 10.1016/j.brainresbull.2021.06.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 06/04/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022]
Abstract
Previous investigations have shown that REM sleep deprivation impairs the hippocampus-dependent memory, long-term potentiation and causing mood changes. The aim of the present study was to explore the effects of exenatide on memory performance, anxiety- and depression like behavior, oxidative stress markers, and synaptic protein levels in REM sleep deprived rats. A total of 40 male Wistar rats were randomly divided to control, exenatide-treated control, sleep deprivation (SD), wide platform (WP) and exenatide-treated SD groups. During experiments, exenatide treatment (0.5 μg/kg, subcutaneously) was applied daily in a single dose for 9 days. Modified multiple platform method was employed to generate REM sleep deprivation for 72 h. The Morris water maze test was used to assess memory performance. Anxiety- and depression-like behaviors were evaluated by open field test (OFT), elevated plus maze (EPM) forced swimming test (FST), respectively 72 h after REMSD. The levels of Ca2+/calmodulin-dependent protein kinase II (CaMKII) and postsynaptic density proteins 95 (PSD95) were measured in tissues of hippocampus and prefrontal cortex. The content of malondialdehyde (MDA) and reduced glutathione (GSH) were also measured. In the present study, an impairment in memory was observed in SD rats at the 24th hour of SD in compare to those of other groups. REMSD increased depression-like behavior in FST as well as the number of rearing and crossing square in OFT. Anxiety is the most common comorbid condition with depressive disorders. Contents of CaMKII and PSD95 decreased in hippocampus of SD rats. Exenatide treatment improved the impaired memory of SD rats and increased CaMKII content in hippocampus There was no difference in MDA and GSH levels among groups. Exenatide treatment also diminished locomotor activity in OFT. In conclusion, treatment with exenatide, at least in part, prevented from these cognitive and behavioral changes possibly through normalizing CaMKII levels in the hippocampus.
Collapse
Affiliation(s)
- Inci Turan
- Zonguldak Bulent Ecevit University Faculty of Medicine, Department of Physiology, Zonguldak, Turkey.
| | - Hale Sayan Ozacmak
- Zonguldak Bulent Ecevit University Faculty of Medicine, Department of Physiology, Zonguldak, Turkey
| | - V Haktan Ozacmak
- Zonguldak Bulent Ecevit University Faculty of Medicine, Department of Physiology, Zonguldak, Turkey
| | - Meryem Ergenc
- Zonguldak Bulent Ecevit University Faculty of Medicine, Institute of Health Sciences Department of Physiology, Zonguldak, Turkey
| | - Taner Bayraktaroğlu
- Zonguldak Bulent Ecevit Unıversity Faculty of Medicine, Department of Endocrinology, Zonguldak, Turkey
| |
Collapse
|
24
|
Mehta K, Behl T, Kumar A, Uddin MS, Zengin G, Arora S. Deciphering the Neuroprotective Role of Glucagon-like Peptide-1 Agonists in Diabetic Neuropathy: Current Perspective and Future Directions. Curr Protein Pept Sci 2021; 22:4-18. [PMID: 33292149 DOI: 10.2174/1389203721999201208195901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 09/15/2020] [Accepted: 11/25/2020] [Indexed: 11/22/2022]
Abstract
Diabetic neuropathy is referred to as a subsequential and debilitating complication belonging to type 1 and type 2 diabetes mellitus. It is a heterogeneous group of disorders with a particularly complex pathophysiology and also includes multiple forms, ranging from normal discomfort to death. The evaluation of diabetic neuropathy is associated with hyperglycemic responses, resulting in an alteration in various metabolic pathways, including protein kinase C pathway, polyol pathway and hexosamine pathway in Schwann and glial cells of neurons. The essential source of neuronal destruction is analogous to these respective metabolic pathways, thus identified as potential therapeutic targets. These pathways regulating therapeutic medications may be used for diabetic neuropathy, however, only target specific drugs could have partial therapeutic activity. Various antidiabetic medications have been approved and marketed, which possess the therapeutic ability to control hyperglycemia and ameliorate the prevalence of diabetic neuropathy. Among all antidiabetic medications, incretin therapy, including Glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors, are the most favorable medications for the management of diabetes mellitus and associated peripheral neuropathic complications. Besides enhancing glucose-evoked insulin release from pancreatic β-cells, these therapeutic agents also play a vital role to facilitate neurite outgrowth and nerve conduction velocity in dorsal root ganglion. Furthermore, incretin therapy also activates cAMP and ERK signalling pathways, resulting in nerve regeneration and repairing. These effects are evidently supported by a series of preclinical data and investigations associated with these medications. However, the literature lacks adequate clinical trial outcomes related to these novel antidiabetic medications. The manuscript emphasizes the pathogenesis, current pharmacological approaches and vivid description of preclinical and clinical data for the effective management of diabetic neuropathy.
Collapse
Affiliation(s)
- Keshav Mehta
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - M Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk Uniersity Campus, Konya, Turkey
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
25
|
Lafferty RA, O’Harte FPM, Irwin N, Gault VA, Flatt PR. Proglucagon-Derived Peptides as Therapeutics. Front Endocrinol (Lausanne) 2021; 12:689678. [PMID: 34093449 PMCID: PMC8171296 DOI: 10.3389/fendo.2021.689678] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
Initially discovered as an impurity in insulin preparations, our understanding of the hyperglycaemic hormone glucagon has evolved markedly over subsequent decades. With description of the precursor proglucagon, we now appreciate that glucagon was just the first proglucagon-derived peptide (PGDP) to be characterised. Other bioactive members of the PGDP family include glucagon-like peptides -1 and -2 (GLP-1 and GLP-2), oxyntomodulin (OXM), glicentin and glicentin-related pancreatic peptide (GRPP), with these being produced via tissue-specific processing of proglucagon by the prohormone convertase (PC) enzymes, PC1/3 and PC2. PGDP peptides exert unique physiological effects that influence metabolism and energy regulation, which has witnessed several of them exploited in the form of long-acting, enzymatically resistant analogues for treatment of various pathologies. As such, intramuscular glucagon is well established in rescue of hypoglycaemia, while GLP-2 analogues are indicated in the management of short bowel syndrome. Furthermore, since approval of the first GLP-1 mimetic for the management of Type 2 diabetes mellitus (T2DM) in 2005, GLP-1 therapeutics have become a mainstay of T2DM management due to multifaceted and sustainable improvements in glycaemia, appetite control and weight loss. More recently, longer-acting PGDP therapeutics have been developed, while newfound benefits on cardioprotection, bone health, renal and liver function and cognition have been uncovered. In the present article, we discuss the physiology of PGDP peptides and their therapeutic applications, with a focus on successful design of analogues including dual and triple PGDP receptor agonists currently in clinical development.
Collapse
Affiliation(s)
| | | | | | - Victor A. Gault
- School of Biomedical Sciences, Ulster University, Coleraine, United Kingdom
| | | |
Collapse
|
26
|
Lietzau G, Ntika S, Pintana H, Tracy L, Klein T, Nyström T, Darsalia V, Patrone C, Krizhanovskii C. A High-Fat Diet Increases Activation of the Glucagon-Like Peptide-1-Producing Neurons in the Nucleus Tractus Solitarii: an Effect that is Partially Reversed by Drugs Normalizing Glycemia. Cell Mol Neurobiol 2021; 42:1995-2002. [PMID: 33811589 PMCID: PMC9239971 DOI: 10.1007/s10571-021-01079-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/12/2021] [Indexed: 11/01/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) is a peripheral incretin and centrally active peptide produced in the intestine and nucleus tractus solitarii (NTS), respectively. GLP-1 not only regulates metabolism but also improves cognition and is neuroprotective. While intestinal GLP-1-producing cells have been well characterized, less is known about GLP-1-producing neurons in NTS. We hypothesized that obesity-induced type 2 diabetes (T2D) impairs the function of NTS GLP-1-producing neurons and glycemia normalization counteracts this effect. We used immunohistochemistry/quantitative microscopy to investigate the number, potential atrophy, and activation (cFos-expression based) of NTS GLP-1-producing neurons, in non-diabetic versus obese/T2D mice (after 12 months of high-fat diet). NTS neuroinflammation was also assessed. The same parameters were quantified in obese/T2D mice treated from month 9 to 12 with two unrelated anti-hyperglycemic drugs: the dipeptidyl peptidase-4 inhibitor linagliptin and the sulfonylurea glimepiride. We show no effect of T2D on the number and volume but increased activation of NTS GLP-1-producing neurons. This effect was partially normalized by both anti-diabetic treatments, concurrent with decreased neuroinflammation. Increased activation of NTS GLP-1-producing neurons could represent an aberrant metabolic demand in T2D/obesity, attenuated by glycemia normalization. Whether this effect represents a pathophysiological process preceding GLP-1 signaling impairment in the CNS, remains to be investigated.
Collapse
Affiliation(s)
- Grazyna Lietzau
- Department of Clinical Science and Education, Karolinska Institutet, Sodersjukhuset Internal Medicine, 118 83, Stockholm, Sweden. .,Faculty of Medicine, Department of Anatomy and Neurobiology, Medical University of Gdansk, Gdansk, Poland.
| | - Stelia Ntika
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Research, Södertälje Hospital, Södertälje, Sweden
| | - Hiranya Pintana
- Department of Clinical Science and Education, Karolinska Institutet, Sodersjukhuset Internal Medicine, 118 83, Stockholm, Sweden
| | - Linda Tracy
- Department of Research, Södertälje Hospital, Södertälje, Sweden
| | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Thomas Nyström
- Department of Clinical Science and Education, Karolinska Institutet, Sodersjukhuset Internal Medicine, 118 83, Stockholm, Sweden
| | - Vladimer Darsalia
- Department of Clinical Science and Education, Karolinska Institutet, Sodersjukhuset Internal Medicine, 118 83, Stockholm, Sweden
| | - Cesare Patrone
- Department of Clinical Science and Education, Karolinska Institutet, Sodersjukhuset Internal Medicine, 118 83, Stockholm, Sweden
| | - Camilla Krizhanovskii
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden. .,Department of Research, Södertälje Hospital, Södertälje, Sweden.
| |
Collapse
|
27
|
Reich N, Hölscher C. Acylated Ghrelin as a Multi-Targeted Therapy for Alzheimer's and Parkinson's Disease. Front Neurosci 2020; 14:614828. [PMID: 33381011 PMCID: PMC7767977 DOI: 10.3389/fnins.2020.614828] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Much thought has been given to the impact of Amyloid Beta, Tau and Alpha-Synuclein in the development of Alzheimer's disease (AD) and Parkinson's disease (PD), yet the clinical failures of the recent decades indicate that there are further pathological mechanisms at work. Indeed, besides amyloids, AD and PD are characterized by the culminative interplay of oxidative stress, mitochondrial dysfunction and hyperfission, defective autophagy and mitophagy, systemic inflammation, BBB and vascular damage, demyelination, cerebral insulin resistance, the loss of dopamine production in PD, impaired neurogenesis and, of course, widespread axonal, synaptic and neuronal degeneration that leads to cognitive and motor impediments. Interestingly, the acylated form of the hormone ghrelin has shown the potential to ameliorate the latter pathologic changes, although some studies indicate a few complications that need to be considered in the long-term administration of the hormone. As such, this review will illustrate the wide-ranging neuroprotective properties of acylated ghrelin and critically evaluate the hormone's therapeutic benefits for the treatment of AD and PD.
Collapse
Affiliation(s)
- Niklas Reich
- Biomedical & Life Sciences Division, Lancaster University, Lancaster, United Kingdom
| | - Christian Hölscher
- Neurology Department, A Second Hospital, Shanxi Medical University, Taiyuan, China.,Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
28
|
Niu Y, Wang X, Li M, Niu B. Exosomes from human umbilical cord Mesenchymal stem cells attenuates stress-induced hippocampal dysfunctions. Metab Brain Dis 2020; 35:1329-1340. [PMID: 32761493 DOI: 10.1007/s11011-019-00514-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/05/2019] [Indexed: 12/30/2022]
Abstract
Human Mesenchymal Stem Cells (MSCs) especially human umbilical cord MSCs is the novel regenerative cell resource for regenerative therapy. However, the biological underpinning of MSCs in neuroprotections requires deep understanding. Exosomes is an important biological factor due to its multiple types of contents with various biological function. In current study, we collected the exosome from umbilical cord mesenchymal stem cells (hUC-MSCs) and tested the neuroprotective effects to brain stress. Proteomic analysis indicates significant enriched protein components display the functions in metabolic regulation. We then injected the exosome (MSC-Ex) to adult mice by i.v injection. On physiological level, treatment of MSC-Ex increased the adiponectin level in peripheral central nervous system (CNS). Moreover, MSC-Ex significantly accelerated the differentiation of adult neural stem cells but did not benefit the related cognitive behavior. We then created acute brain disorder model with STZ intra-hippocampal injection. Compared with STZ group, treatment of MSC-Ex improved cognitive function. Moreover, MSC-Ex promotes hippocampal neurogenesis that was suppressed by STZ injection. In conclusion, hUC-MSCs derived exosome would exert the neural regenerative effects associating with its metabolism regulatory capacity.
Collapse
Affiliation(s)
- Yuhu Niu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, 56, Xinjian South Rd., Taiyuan, China
| | - Xiuwei Wang
- Department of Biotechnology, Capital Institute of Pediatric, Beijing, China
| | - Meining Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, 56, Xinjian South Rd., Taiyuan, China
| | - Bo Niu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, 56, Xinjian South Rd., Taiyuan, China.
- Department of Biotechnology, Capital Institute of Pediatric, Beijing, China.
| |
Collapse
|
29
|
Albazal A, Delshad AA, Roghani M. Melatonin reverses cognitive deficits in streptozotocin-induced type 1 diabetes in the rat through attenuation of oxidative stress and inflammation. J Chem Neuroanat 2020; 112:101902. [PMID: 33276072 DOI: 10.1016/j.jchemneu.2020.101902] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 11/06/2020] [Accepted: 11/28/2020] [Indexed: 12/30/2022]
Abstract
Uncontrolled diabetes mellitus (DM) is linked to attentional deficits and cognition deterioration. The neurohormone melatonin is an endogenous synchronizer of circadian rhythms with multiple protective properties. This research was designed to assess its effect against learning and memory decline in streptozotocin (STZ)-induced diabetic rats. Rats were assigned to control, melatonin-treated control, diabetic, and melatonin-treated diabetic groups. Melatonin was administered i.p. at a dose of 10 mg/kg/day for 47 days. Treatment of diabetic rats with melatonin reversed decline of spatial recognition memory in Y maze, performance of rats in novel object discrimination, and retention and recall in passive avoidance tasks. Furthermore, melatonin appropriately attenuated hippocampal malondialdehyde (MDA) and reactive oxygen species (ROS) and improved superoxide dismutase (SOD) activity and improved mitochondrial membrane potential (MMP) and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) with no significant effect on nitrite, glutathione (GSH) and catalase activity. Besides, hippocampal level of acetylcholinesterase (AChE), glial fibrillary acidic protein (GFAP), nuclear factor-kappaB (NF-κB), interleukin 6 (IL-6) and tumor necrosis factor α (TNFα) decreased following melatonin treatment. There was also a reduction of dendritic spines of pyramidal neurons of hippocampal CA1 area in diabetic group that was significantly alleviated upon melatonin treatment. Melatonin could ameliorate learning and memory disturbances in diabetic rats through mitigation of cholinesterase activity, astrocytes, oxidative stress and inflammation and also via upregulation of some antioxidants in addition to its prevention of dendritic spine loss.
Collapse
Affiliation(s)
- Ala Albazal
- School of Medicine, Shahed University, Tehran, Iran
| | | | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran.
| |
Collapse
|
30
|
Meyhöfer S, Schmid SM. [Diabetes complications - diabetes and the nervous system]. Dtsch Med Wochenschr 2020; 145:1599-1605. [PMID: 33142326 DOI: 10.1055/a-1038-0102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Diabetes mellitus is a chronic metabolic disease associated with multiple long-term complications. Besides macro- and microvascular complications, patient's well-being can be severely impaired by complications affecting the nervous system. About 50 % of patients with diabetes suffer from polyneuropathy. Moreover, the risk of developing cognitive impairment and dementia is also increased in older people with diabetes. Insufficient glycemic control, young age at diagnosis of diabetes are discussed as risk factors for developing diabetes complications. The early identification and prevention of factors predicting diabetes complications that affect the nervous system are still challenging and in need for further research.
Collapse
|
31
|
Yaribeygi H, Rashidy-Pour A, Atkin SL, Jamialahmadi T, Sahebkar A. GLP-1 mimetics and cognition. Life Sci 2020; 264:118645. [PMID: 33121988 DOI: 10.1016/j.lfs.2020.118645] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/12/2020] [Accepted: 10/20/2020] [Indexed: 12/14/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) receptor agonists are a class of antidiabetic drugs that improve the glycaemia via several molecular pathways. Recent evidence suggest that they also have additional effects modulating pathophysiologic pathways included in cognitive disorders. Since some forms of cognitive dysfunction such as Alzheimer's disease are more common among diabetic patients than in the normal population, antidiabetic drugs that have neuroprotective effects affording protection for cognitive disorders would be of benefit. Therefore, we reviewed the pharmacologic effects of GLP-1 analogues and found that they may have the additional benefit of improving cognitive performance via at least eight molecular mechanisms.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | | | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran; Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Halal Research Center of IRI, FDA, Tehran, Iran; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland.
| |
Collapse
|
32
|
An Almond-Based Low Carbohydrate Diet Improves Depression and Glycometabolism in Patients with Type 2 Diabetes through Modulating Gut Microbiota and GLP-1: A Randomized Controlled Trial. Nutrients 2020; 12:nu12103036. [PMID: 33022991 PMCID: PMC7601479 DOI: 10.3390/nu12103036] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/18/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Alow carbohydrate diet (LCD) is more beneficial for the glycometabolism in type 2 diabetes (T2DM) and may be effective in reducing depression. Almond, which is a common nut, has been shown to effectively improve hyperglycemia and depression symptoms. This study aimed to determine the effect of an almond-based LCD (a-LCD) on depression and glycometabolism, as well as gut microbiota and fasting glucagon-like peptide 1 (GLP-1) in patients with T2DM. Methods: This was a randomized controlled trial which compared an a-LCD with a low-fat diet (LFD). Forty-five participants with T2DM at a diabetes club and the Endocrine Division of the First and Second Affiliated Hospital of Soochow University between December 2018 to December 2019 completed each dietary intervention for 3 months, including 22 in the a-LCD group and 23 in the LFD group. The indicators for depression and biochemical indicators including glycosylated hemoglobin (HbA1c), gut microbiota, and GLP-1 concentration were assessed at the baseline and third month and compared between the two groups. Results: A-LCD significantly improved depression and HbA1c (p < 0.01). Meanwhile, a-LCD significantly increased the short chain fatty acid (SCFAs)-producing bacteria Roseburia, Ruminococcus and Eubacterium. The GLP-1 concentration in the a-LCD group was higher than that in the LFD group (p < 0.05). Conclusions: A-LCD could exert a beneficial effect on depression and glycometabolism in patients with T2DM. We speculate that the role of a-LCD in improving depression in patients with T2DM may be associated with it stimulating the growth of SCFAs-producing bacteria, increasing SCFAs production and GPR43 activation, and further maintaining GLP-1 secretion. In future studies, the SCFAs and GPR43 activation should be further examined.
Collapse
|
33
|
Jin Y, Zhao H, Hou Y, Song G. The effects of dipeptidyl peptidase-4 inhibitors and glucagon-like peptide 1 receptor agonists on cognitive functions in adults with type 2 diabetes mellitus: a systematic review and meta-analysis. Acta Diabetol 2020; 57:1129-1144. [PMID: 32300876 DOI: 10.1007/s00592-020-01529-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 03/27/2020] [Indexed: 12/30/2022]
Abstract
AIMS The effects of dipeptidyl peptidase-4 inhibitors (DPP-4 inhibitors/DPP-4I) and glucagon-like peptide 1 receptor agonists (GLP-1 RA) on cognition in patients with type 2 diabetes mellitus (T2DM) remain controversial. We aimed to explore this clinical issue through a systematic review and meta-analysis. METHODS PubMed, EMBASE and the Cochrane Library were searched, and data were expressed as mean difference (MD) or hazard ratio (HR)/odds ratio (OR) with a 95% confidence interval (CI). Heterogeneity was assessed using the Chi-squared test and the I2 statistic. The study was registered with PROSPERO (ID: CRD42019138777). RESULTS Eleven studies (n = 304,258 T2DM patients) were included in our review. In the DPP-4I group, six studies were enrolled to estimate ΔMini-Mental State Examination (MMSE) scores from baseline to the final evaluations after DPP-4I treatment, which showed no statistical difference (MD 0.20; 95% CI - 0.75 to 1.15, p = 0.68). ΔMMSE scores in the DPP-4I group and the other antidiabetic groups were compared, revealing no statistical difference (MD 0.57; 95% CI - 0.05 to 1.19, p = 0.07). Two cohort studies were pooled to determine the HRs for dementia, showing a lower risk of dementia after DPP-4I treatment (HR 0.52; 95% CI 0.29-0.93, p = 0.03). In the GLP-1 analogs group, two studies were included, one of which revealed a downward trend in the risk of dementia after GLP-1 analog treatment, while the other revealed no significant difference after incretins treatment. CONCLUSIONS Currently there is not enough irrefutable evidence to support the hypothesis of positive effects of incretins on cognition. Further clinical studies need to be performed.
Collapse
Affiliation(s)
- Yuxin Jin
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, Hebei, People's Republic of China
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Hang Zhao
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Yilin Hou
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, Hebei, People's Republic of China
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Guangyao Song
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, Hebei, People's Republic of China.
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, 050051, Hebei, People's Republic of China.
| |
Collapse
|
34
|
Hölscher C. Evidence for pathophysiological commonalities between metabolic and neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 155:65-89. [PMID: 32854859 DOI: 10.1016/bs.irn.2020.01.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Diabetes mellitus is a risk factor for developing neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases. This relationship seems counter-intuitive as these pathological syndromes appear to be very different. However, they share underlying mechanisms such as desensitization of insulin signaling. Insulin not only regulates blood glucose levels, but also acts as a growth factor that is important for neuronal activity and repair. Insulin signaling desensitization has been found in the brains of people with progressive neurodegenerative diseases, which is most likely driven by chronic inflammation. Based on this, insulin has been tested in patients with Alzheimer's disease, and it was found that memory formation was improved and brain pathology reduced. Glucagon-like peptide-1 (GLP-1) is an incretin hormone, and numerous drugs that mimic this peptide are on the market to treat type 2 diabetes mellitus. Preclinical studies have provided robust evidence that some of these drugs, such as liraglutide or lixisenatide can enter the brain and improve key pathological parameters, such as memory loss, impairment of motor activity, synapse loss, reduced energy utilization by neurons and chronic inflammation in the brain. First clinical trials with a GLP-1 mimetic show good effects in patients with Parkinson's disease, improving motor control and insulin signaling in the brain. This is a proof of concept that this approach is viable and that drug treatment affects the main drivers of the disease and does not just modify the symptoms. It demonstrates that this new research area is a promising and fertile space for the development of novel treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Christian Hölscher
- Neurology Department of the Second Associated Hospital of Shanxi Medical University, Taiyuan, Shanxi, PR China; Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan, PR China.
| |
Collapse
|
35
|
Diabetes and dementia - the two faces of Janus. ACTA ACUST UNITED AC 2020; 5:e186-e197. [PMID: 32832719 PMCID: PMC7433787 DOI: 10.5114/amsad.2020.97433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/18/2020] [Indexed: 01/03/2023]
Abstract
Patients with type 2 diabetes are at high risk for cognitive decline and dementia. Despite the limited data on the possible pathogenetic mechanisms, evidence suggests that cognitive decline, and thus dementia and Alzheimer’s disease, might arise from a complex interplay between type 2 diabetes and the aging brain, including decreased insulin signalling and glucose metabolism, mitochondrial dysfunction, neuroinflammation, and vascular disease. Furthermore, there is increasing interest on the effects of antidiabetic agents on cognitive decline. There are many studies showing that antidiabetic agents might have beneficial effects on the brain, mainly through inhibition of oxidative stress, inflammation, and apoptosis. In addition, experimental studies on patients with diabetes and Alzheimer’s disease have shown beneficial effects on synaptic plasticity, metabolism of amyloid-β, and microtubule-associated protein tau. Therefore, in the present review, we discuss the effects of antidiabetic agents in relation to cognitive decline, and in particular dementia and Alzheimer’s disease, in patients with type 2 diabetes.
Collapse
|
36
|
Géa LP, da Rosa ED, Panizzutti BS, de Aguiar ÉZ, de Oliveira LF, Ferrari P, Piato A, Gomez R, Colombo R, Rosa AR. Reduction of hippocampal IL-6 levels in LPS-injected rats following acute exendin-4 treatment. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:1303-1311. [PMID: 32363414 DOI: 10.1007/s00210-020-01867-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 04/02/2020] [Indexed: 12/12/2022]
Abstract
Preclinical evidence on the role of glucagon-like peptide-1 receptor (GLP-1r) agonists in the brain led to an increased interest in repurposing these compounds as a therapy for central nervous system (CNS) disorders and associated comorbidities. We aimed to investigate the neuroprotective effects of acute treatment with exendin (EX)-4, a GLP-1r agonist, in an animal model of inflammation. We evaluated the effect of different doses of EX-4 on inflammatory, neurotrophic, and oxidative stress parameters in the hippocampus and serum of lipopolysaccharide (LPS)-injected animals. Male Wistar rats were injected with LPS (0.25 mg/kg i.p.) and treated with different doses of EX-4 (0.1, 0.3, or 0.5 μg/kg i.p.). Sickness behavior was assessed by locomotor activity and body weight, and depressive-like behavior was also evaluated using forced swim test (FST). Brain-derived neurotrophic factor (BDNF), thiobarbituric acid reactive species (TBARS), and interleukin (IL)-6 were quantified in the serum and hippocampus. Glycemia was also analyzed pre- and post-EX-4 treatment. LPS groups exhibited decreased frequency of crossing and reduced body weight (p < 0.001), while alterations on FST were not observed. The higher dose of EX-4 reduced IL-6 in the hippocampus of LPS-injected animals (p = 0.018), and EX-4 per se reduced TBARS serum levels with a modest antioxidant effect in the LPS groups (p ≤ 0.005). BDNF hippocampal levels seemed to be increased in the LPS+EX-4 0.5 group compared with LPS+Saline (p > 0.05). Our study provides evidence on acute anti-inflammatory effects of EX-4 in the hippocampus of rats injected with LPS, contributing to future studies on repurposing compounds with potential neuroprotective properties.
Collapse
Affiliation(s)
- Luiza P Géa
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil
- Programa de Pós-graduação em Ciências Biológicas: Farmacologia e Terapêutica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Eduarda D da Rosa
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil
- Programa de Pós-gradução em Ciências Biológicas: Bioquímica, UFRGS, Porto Alegre, RS, Brazil
| | - Bruna S Panizzutti
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil
- Programa de Pós-graduação em Psiquiatria e Ciências do Comportamento, UFRGS, Porto Alegre, RS, Brazil
| | - Érica Z de Aguiar
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil
| | - Larissa F de Oliveira
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil
| | - Pamela Ferrari
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil
| | - Angelo Piato
- Programa de Pós-graduação em Ciências Biológicas: Farmacologia e Terapêutica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Rosane Gomez
- Programa de Pós-graduação em Ciências Biológicas: Farmacologia e Terapêutica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Rafael Colombo
- Laboratório de Farmacologia e Fisiologia, Universidade de Caxias do Sul (UCS), Caixas do Sul, RS, Brazil
| | - Adriane R Rosa
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil.
- Programa de Pós-graduação em Ciências Biológicas: Farmacologia e Terapêutica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
- Programa de Pós-graduação em Psiquiatria e Ciências do Comportamento, UFRGS, Porto Alegre, RS, Brazil.
| |
Collapse
|
37
|
Zhang Q, Zhang XF, Niu CY. Application prospects of glucagon-like peptide-1 receptor agonists in treatment of metabolic diseases. Shijie Huaren Xiaohua Zazhi 2020; 28:393-400. [DOI: 10.11569/wcjd.v28.i11.393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome [overweight, obesity, type 2 diabetes mellitus (T2DM), and lipid metabolism disorder] is directly related to cardiovascular diseases, non-alcoholic fatty liver disease (NAFLD), tumors, and other diseases, and its prevalence has been increasing sharply, bringing an increasingly heavy burden on society. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are a novel class of glucose-dependent hypoglycemic drugs modified from glucagon-like peptide-1. Since their mechanism of action is different from that of traditional insulin and oral secretagogues, they overcome the adverse reactions associated with traditional oral medications and insulin, can improve the success rate of T2DM control, and has been widely used in the treatment of T2DM. Recent studies have found that, in addition to hypoglycemic effect, GLP-1RAs can improve metabolic diseases, such as diabetes-related complications, NAFLD, and cardiovascular diseases, and therefore have broad application prospects. This paper reviews the potential role of GLP-1RAs in these diseases.
Collapse
Affiliation(s)
- Qiang Zhang
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Xiu-Fang Zhang
- Department of Pediatrics, Xiang'an Hospital of Xiamen University, Xiamen 361101, Fujian Province, China
| | - Chun-Yan Niu
- Department of Gastroenterology, Nanjing Lishui People's Hospital (Zhongda Hospital Lishui Branch, Southeast University), Nanjing 211200, Jiangsu Province, China
| |
Collapse
|
38
|
Sun LN, Liu XL. Functions of adiponectin signaling in regulating neural plasticity and its application as the therapeutic target to neurological and psychiatric diseases. Rev Neurosci 2020; 30:485-495. [PMID: 30864396 DOI: 10.1515/revneuro-2018-0062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/06/2018] [Indexed: 12/15/2022]
Abstract
Convergent lines of evidence indicate the critical roles of adiponectin in regulating neural functions on different levels. Because of the importance in maintaining neural plasticity including adult neurogenesis and synaptic plasticity, adiponectin has the potential to serve as the treatment targets in therapies of neurological and psychiatric disorders. Hence, systematic review is needed to summarize how adiponectin works in the brain, and how the adiponectin pathway is employed as the treatment method needs to be determined. Moreover, the benefits of adiponectin as the regulator for neural plasticity such as synaptic plasticity and neurogenesis have been supported by many literatures. In the current article, we reviewed the functions of adiponectin in different types of neural plasticity. We also demonstrated the potential value of adiponectin as the treatment target for different types of neurodegenerative and psychiatric disorders. Taken together, this review offers a new insight about adiponectin as the ideal target to develop the new treatment methods against neurodegeneration or psychiatric diseases.
Collapse
Affiliation(s)
- Li-Na Sun
- School of PE and Sport, Beijing Normal University, Beijing 100875, China
| | - Xiao-Li Liu
- School of PE and Sport, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
39
|
Fortin SM, Lipsky RK, Lhamo R, Chen J, Kim E, Borner T, Schmidt HD, Hayes MR. GABA neurons in the nucleus tractus solitarius express GLP-1 receptors and mediate anorectic effects of liraglutide in rats. Sci Transl Med 2020; 12:eaay8071. [PMID: 32132220 PMCID: PMC7211411 DOI: 10.1126/scitranslmed.aay8071] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/22/2019] [Accepted: 02/13/2020] [Indexed: 01/04/2023]
Abstract
The glucagon-like peptide-1 receptor (GLP-1R) agonist liraglutide is approved for the treatment of obesity; however, there is still much to be learned regarding the neuronal sites of action that underlie its suppressive effects on food intake and body weight. Peripherally administered liraglutide in rats acts in part through central GLP-1Rs in both the hypothalamus and the hindbrain. Here, we extend findings supporting a role for hindbrain GLP-1Rs in mediating the anorectic effects of liraglutide in male rats. To dissociate the contribution of GLP-1Rs in the area postrema (AP) and the nucleus tractus solitarius (NTS), we examined the effects of liraglutide in both NTS AAV-shRNA-driven Glp1r knockdown and AP-lesioned animals. Knockdown of NTS GLP-1Rs, but not surgical lesioning of the AP, attenuated the anorectic and body weight-reducing effects of acutely delivered liraglutide. In addition, NTS c-Fos responses were maintained in AP-lesioned animals. Moreover, NTS Glp1r knockdown was sufficient to attenuate the intake- and body weight-reducing effects of chronic daily administered liraglutide over 3 weeks. Development of improved obesity pharmacotherapies requires an understanding of the cellular phenotypes targeted by GLP-1R agonists. Fluorescence in situ hybridization identified Glp1r transcripts in NTS GABAergic neurons, which when inhibited using chemogenetics, attenuated the food intake- and body weight-reducing effects of liraglutide. This work demonstrates the contribution of NTS GLP-1Rs to the anorectic potential of liraglutide and highlights a phenotypically distinct (GABAergic) population of neurons within the NTS that express the GLP-1R and are involved in the mediation of liraglutide signaling.
Collapse
Affiliation(s)
- Samantha M Fortin
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rachele K Lipsky
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rinzin Lhamo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jack Chen
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eun Kim
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tito Borner
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Heath D Schmidt
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
40
|
Zhang ZQ, Hölscher C. GIP has neuroprotective effects in Alzheimer and Parkinson's disease models. Peptides 2020; 125:170184. [PMID: 31705913 DOI: 10.1016/j.peptides.2019.170184] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/17/2022]
Abstract
Glucose-dependent Insulinotropic polypeptide (GIP) is a peptide hormone of the incretin family. It has growth factor properties and can re-activate energy utilization. In progressive neurodegenerative disorders such as Alzheimer's and Parkinson's disease, energy utilization is much reduced, and GIP has the potential to reverse this. Furthermore, GIP can reduce the inflammation response in the brain and reduce levels of pro-inflammatory cytokines. Tests in animal models of Alzheimer's and Parkinson's disease show good neuroprotective effects. In Parkinson's disease models, motor activity is normalized, dopaminergic neurons are protected, synapse numbers and dopamine levels are maintained. Levels of growth factors that are essential for neuronal and synaptic function are increased and alpha-synuclein levels are reduced. The chronic inflammation response and mitochondrial damage is reduced. In Alzheimer's disease models, memory is rescued, synapse numbers and synaptic plasticity in the hippocampus is normalized, amyloid plaque load and the chronic inflammation is reduced. Similar protective effects have been previously reported with analogues of glucagon-like peptide 1 (GLP-1), the sister incretin hormone. First clinical trials show good protective effects in both diseases. Recently, novel dual GLP-1/GIP receptor agonists have been developed. The ability to cross the blood-brain barrier (BBB) is key to their neuroprotective effects. We have developed two dual GLP-1/GIP receptor agonist that have cell penetrating sequences added for better BBB penetration. In direct comparisons, these dual agonists show improved neuroprotection in a mouse model of Parkinson's disease. Therefore, such novel multiple receptor agonists hold great promise as potential treatments for Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Zhen Qiang Zhang
- Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Christian Hölscher
- Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan Province, China.
| |
Collapse
|
41
|
|
42
|
Gad SN, Nofal S, Raafat EM, Ahmed AAE. Lixisenatide Reduced Damage in Hippocampus CA1 Neurons in a Rat Model of Cerebral Ischemia-Reperfusion Possibly Via the ERK/P38 Signaling Pathway. J Mol Neurosci 2020; 70:1026-1037. [PMID: 32040827 DOI: 10.1007/s12031-020-01497-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) is a gut-derived peptide that has various physiological actions. One of its main actions is the regulation of blood glucose level when it is elevated as it potentiates insulin release. It is also known that GLP-1 protects neurons from damage caused by neurodegenerative diseases. Lixisenatide is one of the GLP-1 analogues that has a strong affinity to the GLP-1 receptor. Experimental animal studies have shown that it holds a neuroprotective effect in Parkinson, myocardial, and cerebral ischemic disease animal models. The beneficial effect of lixisenatide on the brain after cerebral ischemia-reperfusion (I/R) is not clarified yet; thus, it needs further explanatory studies. Our research is the first to study the effect of lixisenatide on myeloperoxidase (MPO) and toll-like receptors (TLRs)/mitogen-activated protein kinase (MAPK) pathway in a rat model of cerebral I/R. Lixisenatide with 2 doses 0.7 and 7 nmol/kg was given intraperitoneal in 2 different groups for 14 days; then, the bilateral common carotid artery was occluded for 1 h followed by reperfusion for 1 h. Examination of hippocampus CA1 neurons by Nissl stain showed that the number of intact neurons was elevated in the lixisenatide-treated group related to the control group (I/R group). Lixisenatide exhibited neuroprotection action possibly via downregulation of MPO, TLR2/4, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and pP38 and upregulation of phosphorylated extracellular signal-regulated kinase (pERK1/2); thus, this study gives possible link between lixisenatide and TLR/MAPK pathway following cerebral I/R and supports the use of lixisenatide for neuroprotection against stroke.
Collapse
Affiliation(s)
- Salma N Gad
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt.
| | - Shahira Nofal
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| | - Eman M Raafat
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| | - Amany A E Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| |
Collapse
|
43
|
Yaribeygi H, Maleki M, Sathyapalan T, Jamialahmadi T, Sahebkar A. Anti-inflammatory potentials of incretin-based therapies used in the management of diabetes. Life Sci 2019; 241:117152. [PMID: 31837333 DOI: 10.1016/j.lfs.2019.117152] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/01/2019] [Accepted: 12/07/2019] [Indexed: 12/25/2022]
Abstract
GLP-1 receptor agonists (GLP-1RA) and dipeptidyl peptidase 4 inhibitors (DPP-4i) are two classes of antidiabetic agents used in the management of diabetes based on incretin hormones. There is emerging evidence that they have anti-inflammatory effects. Since most long-term complications of diabetes have a background of chronic inflammation, these agents may be beneficial against diabetic complications not only due to their hypoglycemic potential but also via their anti-inflammatory effects. However, the exact molecular mechanisms by which GLP-1RAs and DPP-4i exert their anti-inflammatory effects are not clearly understood. In this review, we discuss the potential molecular pathways by which these incretin-based therapies exert their anti-inflammatory effects.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Mina Maleki
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, United Kingdom of Great Britain and Northern Ireland
| | - Tannaz Jamialahmadi
- Halal Research Center of IRI, FDA, Tehran, Iran; Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
44
|
Verma N, Despa F. Contributing Factors to Diabetic Brain Injury and Cognitive Decline. Diabetes Metab J 2019; 43:560-567. [PMID: 31694078 PMCID: PMC6834839 DOI: 10.4093/dmj.2019.0153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/02/2019] [Indexed: 01/11/2023] Open
Abstract
The link of diabetes with co-occurring disorders in the brain involves complex and multifactorial pathways. Genetically engineered rodents that express familial Alzheimer's disease-associated mutant forms of amyloid precursor protein and presenilin 1 (PSEN1) genes provided invaluable insights into the mechanisms and consequences of amyloid deposition in the brain. Adding diabetes factors (obesity, insulin impairment) to these animal models to predict success in translation to clinic have proven useful at some extent only. Here, we focus on contributing factors to diabetic brain injury with the aim of identifying appropriate animal models that can be used to mechanistically dissect the pathophysiology of diabetes-associated cognitive dysfunction and how diabetes medications may influence the development and progression of cognitive decline in humans with diabetes.
Collapse
Affiliation(s)
- Nirmal Verma
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Florin Despa
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
45
|
Buchmann N, Fink A, Tegeler C, Demuth I, Doblhammer G, Steinhagen-Thiessen E. Different treatment forms of type II diabetes and the risk of dementia in German health claims data. Acta Diabetol 2019; 56:995-1003. [PMID: 31119454 DOI: 10.1007/s00592-019-01332-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 03/22/2019] [Indexed: 12/11/2022]
Abstract
AIMS The association between type II diabetes (T2D) and increased all-cause dementia risk is well established. However, to date, there is no definite proof that a specific therapy for diabetes can halt a progress of cognitive decline. Therefore, we analyzed a large longitudinal random sample of German health claims data to focus on associations between T2D and dementia and to elucidate the role of different treatment forms of T2D on the risk for dementia. METHODS We used a longitudinal random sample (n = 250,000) of claims data of the largest public sickness fund in Germany, the Allgemeine Ortskrankenkasse (AOK). Dementia was defined as ICD-10 codes G31.0, G31.82, G23.1, F00, F01, F02, F03, and F05, and T2D was defined as E11-E14. We performed Cox proportional hazard models to explore the transition into dementia and to calculate the relative risk of dementia dependent on T2D and different T2D treatment forms. RESULTS All models were adjusted for sex, age, and each patient's history of depression, renal insufficiency, and cardiovascular comorbidities. Non-pharmacologic-treated diabetics showed a 23% increased dementia risk (p < 0.001) and oral ADM-treated diabetics showed a 16% increased risk (p < 0.001). Insulin-dependent diabetics is still the highest dementia risk (40%; p < 0.001) and obesity additionally attenuated this risk (75%; p < 0.001) increased risk. CONCLUSIONS We found that diabetes is an independent risk factor for all-cause dementia. An increased risk for dementia in insulin-dependent and obese subjects with diabetes was evident. Longitudinal studies on the effect of different forms of therapy and weight reduction are needed to verify the results of this study.
Collapse
Affiliation(s)
- Nikolaus Buchmann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Lipid Clinic at the Interdisciplinary Metabolism Center, Berlin, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Anne Fink
- German Center for Neurodegenerative Diseases, Sigmund-Freud-Str. 27, 53127, Bonn, Germany
- Rostock Center for the Study of Demographic Change, Konrad-Zuse-Str.1, 18057, Rostock, Germany
| | - Christina Tegeler
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Lipid Clinic at the Interdisciplinary Metabolism Center, Berlin, Germany
| | - Ilja Demuth
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Lipid Clinic at the Interdisciplinary Metabolism Center, Berlin, Germany
| | - Gabriele Doblhammer
- German Center for Neurodegenerative Diseases, Sigmund-Freud-Str. 27, 53127, Bonn, Germany.
- Rostock Center for the Study of Demographic Change, Konrad-Zuse-Str.1, 18057, Rostock, Germany.
- Institute for Sociology and Demography, University Rostock, Ulmenstr. 69, 18057, Rostock, Germany.
| | - Elisabeth Steinhagen-Thiessen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Lipid Clinic at the Interdisciplinary Metabolism Center, Berlin, Germany
| |
Collapse
|
46
|
Dong D, Xie J, Wang J. Neuroprotective Effects of Brain-Gut Peptides: A Potential Therapy for Parkinson's Disease. Neurosci Bull 2019; 35:1085-1096. [PMID: 31286411 DOI: 10.1007/s12264-019-00407-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 04/29/2019] [Indexed: 12/16/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and is typically associated with progressive motor and non-motor dysfunctions. Currently, dopamine replacement therapy is mainly used to relieve the motor symptoms, while its long-term application can lead to various complications and does not cure the disease. Numerous studies have demonstrated that many brain-gut peptides have neuroprotective effects in vivo and in vitro, and may be a promising treatment for PD. In recent years, some progress has been made in studies on the neuroprotective effects of some newly-discovered brain-gut peptides, such as glucagon-like peptide 1, pituitary adenylate cyclase activating polypeptide, nesfatin-1, and ghrelin. However, there is still no systematic review on the neuroprotective effects common to these peptides. Thus, here we review the neuroprotective effects and the associated mechanisms of these four peptides, as well as other brain-gut peptides related to PD, in the hope of providing new ideas for the treatment of PD and related clinical research.
Collapse
Affiliation(s)
- Dong Dong
- Department of Physiology and Pathophysiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China
| | - Junxia Xie
- Department of Physiology and Pathophysiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China.
| | - Jun Wang
- Department of Physiology and Pathophysiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
47
|
Hölscher C. Insulin Signaling Impairment in the Brain as a Risk Factor in Alzheimer's Disease. Front Aging Neurosci 2019; 11:88. [PMID: 31068799 PMCID: PMC6491455 DOI: 10.3389/fnagi.2019.00088] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 04/03/2019] [Indexed: 12/14/2022] Open
Abstract
Type 2 diabetes is a risk factor for developing Alzheimer’s disease (AD). The underlying mechanism that links up the two conditions seems to be the de-sensitization of insulin signaling. In patients with AD, insulin signaling was found to be de-sensitized in the brain, even if they did not have diabetes. Insulin is an important growth factor that regulates cell growth, energy utilization, mitochondrial function and replacement, autophagy, oxidative stress management, synaptic plasticity, and cognitive function. Insulin desensitization, therefore, can enhance the risk of developing neurological disorders in later life. Other risk factors, such as high blood pressure or brain injury, also enhance the likelihood of developing AD. All these risk factors have one thing in common – they induce a chronic inflammation response in the brain. Pro-inflammatory cytokines block growth factor signaling and enhance oxidative stress. The underlying molecular processes for this are described in the review. Treatments to re-sensitize insulin signaling in the brain are also described, such as nasal insulin tests in AD patients, or treatments with re-sensitizing hormones, such as leptin, ghrelin, glucagon-like peptide 1 (GLP-1),and glucose-dependent insulinotropic polypeptide (GIP). The first clinical trials show promising results and are a proof of concept that utilizing such treatments is valid.
Collapse
Affiliation(s)
- Christian Hölscher
- Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
48
|
Liu W, Liu J, Huang Z, Cui Z, Li L, Liu W, Qi Z. Possible role of GLP-1 in antidepressant effects of metformin and exercise in CUMS mice. J Affect Disord 2019; 246:486-497. [PMID: 30599373 DOI: 10.1016/j.jad.2018.12.112] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 12/18/2018] [Accepted: 12/24/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Both depression itself and antidepressant medication have been reported to be significantly related to the risk of type 2 diabetes mellitus (T2DM). Glucagon-like peptide-1 (GLP-1), a treatment target for T2DM, has a neuroprotective effect. As an enhancer and sensitiser of GLP-1, metformin has been reported to be safe for the neurodevelopment. The present study aimed to determine whether and how GLP-1 mediates antidepressant effects of metformin and exercise in mice. METHODS Male C57BL/6 mice were exposed to chronic unpredictable mild stress (CUMS) for 8 weeks. From the 4th week, CUMS mice were subjected to oral metformin treatment and/or treadmill running. A videocomputerized tracking system was used to record behaviors of mice for a 5-min session. ELISA, western blotting and immunohistochemistry were used to examine serum protein concentrations, protein levels in whole hippocampus, protein distribution and expression in dorsal and ventral hippocampus, respectively. RESULTS Our results supported the validity of metformin as a useful antidepressant; moreover, treadmill running favored metformin effects on exploratory behaviors and serum corticosterone levels. CUMS reduced GLP-1 protein levels and phosphorylation levels of extracellular signal-regulated kinase 1/2 (ERK1/2), but increased protein levels of B-cell lymphoma 2-associated X-protein (BAX) in mice hippocampus. All these changes were restored by both single and combined treatment with metformin and exercise. LIMITATIONS We did not establish a causal relationship between GLP-1 expression and related signaling, using GLP-1 agonist and antagonist or knockout techniques. CONCLUSIONS Our findings have demonstrated that protein levels of pERK and BAX may be relevant to the role of GLP-1 in antidepressant effects of metformin and exercise, which may provide a novel topic for future clinical research.
Collapse
Affiliation(s)
- Weina Liu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China.
| | - Jiatong Liu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China
| | - Zhuochun Huang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China
| | - Zhiming Cui
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China
| | - Lingxia Li
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China
| | - Wenbin Liu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China
| | - Zhengtang Qi
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
49
|
Exendin-4 improves behaviorial deficits via GLP-1/GLP-1R signaling following partial hepatectomy. Brain Res 2019; 1706:116-124. [DOI: 10.1016/j.brainres.2018.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 11/03/2018] [Accepted: 11/03/2018] [Indexed: 02/07/2023]
|
50
|
Zhu HL, Liu ZP, Yang WY, Dong DW, Zhao Y, Yang B, Huang LA, Zhang YS, Xu AD. Liraglutide Ameliorates β-Amyloid Deposits and Secondary Damage in the Ipsilateral Thalamus and Sensory Deficits After Focal Cerebral Infarction in Rats. Front Neurosci 2018; 12:962. [PMID: 30618584 PMCID: PMC6304750 DOI: 10.3389/fnins.2018.00962] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022] Open
Abstract
Focal cerebral infarction causes β-amyloid (Aβ) deposition and secondary neuronal degeneration in the ipsilateral thalamus. Thalamus is the subcortical center of sensory, the damage of thalamus could cause sensory deficits. The present study aimed to investigate the protective effects of liraglutide, a long-acting glucagon-like peptide-1 (GLP)-1 receptor agonist, on Aβ deposits and secondary damage in the ipsilateral thalamus after focal cerebral infarction. In addition, this study was conducted to investigate whether liraglutide could improve sensory function after focal cerebral infarction. Forty-two male Sprague–Dawley rats were subjected to distal middle cerebral artery occlusion (MCAO) and then randomly divided into liraglutide and vehicle groups, and 14 sham-operated rats as control. At 1 h after MCAO, rats in the liraglutide and vehicle groups were subcutaneously injected with liraglutide (100 μg/kg/d) and isopyknic vehicle, respectively, once a day for 7 days. Sensory function and secondary thalamic damage were assessed using adhesive-removal test and Nissl staining and immunostaining, respectively, at 7 days after MCAO. Terminal deoxynucleotidyl transferase 2’-deoxyuridine 5’-triphosphate nick end labeling and Western blot were used to detect neuronal apoptosis. The results showed that liraglutide improved sensory deficit compared to the controls. Liraglutide treatment significantly reduced Aβ deposition compared with the vehicle treatment. Liraglutide treatment decreased the neuronal loss, astroglial and microglial activation, and apoptosis compared with the vehicle treatment. Liraglutide significantly down-regulated the expression of Bcl-2 and up-regulated that of Bax in the ipsilateral thalamus compared with the vehicle group. These results suggest that liraglutide ameliorates the deposition of Aβ and secondary damage in the ipsilateral thalamus, potentially contributing to improve sensory deficit after focal cerebral infarction.
Collapse
Affiliation(s)
- Hui-Li Zhu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Zhang-Pei Liu
- Clinical Neuroscience Institute, Jinan University, Guangzhou, China.,Department of Neurology, Stroke Center, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Wan-Yong Yang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Da-Wei Dong
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Ying Zhao
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Bing Yang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Li-An Huang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Yu-Sheng Zhang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - An-Ding Xu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| |
Collapse
|