1
|
Duranti E, Villa C. Insights into Dysregulated Neurological Biomarkers in Cancer. Cancers (Basel) 2024; 16:2680. [PMID: 39123408 PMCID: PMC11312413 DOI: 10.3390/cancers16152680] [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: 06/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
The link between neurodegenerative diseases (NDs) and cancer has generated greater interest in biomedical research, with decades of global studies investigating neurodegenerative biomarkers in cancer to better understand possible connections. Tau, amyloid-β, α-synuclein, SOD1, TDP-43, and other proteins associated with nervous system diseases have also been identified in various types of solid and malignant tumors, suggesting a potential overlap in pathological processes. In this review, we aim to provide an overview of current evidence on the role of these proteins in cancer, specifically examining their effects on cell proliferation, apoptosis, chemoresistance, and tumor progression. Additionally, we discuss the diagnostic and therapeutic implications of this interconnection, emphasizing the importance of further research to completely comprehend the clinical implications of these proteins in tumors. Finally, we explore the challenges and opportunities in targeting these proteins for the development of new targeted anticancer therapies, providing insight into how to integrate knowledge of NDs in oncology research.
Collapse
Affiliation(s)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
2
|
Strekalova T, Svirin E, Gorlova A, Sheveleva E, Burova A, Khairetdinova A, Sitdikova K, Zakharova E, Dudchenko AM, Lyundup A, Morozov S. Resilience and Vulnerability to Stress-Induced Anhedonia: Unveiling Brain Gene Expression and Mitochondrial Dynamics in a Mouse Chronic Stress Depression Model. Biomolecules 2023; 13:1782. [PMID: 38136653 PMCID: PMC10741640 DOI: 10.3390/biom13121782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The role of altered brain mitochondrial regulation in psychiatric pathologies, including Major Depressive Disorder (MDD), has attracted increasing attention. Aberrant mitochondrial functions were suggested to underlie distinct inter-individual vulnerability to stress-related MDD syndrome. In this context, insulin receptor sensitizers (IRSs) that regulate brain metabolism have become a focus of recent research, as their use in pre-clinical studies can help to elucidate the role of mitochondrial dynamics in this disorder and contribute to the development of new antidepressant treatment. Here, following 2-week chronic mild stress (CMS) using predation, social defeat, and restraint, MDD-related behaviour and brain molecular markers have been investigated along with the hippocampus-dependent performance and emotionality in mice that received the IRS dicholine succinate (DS). In a sucrose test, mice were studied for the key feature of MDD, a decreased sensitivity to reward, called anhedonia. Based on this test, animals were assigned to anhedonic and resilient-to-stress-induced-anhedonia groups, using a previously established criterion of a decrease in sucrose preference below 65%. Such assignment was based on the fact that none of control, non-stressed animals displayed sucrose preference that would be smaller than this value. DS-treated stressed mice displayed ameliorated behaviours in a battery of assays: sucrose preference, coat state, the Y-maze, the marble test, tail suspension, and nest building. CMS-vulnerable mice exhibited overexpression of the inflammatory markers Il-1β, tnf, and Cox-1, as well as 5-htt and 5-ht2a-R, in various brain regions. The alterations in hippocampal gene expression were the closest to clinical findings and were studied further. DS-treated, stressed mice showed normalised hippocampal expression of the plasticity markers Camk4, Camk2, Pka, Adcy1, Creb-ar, Nmda-2r-ar, and Nmda-2r-s. DS-treated and non-treated stressed mice who were resilient or vulnerable to anhedonia were compared for hippocampal mitochondrial pathway regulation using Illumina profiling. Resilient mice revealed overexpression of the mitochondrial complexes NADH dehydrogenase, succinate dehydrogenase, cytochrome bc1, cytochrome c oxidase, F-type and V-type ATPases, and inorganic pyrophosphatase, which were decreased in anhedonic mice. DS partially normalised the expression of both ATPases. We conclude that hippocampal reduction in ATP synthesis is associated with anhedonia and pro-inflammatory brain changes that are ameliorated by DS.
Collapse
Affiliation(s)
- Tatyana Strekalova
- Division of Molecular Psychiatry, Center of Mental Health, University of Hospital Würzburg, 97080 Wuerzburg, Germany
| | - Evgeniy Svirin
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Anna Gorlova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Elizaveta Sheveleva
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Alisa Burova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Adel Khairetdinova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Kseniia Sitdikova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Elena Zakharova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Alexander M. Dudchenko
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Aleksey Lyundup
- Endocrinology Research Centre, Dmitry Ulyanov St. 19, Moscow 117036, Russia;
- Research and Education Resource Center, Peoples Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russia
| | - Sergey Morozov
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| |
Collapse
|
3
|
Gence L, Fernezelian D, Meilhac O, Rastegar S, Bascands JL, Diotel N. Insulin signaling promotes neurogenesis in the brain of adult zebrafish. J Comp Neurol 2023; 531:1812-1827. [PMID: 37750011 DOI: 10.1002/cne.25542] [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: 03/02/2023] [Revised: 06/20/2023] [Accepted: 09/08/2023] [Indexed: 09/27/2023]
Abstract
Insulin is a peptide hormone that plays a central role in the regulation of circulating blood glucose in vertebrates, including zebrafish. Increasing evidence has demonstrated the important role of insulin in many brain functions. In zebrafish, two insulin receptor genes (insra and insrb) have been identified. However, their biodistribution in the adult brain as well as their cell-specific expression pattern has not been well described. Using gene expression analysis, in situ hybridization and transgenic fish, we confirmed the expression of insra, insrb, and irs1 (insulin receptor substrate 1, the downstream effector of insulin receptor) in the brain of adult zebrafish and characterized their specific expression in neurons and neural stem cells (radial glia). After demonstrating that intracerebroventricular (ICV) injection resulted in the diffusion of the injected solution within the ventricular system, we analyzed the effect of insulin ICV injection on neurogenesis. We showed that insulin promotes ventricular cell proliferation 24 h postinjection. This neurogenic effect appeared to be independent of neuroinflammatory processes. Also, after a mechanical telencephalic stab-wound injury, we highlighted the overexpression of irs1 gene 5 days postlesion notably in the ventricular zone where radial glial cells (RGCs) are localized, suggesting key roles of insulin signaling in regenerative processes. Finally, our results reinforced the expression of insulin-related proteins in the brain of adult zebrafish, highlighting the potential role of insulin signaling on neurogenesis.
Collapse
Affiliation(s)
- Laura Gence
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis, La Réunion, France
| | - Danielle Fernezelian
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis, La Réunion, France
| | - Olivier Meilhac
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis, La Réunion, France
- CHU de La Réunion, Saint-Denis, La Réunion, France
| | - Sepand Rastegar
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Jean-Loup Bascands
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis, La Réunion, France
| | - Nicolas Diotel
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis, La Réunion, France
| |
Collapse
|
4
|
Chen ZG, Shi X, Zhang XX, Yang FF, Li KR, Fang Q, Cao C, Chen XH, Peng Y. Neuron-secreted NLGN3 ameliorates ischemic brain injury via activating Gαi1/3-Akt signaling. Cell Death Dis 2023; 14:700. [PMID: 37880221 PMCID: PMC10600254 DOI: 10.1038/s41419-023-06219-8] [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: 09/05/2022] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023]
Abstract
We here tested the potential activity and the underlying mechanisms of neuroligin-3 (NLGN3) against ischemia-reperfusion-induced neuronal cell injury. In SH-SY5Y neuronal cells and primary murine cortical neurons, NLGN3 activated Akt-mTOR and Erk signalings, and inhibited oxygen and glucose deprivation (OGD)/re-oxygenation (OGD/R)-induced cytotoxicity. Akt activation was required for NLGN3-induced neuroprotection. Gαi1/3 mediated NLGN3-induced downstream signaling activation. NLGN3-induced Akt-S6K1 activation was largely inhibited by Gαi1/3 silencing or knockout. Significantly, NLGN3-induced neuroprotection against OGD/R was almost abolished by Gαi1/3 silencing or knockout. In vivo, the middle cerebral artery occlusion (MCAO) procedure induced NLGN3 cleavage and secretion, and increased its expression and Akt activation in mouse brain tissues. ADAM10 (A Disintegrin and Metalloproteinase 10) inhibition blocked MCAO-induced NLGN3 cleavage and secretion, exacerbating ischemic brain injury in mice. Neuronal silencing of NLGN3 or Gαi1/3 in mice also inhibited Akt activation and intensified MCAO-induced ischemic brain injury. Conversely, neuronal overexpression of NLGN3 increased Akt activation and alleviated MCAO-induced ischemic brain injury. Together, NLGN3 activates Gαi1/3-Akt signaling to protect neuronal cells from ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Zhi-Guo Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin Shi
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Xian-Xian Zhang
- Department of Neurology, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, China
| | - Fang-Fang Yang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Ke-Ran Li
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Cong Cao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China.
| | - Xiong-Hui Chen
- Department of Emergency Surgery, First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Ya Peng
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| |
Collapse
|
5
|
Alberry B, Silveira PP. Brain insulin signaling as a potential mediator of early life adversity effects on physical and mental health. Neurosci Biobehav Rev 2023; 153:105350. [PMID: 37544390 DOI: 10.1016/j.neubiorev.2023.105350] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
In numerous brain structures, insulin signaling modulates the homeostatic processes, sensitivity to reward pathways, executive function, memory, and cognition. Through human studies and animal models, mounting evidence implicates central insulin signaling in the metabolic, physiological, and psychological consequences of early life adversity. In this review, we describe the consequences of early life adversity in the brain where insulin signaling is a key factor and how insulin may moderate the effects of adversity on psychiatric and cardio-metabolic health outcomes. Further understanding of how early life adversity and insulin signaling impact specific brain regions and mental and physical health outcomes will assist in prevention, diagnosis, and potential intervention following early life adversity.
Collapse
Affiliation(s)
- Bonnie Alberry
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Patricia Pelufo Silveira
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
| |
Collapse
|
6
|
Ezkurdia A, Ramírez MJ, Solas M. Metabolic Syndrome as a Risk Factor for Alzheimer's Disease: A Focus on Insulin Resistance. Int J Mol Sci 2023; 24:ijms24054354. [PMID: 36901787 PMCID: PMC10001958 DOI: 10.3390/ijms24054354] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Alzheimer's disease (AD) is the main type of dementia and is a disease with a profound socioeconomic burden due to the lack of effective treatment. In addition to genetics and environmental factors, AD is highly associated with metabolic syndrome, defined as the combination of hypertension, hyperlipidemia, obesity and type 2 diabetes mellitus (T2DM). Among these risk factors, the connection between AD and T2DM has been deeply studied. It has been suggested that the mechanism linking both conditions is insulin resistance. Insulin is an important hormone that regulates not only peripheral energy homeostasis but also brain functions, such as cognition. Insulin desensitization, therefore, could impact normal brain function increasing the risk of developing neurodegenerative disorders in later life. Paradoxically, it has been demonstrated that decreased neuronal insulin signalling can also have a protective role in aging and protein-aggregation-associated diseases, as is the case in AD. This controversy is fed by studies focused on neuronal insulin signalling. However, the role of insulin action on other brain cell types, such as astrocytes, is still unexplored. Therefore, it is worthwhile exploring the involvement of the astrocytic insulin receptor in cognition, as well as in the onset and/or development of AD.
Collapse
Affiliation(s)
- Amaia Ezkurdia
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - María J. Ramírez
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Maite Solas
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Correspondence:
| |
Collapse
|
7
|
Cullinane PW, de Pablo Fernandez E, König A, Outeiro TF, Jaunmuktane Z, Warner TT. Type 2 Diabetes and Parkinson's Disease: A Focused Review of Current Concepts. Mov Disord 2023; 38:162-177. [PMID: 36567671 DOI: 10.1002/mds.29298] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/25/2022] [Accepted: 11/15/2022] [Indexed: 12/27/2022] Open
Abstract
Highly reproducible epidemiological evidence shows that type 2 diabetes (T2D) increases the risk and rate of progression of Parkinson's disease (PD), and crucially, the repurposing of certain antidiabetic medications for the treatment of PD has shown early promise in clinical trials, suggesting that the effects of T2D on PD pathogenesis may be modifiable. The high prevalence of T2D means that a significant proportion of patients with PD may benefit from personalized antidiabetic treatment approaches that also confer neuroprotective benefits. Therefore, there is an immediate need to better understand the mechanistic relation between these conditions and the specific molecular pathways affected by T2D in the brain. Although there is considerable evidence that processes such as insulin signaling, mitochondrial function, autophagy, and inflammation are involved in the pathogenesis of both PD and T2D, the primary aim of this review is to highlight the evidence showing that T2D-associated dysregulation of these pathways occurs not only in the periphery but also in the brain and how this may facilitate neurodegeneration in PD. We also discuss the challenges involved in disentangling the complex relationship between T2D, insulin resistance, and PD, as well as important questions for further research. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Patrick W Cullinane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Eduardo de Pablo Fernandez
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Annekatrin König
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom.,Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Zane Jaunmuktane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, London, United Kingdom.,Queen Square Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Thomas T Warner
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Queen Square Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
8
|
Cummings J, Ortiz A, Castellino J, Kinney J. Diabetes: Risk factor and translational therapeutic implications for Alzheimer's disease. Eur J Neurosci 2022; 56:5727-5757. [PMID: 35128745 PMCID: PMC9393901 DOI: 10.1111/ejn.15619] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 12/31/2022]
Abstract
Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) commonly co-occur. T2DM increases the risk for AD by approximately twofold. Animal models provide one means of interrogating the relationship of T2DM to AD and investigating brain insulin resistance in the pathophysiology of AD. Animal models show that persistent hyperglycaemia results in chronic low-grade inflammation that may contribute to the development of neuroinflammation and accelerate the pathobiology of AD. Epidemiological studies suggest that patients with T2DM who received treatment with specific anti-diabetic agents have a decreased risk for the occurrence of AD and all-cause dementia. Agents such as metformin ameliorate T2DM and may have other important systemic effects that lower the risk of AD. Glucagon-like peptide 1 (GLP-1) agonists have been associated with a decreased risk for AD in patients with T2DM. Both insulin and non-insulin anti-diabetic treatments have been evaluated for the treatment of AD in clinical trials. In most cases, patients included in the trials have clinical features of AD but do not have T2DM. Many of the trials were conducted prior to the use of diagnostic biomarkers for AD. Trials have had a wide range of durations and population sizes. Many of the agents used to treat T2DM do not cross the blood brain barrier, and the effects are posited to occur via lowering of peripheral hyperglycaemia and reduction of peripheral and central inflammation. Clinical trials of anti-diabetic agents to treat AD are ongoing and will provide insight into the therapeutic utility of these agents.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Chambers‐Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | - Andrew Ortiz
- Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | | | - Jefferson Kinney
- Chambers‐Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA,Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| |
Collapse
|
9
|
The effect of chronic exposure to metformin in a new type-2 diabetic NONcNZO10/LtJ mouse model of stroke. Pharmacol Rep 2022; 74:696-708. [PMID: 35792967 DOI: 10.1007/s43440-022-00382-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/18/2022] [Accepted: 06/10/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND Diabetes is an independent risk factor of stroke and previous studies have confirmed that diabetic patients and animals experience poorer clinical outcomes following stroke. In this study, we aim to determine the effect of chronic exposure of the first-line antidiabetic agent, metformin, to restore euglycemia and to impact brain cell death following stroke in a new type-2 diabetes, NONcNZO10/LtJ (RCS-10) mouse model of stroke. METHODS Male RCS-10 mice received a moderate (11%) fat diet post-weaning, at 4 weeks of age, and became diabetic by 12-14 weeks, thus resembling human maturity-onset diabetes. The mice received either metformin or vehicle for 4 weeks before undergoing a hypoxic/ischemic (HI) insult. Blood samples were collected pre-, post-treatment, and post HI for glucose and lipid measurements, and brains were analyzed for infarct size, glial activation, neuronal cell death, and metformin-mediated adenosine monophosphate-activated protein kinase (AMPK) signaling at 48 h post HI. RESULTS Pretreatment with metformin maintained euglycemia for 4 weeks but did not change body weight or lipid profile. Metformin treatment significantly enhanced the microglial Bfl-1 mRNA expression and showed a non-significant increase in GFAP mRNA, however, GFAP protein levels were reduced. Metformin treatment slightly increased neuronal NeuN and MAP-2 protein levels and significantly reduced overall mortality post HI but did not elicit any significant change in infarct size. CONCLUSION The study suggests that the prolonged effect of metformin-induced euglycemia promoted the microglial activation, reduced neuronal cell death, and improved the overall survival following stroke, without any change in infarct size.
Collapse
|
10
|
Examples of Inverse Comorbidity between Cancer and Neurodegenerative Diseases: A Possible Role for Noncoding RNA. Cells 2022; 11:cells11121930. [PMID: 35741059 PMCID: PMC9221903 DOI: 10.3390/cells11121930] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/25/2022] [Accepted: 06/13/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer is one of the most common causes of death; in parallel, the incidence and prevalence of central nervous system diseases are equally high. Among neurodegenerative diseases, Alzheimer’s dementia is the most common, while Parkinson’s disease (PD) is the second most frequent neurodegenerative disease. There is a significant amount of evidence on the complex biological connection between cancer and neurodegeneration. Noncoding RNAs (ncRNAs) are defined as transcribed nucleotides that perform a variety of regulatory functions. The mechanisms by which ncRNAs exert their functions are numerous and involve every aspect of cellular life. The same ncRNA can act in multiple ways, leading to different outcomes; in fact, a single ncRNA can participate in the pathogenesis of more than one disease—even if these seem very different, as cancer and neurodegenerative disorders are. The ncRNA activates specific pathways leading to one or the other clinical phenotype, sometimes with obvious mechanisms of inverse comorbidity. We aimed to collect from the existing literature examples of inverse comorbidity in which ncRNAs seem to play a key role. We also investigated the example of mir-519a-3p, and one of its target genes Poly (ADP-ribose) polymerase 1, for the inverse comorbidity mechanism between some cancers and PD. We believe it is very important to study the inverse comorbidity relationship between cancer and neurodegenerative diseases because it will help us to better assess these two major areas of human disease.
Collapse
|
11
|
El Idrissi A, Alonso ADC. Pathological Human Tau Induces Alterations in the Brain Insulin Signaling Cascade. Front Neurosci 2022; 16:805046. [PMID: 35264925 PMCID: PMC8899662 DOI: 10.3389/fnins.2022.805046] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/17/2022] [Indexed: 11/25/2022] Open
Abstract
The process of neurodegeneration in Alzheimer's disease has been associated with a disruption of insulin signaling cascade in neurons, and to insulin resistance. T2DM correlates with Alzheimer's disease, but mechanisms of interaction are unknown. We have developed a mouse model of tau induced neurodegeneration expressing pseudo-phosphorylated tau [Pathological Human Tau (PH-Tau)] in neurons. This model (PH-Tau-Tg) recapitulated cognitive decline and neurodegeneration observed in AD. In this study we examined if expression of PH-Tau could affect neuronal excitability and insulin receptor signaling. Neuronal excitability was investigated using intracerebral recordings of extracellular field potentials from prefrontal cortex after insulin and kainic acid (KA) injection. Analysis of baseline recordings indicated an increased excitability of PH-Tau-Tg as evidenced by higher spectrum densities (PSDs) of high frequencies brain waves. Injection of insulin (1IU, s.c) led to a decrease of fast ripples PSDs, more pronounced in PH-Tau-Tg mice than controls. Subsequent injection of kainic acid (KA, 5 mg/kg, s.c) led to significant increase in firing rate, amplitude of extracellular field potentials and PSDs of high frequency brain waves in control mice only. To further investigate the role of insulin in PH-Tau-Tg mice, we subjected mice to a glucose tolerance test. We found that PH-Tau-Tg mice were significantly hyperglycemic prior to glucose injection. Interestingly, the PH-Tau-Tg mice showed a moderate increase at 30 min due to the higher baseline, indicating a low sensitivity of insulin receptor in these mice. This is consistent with increased levels of activated insulin receptors in the brain and the inhibitory effect of insulin on ictal activity post KA injection in PH-Tau-Tg mice. We suggest that these mice have reduced insulin sensitivity (hyperglycemia) and as a compensatory mechanism there is overactivation/expression of insulin receptor in the brain rendering neuronal circuits resistant to seizure induction after injection of insulin. These data indicate that insulin signal transduction pathway is altered in PH-Tau-Tg mice, and that injection of exogenous insulin reduces hypersynchronous bursting activity of field potentials recorded from cortical neuronal circuits. We propose that the appearance of abnormal tau might potentiate the toxic environment by interfering with the insulin signaling cascade in the brain of patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Abdeslem El Idrissi
- Department of Biology and Center for Developmental Neuroscience, College of Staten Island, The City University of New York, New York, NY, United States,Biology Program, The Graduate Center, The City University of New York, New York, NY, United States,*Correspondence: Abdeslem El Idrissi,
| | - Alejandra del Carmen Alonso
- Department of Biology and Center for Developmental Neuroscience, College of Staten Island, The City University of New York, New York, NY, United States,Biology Program, The Graduate Center, The City University of New York, New York, NY, United States,Alejandra del Carmen Alonso,
| |
Collapse
|
12
|
Bhattacharya A, Choi WWY, Muffat J, Li Y. Modeling Developmental Brain Diseases Using Human Pluripotent Stem Cells-Derived Brain Organoids - Progress and Perspective. J Mol Biol 2021; 434:167386. [PMID: 34883115 DOI: 10.1016/j.jmb.2021.167386] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023]
Abstract
Developmental brain diseases encompass a group of conditions resulting from genetic or environmental perturbations during early development. Despite the increased research attention in recent years following recognition of the prevalence of these diseases, there is still a significant lack of knowledge of their etiology and treatment options. The genetic and clinical heterogeneity of these diseases, in addition to the limitations of experimental animal models, contribute to this difficulty. In this regard, the advent of brain organoid technology has provided a new means to study the cause and progression of developmental brain diseases in vitro. Derived from human pluripotent stem cells, brain organoids have been shown to recapitulate key developmental milestones of the early human brain. Combined with technological advancements in genome editing, tissue engineering, electrophysiology, and multi-omics analysis, brain organoids have expanded the frontiers of human neurobiology, providing valuable insight into the cellular and molecular mechanisms of normal and pathological brain development. This review will summarize the current progress of applying brain organoids to model human developmental brain diseases and discuss the challenges that need to be overcome to further advance their utility.
Collapse
Affiliation(s)
- Afrin Bhattacharya
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Wendy W Y Choi
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Julien Muffat
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Program in Neurosciences and Mental Health, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Yun Li
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
13
|
Behl T, Arora A, Sehgal A, Singh S, Sharma N, Bhatia S, Al-Harrasi A, Bungau S, Mostafavi E. Molecular and Biochemical Pathways Encompassing Diabetes Mellitus and Dementia. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:542-556. [PMID: 34758720 DOI: 10.2174/1871527320666211110115257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/25/2021] [Accepted: 09/15/2021] [Indexed: 11/22/2022]
Abstract
Diabetes mellitus is a major metabolic disorder that has now emerged as an epidemic, and it affects the brain through an array of pathways. Diabetes mellitus patients can develop pathological changes in the brain, which eventually take the shape of mild cognitive impairment progressing to Alzheimer's Disease. A number of preclinical and clinical studies demonstrate this fact, and it comes out to be those molecular pathways such as amyloidogenesis, oxidative stress, inflammation, and impaired insulin signaling are identical in diabetes mellitus and dementia. However, the critical player involved in the vicious cycle of diabetes mellitus and dementia is insulin, whose signaling, when impaired in diabetes mellitus (both type 1 and 2), leads to a decline in cognition, although other pathways are also essential contributors. Moreover, it is not only that diabetes mellitus patients indicate cognitive decline at a later stage; many Alzheimer's Disease patients also reflect symptoms of diabetes mellitus, thus creating a vicious cycle inculcating a web of complex molecular mechanisms and hence categorizing Alzheimer's Disease as 'brain diabetes'. Thus, it is practical to suggest that anti-diabetic drugs are beneficial in Alzheimer's Disease; but only smaller trials, not the larger ones, have showcased positive outcomes mainly because of the late onset of therapy. Therefore, it is extremely important to develop more of such molecules that target insulin in dementia patients along with such methods that diagnose impaired insulin signaling and the associated cognitive decline so that early therapy may be initiated and the progression of the disease be prevented.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Arpita Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Saurabh Bhatia
- Amity Institute of Pharmacy, Amity University, Haryana. India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa. Oman
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea. Romania
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA. United States
| |
Collapse
|
14
|
Mor ME, Harvey A, Familari M, St Clair-Glover M, Viventi S, de Iongh RU, Cameron FJ, Dottori M. Neural differentiation medium for human pluripotent stem cells to model physiological glucose levels in human brain. Brain Res Bull 2021; 173:141-149. [PMID: 34022288 DOI: 10.1016/j.brainresbull.2021.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 05/05/2021] [Accepted: 05/17/2021] [Indexed: 12/30/2022]
Abstract
Cortical neurospheres (NSPs) derived from human pluripotent stem cells (hPSC), have proven to be a successful platform to investigate human brain development and neuro-related diseases. Currently, many of the standard hPSC neural differentiation media, use concentrations of glucose (approximately 17.5-25 mM) and insulin (approximately 3.2 μM) that are much greater than the physiological concentrations found in the human brain. These culture conditions make it difficult to analyse perturbations of glucose or insulin on neuronal development and differentiation. We established a new hPSC neural differentiation medium that incorporated physiological brain concentrations of glucose (2.5 mM) and significantly reduced insulin levels (0.86 μM). This medium supported hPSC neural induction and formation of cortical NSPs. The revised hPSC neural differentiation medium, may provide an improved platform to model brain development and to investigate neural differentiation signalling pathways impacted by abnormal glucose and insulin levels.
Collapse
Affiliation(s)
- Michal E Mor
- Department of Anatomy & Physiology, University of Melbourne, Australia
| | | | - Mary Familari
- School of BioSciences, University of Melbourne, Australia
| | - Mitchell St Clair-Glover
- Illawarra Health and Medical Research Institute, Molecular Horizons, University of Wollongong, Australia
| | - Serena Viventi
- The Florey Institute of Neuroscience and Mental Health, Australia
| | - Robb U de Iongh
- Department of Anatomy & Physiology, University of Melbourne, Australia
| | - Fergus J Cameron
- Murdoch Children's Research Institute, The Royal Children's Hospital, Australia; Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Australia
| | - Mirella Dottori
- Department of Anatomy & Physiology, University of Melbourne, Australia; Illawarra Health and Medical Research Institute, Molecular Horizons, University of Wollongong, Australia; Department of Biomedical Engineering, University of Melbourne, Australia.
| |
Collapse
|
15
|
Kuboki A, Kikuta S, Otori N, Kojima H, Matsumoto I, Reisert J, Yamasoba T. Insulin-Dependent Maturation of Newly Generated Olfactory Sensory Neurons after Injury. eNeuro 2021; 8:ENEURO.0168-21.2021. [PMID: 33906971 PMCID: PMC8143024 DOI: 10.1523/eneuro.0168-21.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/24/2022] Open
Abstract
Loss of olfactory sensory neurons (OSNs) after injury to the olfactory epithelium (OE) triggers the generation of OSNs that are incorporated into olfactory circuits to restore olfactory sensory perception. This study addresses how insulin receptor-mediated signaling affects the functional recovery of OSNs after OE injury. Insulin levels were reduced in mice by ablating the pancreatic β cells via streptozotocin (STZ) injections. These STZ-induced diabetic and control mice were then intraperitoneally injected with the olfactotoxic drug methimazole to selectively ablate OSNs. The OE of diabetic and control mice regenerated similarly until day 14 after injury. Thereafter, the OE of diabetic mice contained fewer mature and more apoptotic OSNs than control mice. Functionally, diabetic mice showed reduced electro-olfactogram (EOG) responses and their olfactory bulbs (OBs) had fewer c-Fos-active cells following odor stimulation, as well as performed worse in an odor-guided task compared with control mice. Insulin administered intranasally during days 8-13 after injury was sufficient to rescue recovery of OSNs in diabetic mice compared with control levels, while insulin administration between days 1 and 6 did not. During this critical time window on days 8-13 after injury, insulin receptors are highly expressed and intranasal application of an insulin receptor antagonist inhibits regeneration. Furthermore, an insulin-enriched environment could facilitate regeneration even in non-diabetic mice. These results indicate that insulin facilitates the regeneration of OSNs after injury and suggest a critical stage during recovery (8-13 d after injury) during which the maturation of newly generated OSNs is highly dependent on and promoted by insulin.
Collapse
Affiliation(s)
- Akihito Kuboki
- Department of Otolaryngology, Jikei University School of Medicine, Tokyo 105-8461, Japan
- Monell Chemical Senses Center, Philadelphia, PA 19104
| | - Shu Kikuta
- Department of Otolaryngology, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Nobuyoshi Otori
- Department of Otolaryngology, Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Hiromi Kojima
- Department of Otolaryngology, Jikei University School of Medicine, Tokyo 105-8461, Japan
| | | | | | - Tatsuya Yamasoba
- Department of Otolaryngology, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
16
|
Manaserh IH, Maly E, Jahromi M, Chikkamenahalli L, Park J, Hill J. Insulin sensing by astrocytes is critical for normal thermogenesis and body temperature regulation. J Endocrinol 2020; 247:39-52. [PMID: 32698146 PMCID: PMC7456332 DOI: 10.1530/joe-20-0052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 07/13/2020] [Indexed: 12/23/2022]
Abstract
The important role of astrocytes in the central control of energy balance and glucose homeostasis has recently been recognized. Changes in thermoregulation can lead to metabolic dysregulation, but the role of astrocytes in this process is not yet clear. Therefore, we generated mice congenitally lacking insulin receptors (Ir) in astrocytes (IrKOGFAP mice) to investigate the involvement of astrocyte insulin signaling. IrKOGFAP mice displayed significantly lower energy expenditure and a strikingly lower basal and fasting body temperature. When exposed to cold, however, they were able to mount a thermogenic response. IrKOGFAP mice displayed sex differences in metabolic function and thermogenesis that may contribute to the development of obesity and type II diabetes as early as 2 months of age. While brown adipose tissue exhibited higher adipocyte size in both sexes, more apoptosis was seen in IrKOGFAP males. Less innervation and lower BAR3 expression levels were also observed in IrKOGFAP brown adipose tissue. These effects have not been reported in models of astrocyte Ir deletion in adulthood. In contrast, body weight and glucose regulatory defects phenocopied such models. These findings identify a novel role for astrocyte insulin signaling in the development of normal body temperature control and sympathetic activation of BAT. Targeting insulin signaling in astrocytes has the potential to serve as a novel target for increasing energy expenditure.
Collapse
Affiliation(s)
- Iyad H Manaserh
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Emily Maly
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Marziyeh Jahromi
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Lakshmikanth Chikkamenahalli
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Joshua Park
- Department of Neuroscience, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Jennifer Hill
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| |
Collapse
|
17
|
Lee H, Kim E. Repositioning medication for cardiovascular and cerebrovascular disease to delay the onset and prevent progression of Alzheimer's disease. Arch Pharm Res 2020; 43:932-960. [PMID: 32909178 DOI: 10.1007/s12272-020-01268-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 08/31/2020] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a complex, progressive, neurodegenerative disorder. As with other common chronic diseases, multiple risk factors contribute to the onset and progression of AD. Many researchers have evaluated the epidemiologic and pathophysiological association between AD, cardiovascular diseases (CVDs), and cerebrovascular diseases (CBVDs), including commonly reported risk factors such as diabetes, hypertension, and dyslipidemia. Relevant therapies of CVDs/CBVDs for the attenuation of AD have also been empirically investigated. Considering the challenges of new drug development, in terms of cost and time, multifactorial approaches such as therapeutic repositioning of CVD/CBVD medication should be explored to delay the onset and progression of AD. Thus, in this review, we discuss our current understanding of the association between cardiovascular risk factors and AD, as revealed by clinical and non-clinical studies, as well as the therapeutic implications of CVD/CBVD medication that may attenuate AD. Furthermore, we discuss future directions by evaluating ongoing trials in the field.
Collapse
Affiliation(s)
- Heeyoung Lee
- Department of Clinical Medicinal Sciences, Konyang University, 121 Daehakro, Nonsan, 32992, Republic of Korea
| | - EunYoung Kim
- Evidence-Based Research Laboratory, Division of Clinical Pharmacotherapy, College of Pharmacy, Chung-Ang University, Seoul, 156-756, Republic of Korea.
| |
Collapse
|
18
|
Pitchaimani V, Arumugam S, Thandavarayan RA, Karuppagounder V, Afrin MR, Sreedhar R, Harima M, Nakamura M, Watanabe K, Kodama S, Fujihara K, Sone H. Brain adaptations of insulin signaling kinases, GLUT 3, p-BADser155 and nitrotyrosine expression in various hypoglycemic models of mice. Neurochem Int 2020; 137:104745. [DOI: 10.1016/j.neuint.2020.104745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 12/29/2022]
|
19
|
Poupon-Bejuit L, Rocha-Ferreira E, Thornton C, Hagberg H, Rahim AA. Neuroprotective Effects of Diabetes Drugs for the Treatment of Neonatal Hypoxia-Ischemia Encephalopathy. Front Cell Neurosci 2020; 14:112. [PMID: 32435185 PMCID: PMC7218053 DOI: 10.3389/fncel.2020.00112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/08/2020] [Indexed: 12/15/2022] Open
Abstract
The perinatal period represents a time of great vulnerability for the developing brain. A variety of injuries can result in death or devastating injury causing profound neurocognitive deficits. Hypoxic-ischemic neonatal encephalopathy (HIE) remains the leading cause of brain injury in term infants during the perinatal period with limited options available to aid in recovery. It can result in long-term devastating consequences with neurologic complications varying from mild behavioral deficits to severe seizure, intellectual disability, and/or cerebral palsy in the newborn. Despite medical advances, the only viable option is therapeutic hypothermia which is classified as the gold standard but is not used, or may not be as effective in preterm cases, infection-associated cases or low resource settings. Therefore, alternatives or adjunct therapies are urgently needed. Ongoing research continues to advance our understanding of the mechanisms contributing to perinatal brain injury and identify new targets and treatments. Drugs used for the treatment of patients with type 2 diabetes mellitus (T2DM) have demonstrated neuroprotective properties and therapeutic efficacy from neurological sequelae following HIE insults in preclinical models, both alone, or in combination with induced hypothermia. In this short review, we have focused on recent findings on the use of diabetes drugs that provide a neuroprotective effect using in vitro and in vivo models of HIE that could be considered for clinical translation as a promising treatment.
Collapse
Affiliation(s)
| | - Eridan Rocha-Ferreira
- Centre for Perinatal Medicine and Health, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Claire Thornton
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Henrik Hagberg
- Centre for Perinatal Medicine and Health, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ahad A. Rahim
- UCL School of Pharmacy, University College London, London, United Kingdom
| |
Collapse
|
20
|
Tang BL. Could metformin be therapeutically useful in Huntington's disease? Rev Neurosci 2020; 31:297-317. [PMID: 31751298 DOI: 10.1515/revneuro-2019-0072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022]
Abstract
Emerging evidence suggest that dimethylbiguanide (metformin), a first-line drug for type 2 diabetes mellitus, could be neuroprotective in a range of brain pathologies, which include neurodegenerative diseases and brain injury. However, there are also contraindications that associate metformin treatment with cognitive impairment as well as adverse outcomes in Alzheimer's disease and Parkinson's disease animal models. Recently, a beneficial effect of metformin in animal models of Huntington's disease (HD) has been strengthened by multiple reports. In this brief review, the findings associated with the effects of metformin in attenuating neurodegenerative diseases are discussed, focusing on HD-associated pathology and the potential underlying mechanisms highlighted by these studies. The mechanism of action of metformin is complex, and its therapeutic efficacy is therefore expected to be dependent on the disease context. The key metabolic pathways that are effectively affected by metformin, such as AMP-activated protein kinase activation, may be altered in the later decades of the human lifespan. In this regard, metformin may nonetheless be therapeutically useful for neurological diseases with early pathological onsets, such as HD.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, Singapore 117596, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Medical Drive, Singapore 119077, Singapore
| |
Collapse
|
21
|
Youssef MM, Abd El-Latif HA, El-Yamany MF, Georgy GS. Aliskiren and captopril improve cognitive deficits in poorly controlled STZ-induced diabetic rats via amelioration of the hippocampal P-ERK, GSK3β, P-GSK3β pathway. Toxicol Appl Pharmacol 2020; 394:114954. [PMID: 32171570 DOI: 10.1016/j.taap.2020.114954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/08/2020] [Accepted: 03/10/2020] [Indexed: 12/22/2022]
Abstract
Learning and memory deficits are obvious symptoms that develop over time in patients with poorly controlled diabetes. Hyperactivity of the renin-angiotensin system (RAS) is directly associated with β-cell dysfunction and diabetic complications, including cognitive impairment. Here, we investigated the protective and molecular effects of two RAS modifiers, aliskiren; renin inhibitor and captopril; angiotensin converting enzyme inhibitor, on cognitive deficits in the rat hippocampus. Injection of low dose streptozotocin for 4 days resulted in type 1 diabetes. Then, poorly controlled diabetes was mimicked with ineffective daily doses of insulin for 4 weeks. The hyperglycaemia and pancreatic atrophy caused memory disturbance that were identifiable in behavioural tests, hippocampal neurodegeneration, and the following significant changes in the hippocampus, increases in the inflammatory marker interleukin 1β, cholinesterase, the oxidative stress marker malondialdehyde and protein expression of phosphorylated extracellular-signal-regulated kinase and glycogen synthase kinase-3 beta versus decrease in the antioxidant reduced glutathione and protein expression of phosphorylated glycogen synthase kinase-3 beta. Blocking RAS with either drugs along with insulin amended all previously mentioned parameters. Aliskiren stabilized the blood glucose level and restored normal pancreatic integrity and hippocampal malondialdehyde level. Aliskiren showed superior protection against the hippocampal degeneration displayed in the earlier behavioural modification in the passive avoidance test, and the aliskiren group outperformed the control group in the novel object recognition test. We therefore conclude that aliskiren and captopril reversed the diabetic state and cognitive deficits in rats with poorly controlled STZ-induced diabetes through reducing oxidative stress and inflammation and modulating protein expression.
Collapse
Affiliation(s)
- Madonna M Youssef
- Department of Pharmacology, National organization for drug control and research (NODCAR), Giza, Egypt.
| | - H A Abd El-Latif
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St, Cairo 11562, Egypt
| | - M F El-Yamany
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St, Cairo 11562, Egypt
| | - Gehan S Georgy
- Department of Pharmacology, National organization for drug control and research (NODCAR), Giza, Egypt
| |
Collapse
|
22
|
Zhang QQ, Li WS, Liu Z, Zhang HL, Ba YG, Zhang RX. Metformin therapy and cognitive dysfunction in patients with type 2 diabetes: A meta-analysis and systematic review. Medicine (Baltimore) 2020; 99:e19378. [PMID: 32150083 PMCID: PMC7478804 DOI: 10.1097/md.0000000000019378] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is a risk factor for cognitive dysfunction. The relationship between metformin therapy and cognitive function in patients with T2D is unknown. Therefore, we determined the relationship between metformin therapy and cognitive function in patients with T2D using a meta-analysis. METHODS We systematically searched the Cochrane library, PubMed, and Embase to identify studies showing correlations, and we calculated hazard ratios (HRs). RESULTS We identified 10 studies including 254,679 participants. Metformin significantly reduced the occurrence of cognitive dysfunction in patients with T2D (HR 0.90; 95% CI [0.88, 0.92]). Compared with other hypoglycemic drugs, sulfonylureas also improved cognitive dysfunction (HR 0.92; 95% CI [0.88, 0.95]). Thiazolidinediones gave no statistically significant improvement in cognitive dysfunction (HR 0.97; 95% CI [0.87, 1.07]). The use of insulin aggravated cognitive dysfunction (HR 1.34; 95% CI [1.24, 1.43]). In the subgroup analysis of various regions controlling for age, gender, education, diabetes course, complications, metformin administration and dosage, and follow-up time, metformin significantly improved cognitive dysfunction in patients in the Americas and Europe (HR 0.69; 95% CI [0.63, 0.74]), (HR 0.71; 95% CI [0.66, 0.76], respectively), while metformin did not significantly improve cognitive dysfunction in Asian patients (HR 0.99; 95% CI [0.96, 1.01]). CONCLUSIONS Metformin significantly improved cognitive dysfunction in patients with T2D. Sulfonylureas also improved cognitive dysfunction. Thiazolidinediones had no significant effect on cognitive dysfunction. The use of insulin aggravated cognitive dysfunction. Metformin improved cognitive dysfunction more significantly in patients in the Americas and Europe than in Asia.
Collapse
Affiliation(s)
- Qing-Qing Zhang
- Department of Endocrinology, Qinghai University Affiliated Hospital
| | - Wen-Shan Li
- Department of Neurosurgery, Qinghai Provincial People's Hospital
| | - Zhou Liu
- Graduate School of Qinghai University
| | - Hui-Li Zhang
- Department of Endocrinology, Qinghai University Affiliated Hospital
| | - Ying-Gui Ba
- Department of Nephrology, Qinghai University Affiliated Hospital Xining, Qinghai, China
| | - Rui-Xia Zhang
- Department of Endocrinology, Qinghai University Affiliated Hospital
| |
Collapse
|
23
|
Nasoohi S, Parveen K, Ishrat T. Metabolic Syndrome, Brain Insulin Resistance, and Alzheimer's Disease: Thioredoxin Interacting Protein (TXNIP) and Inflammasome as Core Amplifiers. J Alzheimers Dis 2019; 66:857-885. [PMID: 30372683 DOI: 10.3233/jad-180735] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Empirical evidence indicates a strong association between insulin resistance and pathological alterations related to Alzheimer's disease (AD) in different cerebral regions. While cerebral insulin resistance is not essentially parallel with systemic metabolic derangements, type 2 diabetes mellitus (T2DM) has been established as a risk factor for AD. The circulating "toxic metabolites" emerging in metabolic syndrome may engage several biochemical pathways to promote oxidative stress and neuroinflammation leading to impair insulin function in the brain or "type 3 diabetes". Thioredoxin-interacting protein (TXNIP) as an intracellular amplifier of oxidative stress and inflammasome activation may presumably mediate central insulin resistance. Emerging data including those from our recent studies has demonstrated a sharp TXNIP upregulation in stroke, aging and AD and well underlining the significance of this hypothesis. With the main interest to illustrate TXNIP place in type 3 diabetes, the present review primarily briefs the potential mechanisms contributing to cerebral insulin resistance in a metabolically deranged environment. Then with a particular focus on plausible TXNIP functions to drive and associate with AD pathology, we present the most recent evidence supporting TXNIP as a promising therapeutic target in AD as an age-associated dementia.
Collapse
|
24
|
Shi Q, Liu S, Fonseca VA, Thethi TK, Shi L. Effect of metformin on neurodegenerative disease among elderly adult US veterans with type 2 diabetes mellitus. BMJ Open 2019; 9:e024954. [PMID: 31366635 PMCID: PMC6677947 DOI: 10.1136/bmjopen-2018-024954] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE This study aimed to evaluate the association between metformin treatment and the risk of neurodegenerative disease (ND) among elderly adults with type 2 diabetes mellitus (T2DM). DESIGN/SETTING/PARTICIPANTS This retrospective longitudinal cohort study examined the effects of the length of metformin exposure on ND among elderly US veterans with T2DM and insulin treatment using the Veterans Affairs electronic medical record database. PRIMARY AND SECONDARY OUTCOME MEASURES The primary clinical outcome was defined as diagnosis of ND including dementia, Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and mild cognitive impairment during the follow-up period. The secondary clinical outcomes were separately measured by AD, PD, HD, dementia and mild cognitive impairment. RESULT Adjusted by propensity score weight, a total of 5528 patients (mean age, 63.2±10.9 years; male, 98%; white, 60%) with a median follow-up of 5.2 years were selected. Those with ND or other mental disorders at baseline or who were on insulin for less than two-thirds of the study period were excluded. The incidence rate of ND was 11.48 per 1000 person-years among patients with metformin treatment, compared with 25.45 per 1000 person-years for those without metformin. Compared with no metformin use, 2-4 years and >4 years of metformin exposure were significantly associated with lower risk of ND (adjusted HR (aHR)=0.62, 95% CI 0.45 to 0.85; aHR=0.19, 95% CI 0.12 to 0.31, respectively), while metformin exposure in the first 2 years showed no significant influence. CONCLUSION We conclude that long-term metformin therapy (>2 years) was associated with lower incidence of ND among elderly veterans with T2DM. We need to conduct a study with more representative population and more robust method for causal inferences. Further investigation into the mechanism involved is needed along with randomised trials to confirm a potential neuroprotective effect of metformin.
Collapse
Affiliation(s)
- Qian Shi
- Global Health Management and Policy, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Shuqian Liu
- Global Health Management and Policy, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Vivian A Fonseca
- Section of Endocrinology, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Endocrinology, Southeast Louisiana Veterans Health Care System, New Orleans, Louisiana, USA
| | - Tina K Thethi
- Section of Endocrinology, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Endocrinology, Southeast Louisiana Veterans Health Care System, New Orleans, Louisiana, USA
| | - Lizheng Shi
- Global Health Management and Policy, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
25
|
Leech T, Chattipakorn N, Chattipakorn SC. The beneficial roles of metformin on the brain with cerebral ischaemia/reperfusion injury. Pharmacol Res 2019; 146:104261. [PMID: 31170502 DOI: 10.1016/j.phrs.2019.104261] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
Cerebral ischaemia/reperfusion (I/R) injury is the transient loss, followed by rapid return, of blood flow to the brain. This condition is often caused by strokes and heart attacks. The underlying mechanisms resulting in brain damage during cerebral I/R injury include mitochondrial dysregulation, increased oxidative stress/reactive oxygen species, blood-brain-barrier breakdown, inflammation of the brain, and increased neuronal apoptosis. Metformin is the first-line antidiabetic drug which has recently been shown to be capable of acting through the aforementioned pathways to improve recovery following cerebral I/R injury. However, some studies have suggested that metformin therapy may have no effect or even worsen recovery following cerebral I/R injury. The present review will compile and examine the available in vivo, in vitro, and clinical data concerning the neuroprotective effects of metformin following cerebral I/R injury. Any contradictory evidence will also be assessed and presented to determine the actual effectiveness of metformin treatment in stroke recovery.
Collapse
Affiliation(s)
- Tom Leech
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, United Kingdom; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
26
|
Frangou S, Shirali M, Adams MJ, Howard DM, Gibson J, Hall LS, Smith BH, Padmanabhan S, Murray AD, Porteous DJ, Haley CS, Deary IJ, Clarke TK, McIntosh AM. Insulin resistance: Genetic associations with depression and cognition in population based cohorts. Exp Neurol 2019; 316:20-26. [PMID: 30965038 PMCID: PMC6503941 DOI: 10.1016/j.expneurol.2019.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/26/2019] [Accepted: 04/03/2019] [Indexed: 01/07/2023]
Abstract
Insulin resistance, broadly defined as the reduced ability of insulin to exert its biological action, has been associated with depression and cognitive dysfunction in observational studies. However, it is unclear whether these associations are causal and whether they might be underpinned by other shared factors. To address this knowledge gap, we capitalized on the stability of genetic biomarkers through the lifetime, and on their unidirectional relationship with depression and cognition. Specifically, we determined the association between quantitative measures of cognitive function and depression and genetic instruments of insulin resistance traits in two large-scale population samples, the Generation Scotland: Scottish Family Health Study (GS: SFHS; N = 19,994) and in the UK Biobank (N = 331,374). In the GS:SFHS, the polygenic risk score (PRS) for fasting insulin was associated with verbal intelligence and depression while the PRS for the homeostasis model assessment of insulin resistance was associated with verbal intelligence. Despite this overlap in genetic architecture, Mendelian randomization analyses in the GS:SFHS and in the UK Biobank samples did not yield evidence for causal associations from insulin resistance traits to either depression or cognition. These findings may be due to weak genetic instruments, limited cognitive measures and insufficient power but they may also indicate the need to identify other biological mechanisms that may mediate the relationship from insulin resistance to depression and cognition.
Collapse
Affiliation(s)
- Sophia Frangou
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Masoud Shirali
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Mark J Adams
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - David M Howard
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Jude Gibson
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Lynsey S Hall
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Blair H Smith
- Division of Population Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Alison D Murray
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - David J Porteous
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK; Generation Scotland, Medical Genetics Section, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Chris S Haley
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK; Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Toni-Kim Clarke
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Andrew M McIntosh
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK; Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK; Department of Psychology, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
27
|
El Idrissi A. Taurine Regulation of Neuroendocrine Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1155:977-985. [PMID: 31468461 DOI: 10.1007/978-981-13-8023-5_81] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Taurine (2-aminoethanesulfonic acid) is a sulfur-containing amino acid. It is one of the most abundant free amino acids in many excitable tissues, including the brain, skeletal and cardiac muscles. Physiological actions of taurine are widespread and include regulation of plasma glucose levels, bile acid conjugation, detoxification, membrane stabilization, blood pressure regulation, osmoregulation, neurotransmission, and modulation of mitochondria function and cellular calcium levels. Taurine plays an important role in modulating glutamate and GABA neurotransmission and prevents excitotoxicity in vitro primarily through modulation of intracellular calcium homeostasis. Taurine supplementation prevents age-dependent decline of cognitive functions. Because of the wide spread actions of taurine, its levels are highly regulated through enzymatic biosynthesis or dietary intake. Furthermore, depletion of endogenous or dietary supplementation of exogenous taurine have been shown to induce wide spread actions on multiple organs. Cysteine sulfonic acid decarboxylase (CSAD) was first identified in the liver and is thought to be the rate-limiting enzyme in taurine biosynthesis. CSAD mRNA is expressed in the brain in astrocytes. Homozygous knockout mice lacking CSAD (CSAD-KO) have very reduced taurine content and show severe functional histopathology in the visual system, skeletal system, heart, pancreas and brain. Conversely, dietary supplementation of taurine results in significant health benefits acting through the same organ systems. Fluctuation of taurine bioavailability lead to changes in the expression levels of taurine transporters in neuronal plasma membranes, endothelial cells forming the blood-brain barrier and proximal cells of the kidneys. Suggesting a highly regulated mechanism for maintaining taurine homeostasis and organ systems function. Here we show how alterations in taurine levels directly affect the function of one organ system and through functional interaction and compensatory adaptation; these effects extend to another organ systems with focus on the nervous system.
Collapse
Affiliation(s)
- Abdeslem El Idrissi
- Department of Biology, Center for Developmental Neuroscience, College of Staten Island, Staten Island, NY, USA.
| |
Collapse
|
28
|
Mamo JC, Lam V, Brook E, Mooranian A, Al-Salami H, Fimognari N, Nesbit M, Takechi R. Probucol prevents blood-brain barrier dysfunction and cognitive decline in mice maintained on pro-diabetic diet. Diab Vasc Dis Res 2019; 16:87-97. [PMID: 30156119 DOI: 10.1177/1479164118795274] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
An emerging body of evidence consistently suggests that compromised blood-brain barrier integrity may be causally associated with cognitive decline induced by type-2 diabetes. Our previous studies demonstrated that selected anti-inflammatory/anti-oxidative agents can preserve the integrity of blood-brain barrier and prevent neuroinflammation in mouse models of dysfunctional blood-brain barrier. Therefore, we have tested whether the previously proven blood-brain barrier protective agent, probucol, can prevent blood-brain barrier breakdown and cognitive decline in a dietary-induced murine model of diabetic insulin resistance. After 6-month chronic ingestion of a diet high in fat and fructose, the mice became insulin resistant. The high-fat and high-fructose-fed mice showed significant cognitive decline assessed by Morris water maze, concomitant with significant elevations in cortical and hippocampal glial acidic fibrillary protein and Fluoro Jade-C staining, indicating heightened neuroinflammation and neurodegeneration, respectively. The integrity of blood-brain barrier in high-fat and high-fructose-fed mice was substantially compromised, and this showed a significant association with heightened neurodegeneration. Co-provision of probucol with high-fat and high-fructose diet completely prevented the cognitive decline and blood-brain barrier dysfunction. Similarly, metformin was able to restore the cognitive function in high-fat and high-fructose-fed mice, while its blood-brain barrier protective effects were modest. These data suggest that probucol may prevent cognitive decline induced by insulin resistance by preserving the integrity of blood-brain barrier, whereas metformin's neuroprotective effects may be mediated through a separate pathway.
Collapse
Affiliation(s)
- John Cl Mamo
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 2 School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Virginie Lam
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 2 School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Emily Brook
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 3 School of Biomedical Sciences, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Armin Mooranian
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 4 School of Pharmacy, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Hani Al-Salami
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 4 School of Pharmacy, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Nicholas Fimognari
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 2 School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Michael Nesbit
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 2 School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Ryusuke Takechi
- 1 Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
- 2 School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| |
Collapse
|
29
|
Rad SK, Arya A, Karimian H, Madhavan P, Rizwan F, Koshy S, Prabhu G. Mechanism involved in insulin resistance via accumulation of β-amyloid and neurofibrillary tangles: link between type 2 diabetes and Alzheimer's disease. Drug Des Devel Ther 2018; 12:3999-4021. [PMID: 30538427 PMCID: PMC6255119 DOI: 10.2147/dddt.s173970] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The pathophysiological link between type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) has been suggested in several reports. Few findings suggest that T2DM has strong link in the development process of AD, and the complete mechanism is yet to be revealed. Formation of amyloid plaques (APs) and neurofibrillary tangles (NFTs) are two central hallmarks in the AD. APs are the dense composites of β-amyloid protein (Aβ) which accumulates around the nerve cells. Moreover, NFTs are the twisted fibers containing hyperphosphorylated tau proteins present in certain residues of Aβ that build up inside the brain cells. Certain factors contribute to the aetiogenesis of AD by regulating insulin signaling pathway in the brain and accelerating the formation of neurotoxic Aβ and NFTs via various mechanisms, including GSK3β, JNK, CamKII, CDK5, CK1, MARK4, PLK2, Syk, DYRK1A, PPP, and P70S6K. Progression to AD could be influenced by insulin signaling pathway that is affected due to T2DM. Interestingly, NFTs and APs lead to the impairment of several crucial cascades, such as synaptogenesis, neurotrophy, and apoptosis, which are regulated by insulin, cholesterol, and glucose metabolism. The investigation of the molecular cascades through insulin functions in brain contributes to probe and perceive progressions of diabetes to AD. This review elaborates the molecular insights that would help to further understand the potential mechanisms linking T2DM and AD.
Collapse
Affiliation(s)
- Sima Kianpour Rad
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Aditya Arya
- Department of Pharmacology and Therapeutics, School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia,
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia,
- Malaysian Institute of Pharmaceuticals and Nutraceuticals (IPharm), Bukit Gambir, Gelugor, Pulau Pinang, Malaysia,
| | - Hamed Karimian
- Department of Pharmacology and Therapeutics, School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia,
| | - Priya Madhavan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Farzana Rizwan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Shajan Koshy
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Girish Prabhu
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| |
Collapse
|
30
|
El Idrissi A, El Hilali F, Rotondo S, Sidime F. Effects of Taurine Supplementation on Neuronal Excitability and Glucose Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 1:271-279. [PMID: 28849462 DOI: 10.1007/978-94-024-1079-2_24] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In this study we examined the role of chronic taurine supplementation on plasma glucose homeostasis and brain excitability through activation of the insulin receptor. FVB/NJ male mice were supplemented with taurine in drinking water (0.05% w/v) for 4 weeks and subjected to a glucose tolerance test (7.5 mg/kg BW) after 12 h fasting. We found that taurine-fed mice were slightly hypoglycemic prior to glucose injection and showed significantly reduced plasma glucose at 30 and 60 min post-glucose injection when compared to control mice. Previously, we reported that taurine supplementation induces biochemical changes that target the GABAergic system. Those studies show that taurine-fed mice are hyperexcitable, have reduced GABAA receptors expression and increased GAD and somatostatin expression in the brain. In this study, we found that taurine-fed mice had a significant increase in insulin receptor (IR) immuno-reactivity in the pancreas and all brain regions examined. At the mRNA level, we found that the IR showed differential regional expression. Surprisingly, we found that neurons express the gene for insulin and that taurine had a significant role in regulating insulin gene expression. We propose that increased insulin production and secretion in taurine-fed mice cause an increase activation of the central IR and may be partially responsible for the increased neuronal excitability observed in taurine supplemented mice. Furthermore, the high levels of neuronal insulin expression and its regulation by taurine implicates taurine in the regulation of metabolic homeostasis.
Collapse
Affiliation(s)
- Abdeslem El Idrissi
- Department of Biology, College of Staten Island, Staten Island, NY, USA.
- Graduate School, City University of New York, New York, NY, USA.
- Center for Developmental Neuroscience, Staten Island, NY, USA.
| | | | - Salvatore Rotondo
- Department of Biology, College of Staten Island, Staten Island, NY, USA
| | - Francoise Sidime
- Department of Biology, College of Staten Island, Staten Island, NY, USA
- Graduate School, City University of New York, New York, NY, USA
- Center for Developmental Neuroscience, Staten Island, NY, USA
| |
Collapse
|
31
|
Pomytkin I, Costa‐Nunes JP, Kasatkin V, Veniaminova E, Demchenko A, Lyundup A, Lesch K, Ponomarev ED, Strekalova T. Insulin receptor in the brain: Mechanisms of activation and the role in the CNS pathology and treatment. CNS Neurosci Ther 2018; 24:763-774. [PMID: 29691988 PMCID: PMC6489906 DOI: 10.1111/cns.12866] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/28/2018] [Accepted: 03/30/2018] [Indexed: 12/16/2022] Open
Abstract
While the insulin receptor (IR) was found in the CNS decades ago, the brain was long considered to be an insulin-insensitive organ. This view is currently revisited, given emerging evidence of critical roles of IR-mediated signaling in development, neuroprotection, metabolism, and plasticity in the brain. These diverse cellular and physiological IR activities are distinct from metabolic IR functions in peripheral tissues, thus highlighting region specificity of IR properties. This particularly concerns the fact that two IR isoforms, A and B, are predominantly expressed in either the brain or peripheral tissues, respectively, and neurons express exclusively IR-A. Intriguingly, in comparison with IR-B, IR-A displays high binding affinity and is also activated by low concentrations of insulin-like growth factor-2 (IGF-2), a regulator of neuronal plasticity, whose dysregulation is associated with neuropathologic processes. Deficiencies in IR activation, insulin availability, and downstream IR-related mechanisms may result in aberrant IR-mediated functions and, subsequently, a broad range of brain disorders, including neurodevelopmental syndromes, neoplasms, neurodegenerative conditions, and depression. Here, we discuss findings on the brain-specific features of IR-mediated signaling with focus on mechanisms of primary receptor activation and their roles in the neuropathology. We aimed to uncover the remaining gaps in current knowledge on IR physiology and highlight new therapies targeting IR, such as IR sensitizers.
Collapse
Affiliation(s)
- Igor Pomytkin
- Department of Advanced Cell TechnologiesInstitute of Regenerative MedicineSechenov First Moscow State Medical UniversityMoscowRussia
| | - João P. Costa‐Nunes
- Department of Normal PhysiologyLaboratory of Psychiatric NeurobiologyInstitute of Molecular MedicineSechenov First Moscow State Medical UniversityMoscowRussia
- Faculdade de Medicina de LisboaInstituto de Medicina MolecularUniversidade de LisboaLisboaPortugal
| | - Vladimir Kasatkin
- Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and ImmunologyMoscowRussia
| | - Ekaterina Veniaminova
- Department of Normal PhysiologyLaboratory of Psychiatric NeurobiologyInstitute of Molecular MedicineSechenov First Moscow State Medical UniversityMoscowRussia
- Laboratory of Cognitive DysfunctionsInstitute of General Pathology and PathophysiologyMoscowRussia
- Department of NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - Anna Demchenko
- Department of Advanced Cell TechnologiesInstitute of Regenerative MedicineSechenov First Moscow State Medical UniversityMoscowRussia
| | - Alexey Lyundup
- Department of Advanced Cell TechnologiesInstitute of Regenerative MedicineSechenov First Moscow State Medical UniversityMoscowRussia
| | - Klaus‐Peter Lesch
- Department of Normal PhysiologyLaboratory of Psychiatric NeurobiologyInstitute of Molecular MedicineSechenov First Moscow State Medical UniversityMoscowRussia
- Department of NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
- Division of Molecular PsychiatryCenter of Mental HealthClinical Research Unit on Disorders of Neurodevelopment and CognitionUniversity of WürzburgWürzburgGermany
| | - Eugene D. Ponomarev
- Faculty of MedicineSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongHong Kong
| | - Tatyana Strekalova
- Department of Normal PhysiologyLaboratory of Psychiatric NeurobiologyInstitute of Molecular MedicineSechenov First Moscow State Medical UniversityMoscowRussia
- Laboratory of Cognitive DysfunctionsInstitute of General Pathology and PathophysiologyMoscowRussia
- Department of NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
32
|
Taibi-Djennah Z, Martin-Eauclaire MF, Laraba-Djebari F. Evaluation of neuroprotective effects of insulin on immuno-inflammatory and systemic disorders induced by kaliotoxin, a Kv1.3 channel blocker. Inflamm Res 2018; 67:863-877. [DOI: 10.1007/s00011-018-1177-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 06/12/2018] [Accepted: 08/01/2018] [Indexed: 12/11/2022] Open
|
33
|
Zhong KL, Chen F, Hong H, Ke X, Lv YG, Tang SS, Zhu YB. New views and possibilities of antidiabetic drugs in treating and/or preventing mild cognitive impairment and Alzheimer's Disease. Metab Brain Dis 2018; 33:1009-1018. [PMID: 29626315 DOI: 10.1007/s11011-018-0227-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 03/28/2018] [Indexed: 12/13/2022]
Abstract
Mounting evidence suggests that diabetes mellitus (DM) is associated with mild cognitive impairment (MCI), vascular dementia and Alzheimer's disease (AD). Biological, clinical and epidemiological data support a close link between DM and AD. Increasingly, studies have found that several antidiabetic agents can promote neurogenesis, and clinically ameliorate cognitive and memory impairments in different clinical settings. Data has shown that these antidiabetic drugs positively affect mitochondrial and synaptic function, neuroinflammation, and brain metabolism. Evidence to date strongly suggests that these antidiabetic drugs could be developed as disease-modifying therapies for MCI and AD in patients with and without diabetes.
Collapse
Affiliation(s)
- Kai Long Zhong
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Fang Chen
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Hao Hong
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Xuan Ke
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Yang Ge Lv
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Su Su Tang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Yu Bing Zhu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
34
|
Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nat Rev Neurol 2018; 14:168-181. [PMID: 29377010 DOI: 10.1038/nrneurol.2017.185] [Citation(s) in RCA: 885] [Impact Index Per Article: 147.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Considerable overlap has been identified in the risk factors, comorbidities and putative pathophysiological mechanisms of Alzheimer disease and related dementias (ADRDs) and type 2 diabetes mellitus (T2DM), two of the most pressing epidemics of our time. Much is known about the biology of each condition, but whether T2DM and ADRDs are parallel phenomena arising from coincidental roots in ageing or synergistic diseases linked by vicious pathophysiological cycles remains unclear. Insulin resistance is a core feature of T2DM and is emerging as a potentially important feature of ADRDs. Here, we review key observations and experimental data on insulin signalling in the brain, highlighting its actions in neurons and glia. In addition, we define the concept of 'brain insulin resistance' and review the growing, although still inconsistent, literature concerning cognitive impairment and neuropathological abnormalities in T2DM, obesity and insulin resistance. Lastly, we review evidence of intrinsic brain insulin resistance in ADRDs. By expanding our understanding of the overlapping mechanisms of these conditions, we hope to accelerate the rational development of preventive, disease-modifying and symptomatic treatments for cognitive dysfunction in T2DM and ADRDs alike.
Collapse
|
35
|
Nday CM, Eleftheriadou D, Jackson G. Shared pathological pathways of Alzheimer's disease with specific comorbidities: current perspectives and interventions. J Neurochem 2018; 144:360-389. [PMID: 29164610 DOI: 10.1111/jnc.14256] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) belongs to one of the most multifactorial, complex and heterogeneous morbidity-leading disorders. Despite the extensive research in the field, AD pathogenesis is still at some extend obscure. Mechanisms linking AD with certain comorbidities, namely diabetes mellitus, obesity and dyslipidemia, are increasingly gaining importance, mainly because of their potential role in promoting AD development and exacerbation. Their exact cognitive impairment trajectories, however, remain to be fully elucidated. The current review aims to offer a clear and comprehensive description of the state-of-the-art approaches focused on generating in-depth knowledge regarding the overlapping pathology of AD and its concomitant ailments. Thorough understanding of associated alterations on a number of molecular, metabolic and hormonal pathways, will contribute to the further development of novel and integrated theranostics, as well as targeted interventions that may be beneficial for individuals with age-related cognitive decline.
Collapse
Affiliation(s)
- Christiane M Nday
- Department of Chemical Engineering, Laboratory of Inorganic Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Despoina Eleftheriadou
- Department of Chemical Engineering, Laboratory of Inorganic Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Graham Jackson
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, South Africa
| |
Collapse
|
36
|
Houck AL, Seddighi S, Driver JA. At the Crossroads Between Neurodegeneration and Cancer: A Review of Overlapping Biology and Its Implications. Curr Aging Sci 2018; 11:77-89. [PMID: 29552989 PMCID: PMC6519136 DOI: 10.2174/1874609811666180223154436] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 02/08/2018] [Accepted: 02/13/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND A growing body of epidemiologic evidence suggests that neurodegenerative diseases occur less frequently in cancer survivors, and vice versa. While unusual, this inverse comorbidity is biologically plausible and could be explained, in part, by the evolutionary tradeoffs made by neurons and cycling cells to optimize the performance of their very different functions. The two cell types utilize the same proteins and pathways in different, and sometimes opposite, ways. However, cancer and neurodegeneration also share many pathophysiological features. OBJECTIVE In this review, we compare three overlapping aspects of neurodegeneration and cancer. METHOD First, we contrast the priorities and tradeoffs of dividing cells and neurons and how these manifest in disease. Second, we consider the hallmarks of biological aging that underlie both neurodegeneration and cancer. Finally, we utilize information from genetic databases to outline specific genes and pathways common to both diseases. CONCLUSION We argue that a detailed understanding of the biologic and genetic relationships between cancer and neurodegeneration can guide future efforts in designing disease-modifying therapeutic interventions. Lastly, strategies that target aging may prevent or delay both conditions.
Collapse
Affiliation(s)
- Alexander L. Houck
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sahba Seddighi
- Clinical and Translational Neuroscience Unit, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Jane A. Driver
- Geriatric Research Education and Clinical Center, VA Boston Healthcare System and the Division of Aging, Department of Medicine, Brigham and Women ‘s Hospital, Harvard Medical School (J.A.D.), Boston, MA, USA
| |
Collapse
|
37
|
Sánchez-Gracia A, Guirao-Rico S, Hinojosa-Alvarez S, Rozas J. Computational prediction of the phenotypic effects of genetic variants: basic concepts and some application examples in Drosophila nervous system genes. J Neurogenet 2017; 31:307-319. [DOI: 10.1080/01677063.2017.1398241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Alejandro Sánchez-Gracia
- Departament de Genètica, Microbiologia i Estadística and Institut de Recerca de la Biodiversitat (IRBio), Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Sara Guirao-Rico
- Center for Research in Agricultural Genomics (CRAG) CSIC-IRTA-UAB-UB, Bellaterra, Spain
| | - Silvia Hinojosa-Alvarez
- Departament de Genètica, Microbiologia i Estadística and Institut de Recerca de la Biodiversitat (IRBio), Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Julio Rozas
- Departament de Genètica, Microbiologia i Estadística and Institut de Recerca de la Biodiversitat (IRBio), Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
38
|
Akın O, Eker İ, Arslan M, Yavuz ST, Akman S, Taşçılar ME, Ünay B. Relation of insulin resistance to neurocognitive function and electroencephalography in obese children. J Pediatr Endocrinol Metab 2017; 30:1027-1032. [PMID: 28841575 DOI: 10.1515/jpem-2017-0186] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/24/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND Childhood obesity may lead to neuronal impairment in both the peripheral and the central nervous system. This study aimed to investigate the impact of obesity and insulin resistance (IR) on the central nervous system and neurocognitive functions in children. METHODS Seventy-three obese children (38 male and 35 female) and 42 healthy children (21 male and 21 female) were recruited. Standard biochemical indices and IR were evaluated. The Wechsler Intelligence Scale for Children-Revised (WISC-R) and electroencephalography (EEG) were administered to all participants. The obese participants were divided into two groups based on the presence or absence of IR, and the data were compared between the subgroups. RESULTS Only verbal scores on the WISC-R in the IR+ group were significantly lower than those of the control and IR- groups. There were no differences between the groups with respect to other parameters of the WISC-R or the EEG. Verbal scores of the WISC-R were negatively correlated with obesity duration and homeostatic model assessment-insulin resistance (HOMA-IR) values. EEGs showed significantly more frequent 'slowing during hyperventilation' (SDHs) in obese children than non-obese children. CONCLUSIONS Neurocognitive functions, particularly verbal abilities, were impaired in obese children with IR. An early examination of cognitive functions may help identify and correct such abnormalities in obese children.
Collapse
|
39
|
Orkaby AR, Cho K, Cormack J, Gagnon DR, Driver JA. Metformin vs sulfonylurea use and risk of dementia in US veterans aged ≥65 years with diabetes. Neurology 2017; 89:1877-1885. [PMID: 28954880 DOI: 10.1212/wnl.0000000000004586] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 08/01/2017] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To determine whether metformin is associated with a lower incidence of dementia than sulfonylureas. METHODS This was a retrospective cohort study of US veterans ≥65 years of age with type 2 diabetes who were new users of metformin or a sulfonylurea and had no dementia. Follow-up began after 2 years of therapy. To account for confounding by indication, we developed a propensity score (PS) and used inverse probability of treatment weighting (IPTW) methods. Cox proportional hazards models estimated the hazard ratio (HR) of incident dementia. RESULTS We identified 17,200 new users of metformin and 11,440 new users of sulfonylureas. Mean age was 73.5 years and mean HbA1c was 6.8%. Over an average follow-up of 5 years, 4,906 cases of dementia were diagnosed. Due to effect modification by age, all analyses were conducted using a piecewise model for age. Crude hazard ratio [HR] for any dementia in metformin vs sulfonylurea users was 0.67 (95% confidence interval [CI] 0.61-0.73) and 0.78 (95% CI 0.72-0.83) for those <75 years of age and ≥75 years of age, respectively. After PS IPTW adjustment, results remained significant in veterans <75 years of age (HR 0.89; 95% CI 0.79-0.99), but not for those ≥75 years of age (HR 0.96; 95% CI 0.87-1.05). A lower risk of dementia was also seen in the subset of younger veterans who had HbA1C values ≥7% (HR 0.76; 95% CI 0.63-0.91), had good renal function (HR 0.86; 95% CI 0.76-0.97), and were white (HR 0.87; 95% CI 0.77-0.99). CONCLUSIONS After accounting for confounding by indication, metformin was associated with a lower risk of subsequent dementia than sulfonylurea use in veterans <75 years of age. Further work is needed to identify which patients may benefit from metformin for the prevention of dementia.
Collapse
Affiliation(s)
- Ariela R Orkaby
- From the VA Boston Geriatric Research, Education, and Clinical Center (GRECC) and Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC) (A.R.O., K.C., J.C., D.R.G., J.A.D.); Division of Aging (A.R.O., K.C., J.A.D.), Brigham & Women's Hospital, Harvard Medical School; and Boston University School of Public Health Department of Biostatistics (D.R.G.), MA.
| | - Kelly Cho
- From the VA Boston Geriatric Research, Education, and Clinical Center (GRECC) and Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC) (A.R.O., K.C., J.C., D.R.G., J.A.D.); Division of Aging (A.R.O., K.C., J.A.D.), Brigham & Women's Hospital, Harvard Medical School; and Boston University School of Public Health Department of Biostatistics (D.R.G.), MA
| | - Jean Cormack
- From the VA Boston Geriatric Research, Education, and Clinical Center (GRECC) and Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC) (A.R.O., K.C., J.C., D.R.G., J.A.D.); Division of Aging (A.R.O., K.C., J.A.D.), Brigham & Women's Hospital, Harvard Medical School; and Boston University School of Public Health Department of Biostatistics (D.R.G.), MA
| | - David R Gagnon
- From the VA Boston Geriatric Research, Education, and Clinical Center (GRECC) and Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC) (A.R.O., K.C., J.C., D.R.G., J.A.D.); Division of Aging (A.R.O., K.C., J.A.D.), Brigham & Women's Hospital, Harvard Medical School; and Boston University School of Public Health Department of Biostatistics (D.R.G.), MA
| | - Jane A Driver
- From the VA Boston Geriatric Research, Education, and Clinical Center (GRECC) and Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC) (A.R.O., K.C., J.C., D.R.G., J.A.D.); Division of Aging (A.R.O., K.C., J.A.D.), Brigham & Women's Hospital, Harvard Medical School; and Boston University School of Public Health Department of Biostatistics (D.R.G.), MA
| |
Collapse
|
40
|
Li L. The Molecular Mechanism of Glucagon-Like Peptide-1 Therapy in Alzheimer's Disease, Based on a Mechanistic Target of Rapamycin Pathway. CNS Drugs 2017; 31:535-549. [PMID: 28540646 DOI: 10.1007/s40263-017-0431-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The mechanistic target of rapamycin (mTOR) is an important molecule that connects aging, lifespan, energy balance, glucose and lipid metabolism, and neurodegeneration. Rapamycin exerts effects in numerous biological activities via its target protein, playing a key role in energy balance, regulation of autophagy, extension of lifespan, immunosuppression, and protection against neurodegeneration. There are many similar pathophysiological processes and molecular pathways between Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM), and pharmacologic agents used to treat T2DM, including glucagon-like peptide-1 (GLP-1) analogs, seem to be beneficial for AD. mTOR mediates the effects of GLP-1 analogs in the treatment of T2DM; hence, I hypothesize that mTOR is a key molecule for mediating the protective effects of GLP-1 for AD.
Collapse
Affiliation(s)
- Lin Li
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
41
|
Association of nerve conduction impairment and insulin resistance in children with obesity. Childs Nerv Syst 2016; 32:2219-2224. [PMID: 27503137 DOI: 10.1007/s00381-016-3210-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/29/2016] [Indexed: 10/21/2022]
Abstract
AIM The objective of our study was to investigate nerve conduction in normoglycemic obese children. METHODS A total of 60 children with obesity (30 female and 30 male) and 30 healthy children (15 female and 15 male) were enrolled in the study. Insulin resistance (IR) and other metabolic disturbances were investigated and nerve conduction was measured in all participants. Obese children were divided into groups according to the presence of IR. All results were compared between these subgroups. RESULTS The nerve conduction velocity (NCV) of motor median nerves in the IR+ group was significantly higher than that in the IR- group and lower than that in the control group. The NCV of the motor peroneal nerve in the IR+ group was significantly lower than that in the IR- group. The sensory nerve action potential (SNAP) of the sensory median nerve was significantly lower in the IR+ group compared to that in the IR- group. The sensory sural nerve's SNAP was significantly lower in the IR+ group than that in the control group. CONCLUSION Nerve conduction tests may help to detect early pathologies in peripheral nerves and to decrease morbidities in obese children.
Collapse
|
42
|
Khallaghi B, Safarian F, Nasoohi S, Ahmadiani A, Dargahi L. Metformin-induced protection against oxidative stress is associated with AKT/mTOR restoration in PC12 cells. Life Sci 2016; 148:286-92. [DOI: 10.1016/j.lfs.2016.02.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 01/26/2016] [Accepted: 02/07/2016] [Indexed: 12/21/2022]
|
43
|
Chen F, Dong RR, Zhong KL, Ghosh A, Tang SS, Long Y, Hu M, Miao MX, Liao JM, Sun HB, Kong LY, Hong H. Antidiabetic drugs restore abnormal transport of amyloid-β across the blood-brain barrier and memory impairment in db/db mice. Neuropharmacology 2015. [PMID: 26211973 DOI: 10.1016/j.neuropharm.2015.07.023] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Previous studies have shown significant changes in amyloid-β (Aβ) transport across the blood-brain barrier (BBB) under diabetic conditions with hypoinsulinemia, which is involved in diabetes-associated cognitive impairment. Present study employed db/db mice with hyperinsulinemia to investigate changes in Aβ transport across the BBB, hippocampal synaptic plasticity, and restorative effects of antidiabetic drugs. Our results showed that db/db mice exhibited similar changes in Aβ transport across the BBB to that of insulin-deficient mice. Chronic treatment of db/db mice with antidiabetic drugs such as metformin, glibenclamide and insulin glargine significantly decreased Aβ influx across the BBB determined by intra-arterial infusion of (125)I-Aβ(1-40), and expression of the receptor for advanced glycation end products (RAGE) participating in Aβ influx. Insulin glargine, but not, metformin or glibenclamide increased Aβ efflux across the BBB determined by stereotaxic intra-cerebral infusion of (125)I-Aβ(1-40), and expression of the low-density lipoprotein receptor related protein 1 (LRP1) participating in Aβ efflux. Moreover, treatment with these drugs significantly decreased hippocampal Aβ(1-40) or Aβ(1-42) and inhibited neuronal apoptosis. The drugs also ameliorated memory impairment confirmed by improved performance on behavioral tasks. However, insulin glargine or glibenclamide, but not metformin, restored hippocampal synaptic plasticity characterized by enhancing in vivo long-term potentiation (LTP). Further study found that these three drugs significantly restrained NF-κB, but only insulin glargine enhanced peroxisome proliferator-activated receptor γ (PPARγ) activity at the BBB in db/db mice. Our data indicate that the antidiabetic drugs can partially restore abnormal Aβ transport across the BBB and memory impairment under diabetic context.
Collapse
Affiliation(s)
- Fang Chen
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Rong Rong Dong
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Kai Long Zhong
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Arijit Ghosh
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Su Su Tang
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Yan Long
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Mei Hu
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Ming Xing Miao
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Jian Min Liao
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Hong Bing Sun
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Ling Yi Kong
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Hong
- Department of Pharmacology, and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
44
|
Yin Z, Yu H, Chen S, Ma C, Ma X, Xu L, Ma Z, Qu R, Ma S. Asiaticoside attenuates diabetes-induced cognition deficits by regulating PI3K/Akt/NF-κB pathway. Behav Brain Res 2015; 292:288-99. [PMID: 26097002 DOI: 10.1016/j.bbr.2015.06.024] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 06/11/2015] [Accepted: 06/13/2015] [Indexed: 10/23/2022]
Abstract
Diabetes-associated cognitive dysfunction, referred as "diabetic encephalopathy", has been confirmed in a great deal of literature. Current evidence support that oxidative stress, inflammation, energy metabolism imbalance, and aberrant insulin signaling are associated with cognition deficits induced by diabetes. The present study explore the effect of asiaticoside on the cognition behaviors, synapses, and oxidative stress in diabetic rats. Asiaticoside could markedly ameliorate the performance in the Morris Water Maze (decreased latency time and path length, and increased time spent in the target quadrant), which was correlated with its capabilities of suppressing oxidative stress, restoring Na(+)-K(+)-ATPase activity and protecting hippocampal synapses. In vitro, asiaticoside could up-regulate synaptic proteins expression via modulating Phosphoinositide 3-kinase (PI3K)/Protein Kinase B(AKT)/Nuclear Factor -kappa B (NF-κB)-mediated inflammatory pathway in SH-SY5Y cells incubated with high glucose chronically. In conclusion, asiaticoside had beneficial effects on the prevention and treatment of diabetes-associated cognitive deficits, which was involved in oxidative stress, PI3K/Akt/NF-κB pathway and synaptic function in the development of cognitive decline induced by diabetes.
Collapse
Affiliation(s)
- Zhujun Yin
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, PR China
| | - Haiyang Yu
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, PR China
| | - She Chen
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, PR China
| | - Chunhua Ma
- School of Life Sciences, Nanjing University, Nanjing 210009, PR China
| | - Xiao Ma
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, PR China
| | - Lixing Xu
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, PR China
| | - Zhanqiang Ma
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, PR China
| | - Rong Qu
- Department of Pharmacology of Traditional Chinese Medical Formulae, Nanjing University of Traditional Chinese Medicine, Nanjing 210029, PR China
| | - Shiping Ma
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
45
|
Rizvi SMD, Shaikh S, Waseem SMA, Shakil S, Abuzenadah AM, Biswas D, Tabrez S, Ashraf GM, Kamal MA. Role of anti-diabetic drugs as therapeutic agents in Alzheimer's disease. EXCLI JOURNAL 2015; 14:684-96. [PMID: 27152105 PMCID: PMC4849108 DOI: 10.17179/excli2015-252] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/05/2015] [Indexed: 12/16/2022]
Abstract
Recent data have suggested a strong possible link between Type 2 Diabetes Mellitus and Alzheimer's disease (AD), although exact mechanisms linking the two are still a matter of research and debate. Interestingly, both are diseases with high incidence and prevalence in later years of life. The link appears so strong that some scientists use Alzheimer's and Type 3 Diabetes interchangeably. In depth study of recent data suggests that the anti diabetic drugs not only have possible role in treatment of Alzheimer's but may also arrest the declining cognitive functions associated with it. The present review gives an insight into the possible links, existing therapeutics and clinical trials of anti diabetic drugs in patients suffering from AD primarily or as co-morbidity. It may be concluded that the possible beneficial effects and usefulness of the current anti diabetic drugs in AD cannot be neglected and further research is required to achieve positive results. Currently, several drug trials are in progress to give conclusive evidence based data.
Collapse
Affiliation(s)
| | | | - Shah Mohammad Abbas Waseem
- Department of Physiology, Integral Institute of Medical Sciences & Research, Integral University, Lucknow, India
| | - Shazi Shakil
- Center of Innovation in Personalized Medicine, Faculty of Applied Medical Sciences,King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adel M. Abuzenadah
- Center of Innovation in Personalized Medicine, Faculty of Applied Medical Sciences,King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md. Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Enzymoic, 7 Peterlee Pl, Hebersham, NSW 2770, Australia
| |
Collapse
|
46
|
Khang R, Park C, Shin JH. Dysregulation of parkin in the substantia nigra of db/db and high-fat diet mice. Neuroscience 2015; 294:182-92. [DOI: 10.1016/j.neuroscience.2015.03.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 03/06/2015] [Accepted: 03/07/2015] [Indexed: 01/28/2023]
|
47
|
El Idrissi A, Sidime F, Tantawy O, Obeysekera D, Wisidagama D, Tariq S, Jmukhadze I, L’Amoreaux WJ. Taurine Supplementation Induces Hyperinsulinemia and Neuronal Hyperexcitability. TAURINE 9 2015; 803:415-23. [DOI: 10.1007/978-3-319-15126-7_32] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
48
|
Werner H, LeRoith D. Insulin and insulin-like growth factor receptors in the brain: physiological and pathological aspects. Eur Neuropsychopharmacol 2014; 24:1947-53. [PMID: 24529663 DOI: 10.1016/j.euroneuro.2014.01.020] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 01/23/2014] [Indexed: 12/31/2022]
Abstract
The involvement of insulin, the insulin-like growth factors (IGF1, IGF2) and their receptors in central nervous system development and function has been the focus of scientific interest for more than 30 years. The insulin-like peptides, both locally-produced proteins as well as those transported from the circulation into the brain via the blood-brain barrier, are involved in a myriad of biological activities. These actions include, among others, neuronal survival, neurogenes, angiogenesis, excitatory and inhibitory neurotransmission, regulation of food intake, and cognition. In recent years, a linkage between brain insulin/IGF1 and certain neuropathologies has been identified. Epidemiological studies have demonstrated a correlation between diabetes (mainly type 2) and Alzheimer׳s disease. In addition, an aberrant decline in IGF1 values was suggested to play a role in the development of Alzheimer׳s disease. The present review focuses on the expression and function of insulin, IGFs and their receptors in the brain in physiological and pathological conditions.
Collapse
Affiliation(s)
- Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Derek LeRoith
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Research Institute at Rambam (LHCRIR), Rambam-Health Care Campus, P.O. Box 9602, Haifa 31096, Israel.
| |
Collapse
|
49
|
Khang R, Park C, Shin JH. The biguanide metformin alters phosphoproteomic profiling in mouse brain. Neurosci Lett 2014; 579:145-50. [PMID: 25067825 DOI: 10.1016/j.neulet.2014.07.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/11/2014] [Accepted: 07/16/2014] [Indexed: 11/29/2022]
Abstract
Metformin, a potent antihyperglycemic agent is recommended as the first-line oral therapy for type 2 diabetes (T2D). Recently, metformin has been reported to be beneficial to neurodegenerative disease models. However, the putative mechanisms underlying the neuroprotective effects of metformin in disease models are unknown. Thus, we applied LC-MS/MS-based pattern analysis and two-dimensional electrophoresis (2DE)-based proteomic approach to understand the global phosphoproteomic alteration in the brain of metformin-administrated mice. Collectively, LC-MS/MS-based pattern analysis reveals that 41 phosphoproteins were upregulated and 22 phosphoproteins were downregulated in the brain of metformin-administrated mice. In addition, 5 differentially expressed phosphoproteins were identified upon metformin administration by 2DE coupled with mass spectrometry. The phosphorylation status of metabolic enzymes was decreased while that of mitochondrial proteins was increased by metformin. Interestingly, phosphorylated α-synuclein was significantly decreased by metformin administration. Taken together, our results might provide potential pathways to understand the pharmacological effect of metformin on neuroprotection.
Collapse
Affiliation(s)
- Rin Khang
- Division of Pharmacology, Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - ChiHu Park
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea; Mass Spectrometry, Research Core Facility, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Joo-Ho Shin
- Division of Pharmacology, Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
50
|
Mansur RB, Cha DS, Woldeyohannes HO, Soczynska JK, Zugman A, Brietzke E, McIntyre RS. Diabetes mellitus and disturbances in brain connectivity: a bidirectional relationship? Neuromolecular Med 2014; 16:658-68. [PMID: 24974228 DOI: 10.1007/s12017-014-8316-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 06/19/2014] [Indexed: 12/26/2022]
Abstract
Diabetes mellitus (DM) is associated with deficits across multiple cognitive domains. The observed impairments in cognitive function are hypothesized to be subserved by alterations in brain structure and function. Several lines of evidence indicate that alterations in glial integrity and function, as well as abnormal synchrony within brain circuits and associated networks, are observed in adults with DM. Microangiopathy and alterations in insulin homeostasis appear to be principal effector systems, although a unitary explanation subsuming the complex etiopathology of white matter in DM is unavailable. A contemporary model of disease pathophysiology for several mental disorders, including but not limited to mood disorders, posits abnormalities in the synchronization of cellular systems in circuits. The observation that similar abnormalities occur in diabetic populations provides the basis for hypothesizing the convergence of pathoetiological factors. Herein, we propose that abnormal structure, function and chemical composition as well as synchrony within and between circuits is an accompaniment of DM and is shared in common with several mental disorders.
Collapse
Affiliation(s)
- Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit (MDPU), University Health Network, University of Toronto, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada,
| | | | | | | | | | | | | |
Collapse
|