251
|
Rohrer J, Lupo N, Bernkop-Schnürch A. Advanced formulations for intranasal delivery of biologics. Int J Pharm 2018; 553:8-20. [PMID: 30316796 DOI: 10.1016/j.ijpharm.2018.10.029] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The global biologics market has been ever increasing over the last decades and is predicted to top Euro 350 by 2020. Facing this scenario, the parenteral route of biologics administration as hitherto standard route is inconvenient for the future. Among the alternatives, the intranasal delivery of therapeutic biologicals seems to be most promising but researchers are still facing challenges as indicated by the scarce number of successfully marketed peptide drugs. AREAS COVERED This review article is a compilation of current research focusing on achievements in the field of auxiliary agents for biologics delivery. First, the key benefits of the nose as most promising alternative route of drug administration are highlighted. Then, the potential of the different auxiliary agents in preclinical research is in detail discussed. Moreover, the most used permeation enhancing agents, mucolytic agents, mucoadhesive agents, in situ gelling agents and enzyme inhibiting agents in the formulation of nasal drug delivery systems are described. Thus, the overall purpose of this review is to highlight recent achievements in nasal delivery of biologics and to encourage researchers to work in the direction of needle-free nasal administration of biologics. EXPERT OPINION The nasal epithelium is a promising route for biologics administration, which is reflected in a number of well-established products on the market treating chronic diseases as well as a large number of clinical trials currently in progress. The nasal route of drug administration might be a chance to improve therapy of biologics however break-through advances, especially for very complex molecules, such as antibodies, are still needed.
Collapse
Affiliation(s)
- Julia Rohrer
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck, Austria
| | - Noemi Lupo
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck, Austria.
| |
Collapse
|
252
|
Jiang R, Prell C, Lönnerdal B. Milk osteopontin promotes brain development by up-regulating osteopontin in the brain in early life. FASEB J 2018; 33:1681-1694. [PMID: 30199283 DOI: 10.1096/fj.201701290rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Osteopontin (OPN) is a pleiotropic protein and is abundantly present in milk. Its functions include immune modulation and cellular proliferation and differentiation. OPN is highly expressed in the brain. We investigated the effects of milk-derived OPN on brain development of mouse pups. Wild-type (WT) dams producing OPN+ milk and OPN knockout (KO) dams producing OPN- milk nursed WT pups (OPN+/+), yielding 2 pup treatment groups, OPN+ OPN+/+ and OPN- OPN+/+, for comparison. Preliminary studies supported use of this model by showing high concentrations of OPN in milk of WT dams and no OPN in milk of OPN KO dams, and production of similar amounts of milk by WT and KO dams. The ability of ingested milk OPN to enter the brain was revealed by appearance of orally gavaged [125I]-labeled and antibody-probed milk OPN in brains of pups. Brain OPN mRNA levels were similar in both nursed groups, but the brain OPN protein level was significantly lower in the OPN- OPN+/+ group at postnatal days 6 and 8. Behavior tests showed impaired memory and learning ability in OPN- OPN+/+ pups. In addition, our study revealed increased expression of myelination-related proteins and elevated proliferation and differentiation of NG-2 glia into oligodendrocytes in the brain of OPN+ OPN+/+ pups, accompanied by increased activation of ERK-1/2 and PI3K/Akt signaling. We concluded that milk OPN can play an important role in brain development and behavior in infancy by promoting myelination.-Jiang, R., Prell, C., Lönnerdal, B. Milk osteopontin promotes brain development by up-regulating osteopontin in the brain in early life.
Collapse
Affiliation(s)
- Rulan Jiang
- Department of Nutrition, University of California Davis, Davis, California, USA
| | - Christine Prell
- Dr. von Hauner Children's Hospital, Ludwig Maximilians University, Munich, Germany
| | - Bo Lönnerdal
- Department of Nutrition, University of California Davis, Davis, California, USA
| |
Collapse
|
253
|
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: 115] [Impact Index Per Article: 16.4] [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
|
254
|
Rhea EM, Rask-Madsen C, Banks WA. Insulin transport across the blood-brain barrier can occur independently of the insulin receptor. J Physiol 2018; 596:4753-4765. [PMID: 30044494 DOI: 10.1113/jp276149] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 07/23/2018] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Insulin enters the brain from the blood via a saturable transport system. It is unclear how insulin is transported across the blood-brain barrier (BBB). Using two models of the signalling-related insulin receptor loss or inhibition, we show insulin transport can occur in vivo without the signalling-related insulin receptor. Insulin in the brain has multiple roles including acting as a metabolic regulator and improving memory. Understanding how insulin is transported across the BBB will aid in developing therapeutics to further increase CNS concentrations. ABSTRACT A saturable system transports insulin from blood across the blood-brain barrier (BBB) and into the central nervous system. Whether or not the classic or signalling-related insulin receptor plays a role in mediating this transport in vivo is controversial. Here, we employed kinetics methods that distinguish between transport across the brain endothelial cell and reversible luminal surface receptor binding. Using a previously established line of mice with endothelial-specific loss of the signalling-related insulin receptor (EndoIRKO) or inhibiting the insulin receptor with the selective antagonist S961, we show insulin transport across the BBB is maintained. Rates of insulin transport were similar in all groups and transport was still saturable. Unlike transport, binding of insulin to the brain endothelial cell was decreased with the loss or inhibition of the signalling-related insulin receptor. These findings demonstrate that the signalling-related insulin receptor is not required for insulin transport across the BBB.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Research and Development, VA Puget Sound, Seattle, WA, USA.,Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | | | - William A Banks
- Research and Development, VA Puget Sound, Seattle, WA, USA.,Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
255
|
Phillips OR, Onopa AK, Zaiko YV, Singh MK. Insulin resistance is associated with smaller brain volumes in a preliminary study of depressed and obese children. Pediatr Diabetes 2018; 19:892-897. [PMID: 29569318 PMCID: PMC6030449 DOI: 10.1111/pedi.12672] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/19/2018] [Accepted: 03/14/2018] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE During childhood, the brain can consume up to 65% of total calories, and a steady supply of the brain's main fuel glucose needs to be maintained. Although the brain itself is not dependent on insulin for the uptake of glucose, insulin plays an important role in energy homeostasis. Thus, the risk for insulin resistance during brain development may negatively impact the whole brain volume. METHODS We investigated the link between the insulin resistance and the whole brain volume as measured by structural Magnetic resonance imaging (MRI) in 46 unmedicated depressed and overweight youths between the ages of 9 and 17 years. RESULTS Smaller whole brain volumes were associated with insulin resistance independent of age, sex, depression severity, body mass index, socioeconomic status, Tanner Stage, and Intelligence quotient (IQ) (r = 0.395, P = .014) CONCLUSIONS There may be a significant cost for developing insulin resistance on the developing brain. Disentangling the precise relationship between the insulin resistance and the developing brain is critical.
Collapse
Affiliation(s)
- Owen R. Phillips
- Department of Psychiatry, Division of Child and Adolescent Psychiatry, Stanford University School of Medicine, Stanford Pediatric Mood Disorders Program, Division of Child and Adolescent Psychiatry, 401 Quarry Road, Stanford, CA 94305-5719,Corresponding Author: Owen Phillips, PhD, Postdoctoral Scholar, Stanford Pediatric Mood Disorders Program, Division of Child and Adolescent Psychiatry, 401 Quarry Road, Stanford, CA 94305-5719 (; Phone: (650) 725-5922; Fax: (650) 724-7389). Website: med.stanford.edu/pedmood
| | - Alexander K. Onopa
- Department of Psychiatry, Division of Child and Adolescent Psychiatry, Stanford University School of Medicine, Stanford Pediatric Mood Disorders Program, Division of Child and Adolescent Psychiatry, 401 Quarry Road, Stanford, CA 94305-5719
| | - Yevgeniya V. Zaiko
- Department of Psychiatry, Division of Child and Adolescent Psychiatry, Stanford University School of Medicine, Stanford Pediatric Mood Disorders Program, Division of Child and Adolescent Psychiatry, 401 Quarry Road, Stanford, CA 94305-5719
| | - Manpreet K. Singh
- Department of Psychiatry, Division of Child and Adolescent Psychiatry, Stanford University School of Medicine, Stanford Pediatric Mood Disorders Program, Division of Child and Adolescent Psychiatry, 401 Quarry Road, Stanford, CA 94305-5719
| |
Collapse
|
256
|
Zhong KL, Chen F, Hong H, Ke X, Lv YG, Tang SS, Zhu YB. New views and possibilities of antidiabetic drugs in treating and/or preventing mild cognitive impairment and Alzheimer's Disease. Metab Brain Dis 2018; 33:1009-1018. [PMID: 29626315 DOI: 10.1007/s11011-018-0227-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 03/28/2018] [Indexed: 12/13/2022]
Abstract
Mounting evidence suggests that diabetes mellitus (DM) is associated with mild cognitive impairment (MCI), vascular dementia and Alzheimer's disease (AD). Biological, clinical and epidemiological data support a close link between DM and AD. Increasingly, studies have found that several antidiabetic agents can promote neurogenesis, and clinically ameliorate cognitive and memory impairments in different clinical settings. Data has shown that these antidiabetic drugs positively affect mitochondrial and synaptic function, neuroinflammation, and brain metabolism. Evidence to date strongly suggests that these antidiabetic drugs could be developed as disease-modifying therapies for MCI and AD in patients with and without diabetes.
Collapse
Affiliation(s)
- Kai Long Zhong
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Fang Chen
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Hao Hong
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Xuan Ke
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Yang Ge Lv
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Su Su Tang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Yu Bing Zhu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
257
|
Lin Y, Wang K, Ma C, Wang X, Gong Z, Zhang R, Zang D, Cheng Y. Evaluation of Metformin on Cognitive Improvement in Patients With Non-dementia Vascular Cognitive Impairment and Abnormal Glucose Metabolism. Front Aging Neurosci 2018; 10:227. [PMID: 30100873 PMCID: PMC6074058 DOI: 10.3389/fnagi.2018.00227] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/09/2018] [Indexed: 12/19/2022] Open
Abstract
Objective: Recent studies have suggested that metformin can penetrate the blood–brain barrier, protecting neurons via anti-inflammatory action and improvement of brain energy metabolism. In this study, we aim to investigate the effect of metformin on cognitive function in patients with abnormal glucose metabolism and non-dementia vascular cognitive impairment (NDVCI). Methods: One hundred patients with NDVCI and abnormal glucose metabolism were randomly allocated into two groups: metformin and donepezil (n = 50) or acarbose and donepezil (n = 50). The neuropsychological status, glucose metabolism, and common carotid arteries intima–media thickness (CCA-IMT) before and after a year of treatment, were measured and compared between the groups. Results: Ninety four patients completed all the assessment and follow-up. After a year of treatment, there was a decrease in Alzheimer’s disease Assessment Scale-Cognitive Subscale scores and the duration of the Trail Making Test in the metformin-donepezil group. Furthermore, these patients showed a significant increase in World Health Organization–University of California–Los Angeles Auditory Verbal Learning Test scores after treatment (all P < 0.05). However, there was no obvious improvement in cognitive function in the acarbose-donepezil group. We also observed a significant decrease in the level of fasting insulin and insulin resistance (IR) index in the metformin-donepezil group, with a lower CCA-IMT value than that in the acarbose-donepezil group after a year of treatment (P < 0.05). Conclusion: We conclude that metformin can improve cognitive function in patients with NDVCI and abnormal glucose metabolism, especially in terms of performance function. Improved cognitive function may be related to improvement of IR and the attenuated progression of IMT. Trial Registration:ChiCTR-IPR-17011855.
Collapse
Affiliation(s)
- Yufeng Lin
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China.,Department of Neurology, Tianjin First Center Clinical College of Tianjin Medical University, Tianjin, China
| | - Kaiyuan Wang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chunchao Ma
- Department of Neurology, Tianjin First Center Clinical College of Tianjin Medical University, Tianjin, China
| | - Xuesong Wang
- Department of Neurology, Tianjin First Center Clinical College of Tianjin Medical University, Tianjin, China
| | - Zhongying Gong
- Department of Neurology, Tianjin First Center Clinical College of Tianjin Medical University, Tianjin, China
| | - Rui Zhang
- Department of Neurology, Tianjin First Center Clinical College of Tianjin Medical University, Tianjin, China
| | - Dawei Zang
- Department of Neurology, Tianjin First Center Clinical College of Tianjin Medical University, Tianjin, China
| | - Yan Cheng
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
258
|
Khanna S, Domingo-Fernández D, Iyappan A, Emon MA, Hofmann-Apitius M, Fröhlich H. Using Multi-Scale Genetic, Neuroimaging and Clinical Data for Predicting Alzheimer's Disease and Reconstruction of Relevant Biological Mechanisms. Sci Rep 2018; 8:11173. [PMID: 30042519 PMCID: PMC6057884 DOI: 10.1038/s41598-018-29433-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 06/29/2018] [Indexed: 01/02/2023] Open
Abstract
Alzheimer's Disease (AD) is among the most frequent neuro-degenerative diseases. Early diagnosis is essential for successful disease management and chance to attenuate symptoms by disease modifying drugs. In the past, a number of cerebrospinal fluid (CSF), plasma and neuro-imaging based biomarkers have been proposed. Still, in current clinical practice, AD diagnosis cannot be made until the patient shows clear signs of cognitive decline, which can partially be attributed to the multi-factorial nature of AD. In this work, we integrated genotype information, neuro-imaging as well as clinical data (including neuro-psychological measures) from ~900 normal and mild cognitively impaired (MCI) individuals and developed a highly accurate machine learning model to predict the time until AD is diagnosed. We performed an in-depth investigation of the relevant baseline characteristics that contributed to the AD risk prediction. More specifically, we used Bayesian Networks to uncover the interplay across biological scales between neuro-psychological assessment scores, single genetic variants, pathways and neuro-imaging related features. Together with information extracted from the literature, this allowed us to partially reconstruct biological mechanisms that could play a role in the conversion of normal/MCI into AD pathology. This in turn may open the door to novel therapeutic options in the future.
Collapse
Affiliation(s)
- Shashank Khanna
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, Sankt Augustin, 53754, Germany.,Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany
| | - Daniel Domingo-Fernández
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, Sankt Augustin, 53754, Germany.,Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany
| | - Anandhi Iyappan
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, Sankt Augustin, 53754, Germany.,Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany
| | - Mohammad Asif Emon
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, Sankt Augustin, 53754, Germany.,Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, Sankt Augustin, 53754, Germany.,Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany
| | - Holger Fröhlich
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany. .,UCB Biosciences GmbH, Alfred-Nobel Str. 10, 40789, Monheim, Germany.
| |
Collapse
|
259
|
Valian N, Ahmadiani A, Dargahi L. Increasing methamphetamine doses inhibit glycogen synthase kinase 3β activity by stimulating the insulin signaling pathway in substantia nigra. J Cell Biochem 2018; 119:8522-8530. [PMID: 30011098 DOI: 10.1002/jcb.27082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/26/2018] [Indexed: 01/01/2023]
Abstract
Methamphetamine (MA), a highly abused psychostimulant, exerts neurotoxic effects on the dopaminergic system via several neurotoxicity mechanisms in the long-term administration. Since the effect of MA on the signaling insulin pathway is less studied, the current study was designed to evaluate the effect of escalating an MA regimen on different insulin signaling elements in substantia nigra (SN) and striatum of a rat. Increasing MA doses (1-14 mg/kg) were administrated intraperitoneally twice a day for 14 days in rats. In the control group, normal saline was injected in the same volume. On days 1, 14, 28, and 60 after MA discontinuation, molecular assessments were performed. Insulin receptor (IR) and insulin receptor substrate (IRS) 1 and 2 gene expression were evaluated using real-time polymerase chain reaction, and protein levels of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K), phospho-PI3K, Akt, phospho-Akt, glycogen synthase kinase 3β (GSK3β), and phospho-GSK3β were measured by the Western blot analysis in SN and striatum. Messenger RNA levels of IR and insulin receptor substrate 2 were increased in SN, 1 day after the last injection. Although no changes were observed in PI3K, phospho-PI3K, Akt, phospho-Akt, and GSK3β levels, increase in the level of inactive form of GSK3β (phosphorylated on serine 9) was indicated in SN on day 28. In striatum, decreases in IR and phospho-Akt were demonstrated, without any change in other elements. Repeated escalating regimen of MA activated the insulin signaling pathway and inhibited GSK3β activity in SN. This response, which did not occur in striatum, may act as an adaptive mechanism to prevent MA-induced neurotoxicity in dopaminergic cell bodies.
Collapse
Affiliation(s)
- Neda Valian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University ofMedical Sciences, Tehran, Iran
| |
Collapse
|
260
|
Geijselaers SLC, Aalten P, Ramakers IHGB, De Deyn PP, Heijboer AC, Koek HL, OldeRikkert MGM, Papma JM, Reesink FE, Smits LL, Stehouwer CDA, Teunissen CE, Verhey FRJ, van der Flier WM, Biessels GJ. Association of Cerebrospinal Fluid (CSF) Insulin with Cognitive Performance and CSF Biomarkers of Alzheimer's Disease. J Alzheimers Dis 2018; 61:309-320. [PMID: 29154275 PMCID: PMC5734123 DOI: 10.3233/jad-170522] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background: Abnormal insulin signaling in the brain has been linked to Alzheimer’s disease (AD). Objective: To evaluate whether cerebrospinal fluid (CSF) insulin levels are associated with cognitive performance and CSF amyloid-β and Tau. Additionally, we explore whether any such association differs by sex or APOE ɛ4 genotype. Methods: From 258 individuals participating in the Parelsnoer Institute Neurodegenerative Diseases, a nationwide multicenter memory clinic population, we selected 138 individuals (mean age 66±9 years, 65.2% male) diagnosed with subjective cognitive impairment (n = 45), amnestic mild cognitive impairment (n = 44), or AD (n = 49), who completed a neuropsychological assessment, including tests of global cognition and memory performance, and who underwent lumbar puncture. We measured CSF levels of insulin, amyloid-β1-42, total (t-)Tau, and phosphorylated (p-)Tau. Results: CSF insulin levels did not differ between the diagnostic groups (p = 0.136). Across the whole study population, CSF insulin was unrelated to cognitive performance and CSF biomarkers of AD, after adjustment for age, sex, body mass index, diabetes status, and clinic site (all p≥0.131). Importantly, however, we observed effect modification by sex and APOE ɛ4 genotype. Specifically, among women, higher insulin levels in the CSF were associated with worse global cognition (standardized regression coefficient –0.483; p = 0.008) and higher p-Tau levels (0.353; p = 0.040). Among non-carriers of the APOE ɛ4 allele, higher CSF insulin was associated with higher t-Tau (0.287; p = 0.008) and p-Tau (0.246; p = 0.029). Conclusion: Our findings provide further evidence for a relationship between brain insulin signaling and AD pathology. It also highlights the need to consider sex and APOE ɛ4 genotype when assessing the role of insulin.
Collapse
Affiliation(s)
- Stefan L C Geijselaers
- Departments of Neurology and Geriatrics Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, the Netherlands.,Department of Internal Medicine and Cardiovascular Research Institute, Maastricht University Medical Centre +, Maastricht, the Netherlands
| | - Pauline Aalten
- Alzheimer Centre Limburg, School for Mental Health and Neuroscience (MHeNS), Maastricht University Medical Centre +, Maastricht, the Netherlands
| | - Inez H G B Ramakers
- Alzheimer Centre Limburg, School for Mental Health and Neuroscience (MHeNS), Maastricht University Medical Centre +, Maastricht, the Netherlands
| | - Peter Paul De Deyn
- Department of Neurology and Alzheimer Research Centre, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Annemieke C Heijboer
- Department of Clinical Chemistry, Endocrine Laboratory, VU University Medical Centre, Amsterdam, the Netherlands
| | - Huiberdina L Koek
- Departments of Neurology and Geriatrics Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Marcel G M OldeRikkert
- Radboudumc Alzheimer Centre, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Janne M Papma
- Departments of Neurology and Radiology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Fransje E Reesink
- Department of Neurology and Alzheimer Research Centre, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Lieke L Smits
- Alzheimer Centre Amsterdam, VU University Medical Centre, Amsterdam, the Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine and Cardiovascular Research Institute, Maastricht University Medical Centre +, Maastricht, the Netherlands
| | - Charlotte E Teunissen
- Department of Clinical Chemistry, Neurochemistry Laboratory and Biobank, VU University Medical Centre, Amsterdam, the Netherlands
| | - Frans R J Verhey
- Alzheimer Centre Limburg, School for Mental Health and Neuroscience (MHeNS), Maastricht University Medical Centre +, Maastricht, the Netherlands
| | | | - Geert Jan Biessels
- Department of Internal Medicine and Cardiovascular Research Institute, Maastricht University Medical Centre +, Maastricht, the Netherlands
| | | |
Collapse
|
261
|
Mehlig K, Lapidus L, Thelle DS, Waern M, Zetterberg H, Björkelund C, Skoog I, Lissner L. Low fasting serum insulin and dementia in nondiabetic women followed for 34 years. Neurology 2018; 91:e427-e435. [PMID: 29997193 PMCID: PMC6093770 DOI: 10.1212/wnl.0000000000005911] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 04/25/2018] [Indexed: 12/03/2022] Open
Abstract
Objective In a representative population of women followed over 34 years, we investigated the prospective association between fasting serum insulin and dementia, taking into account the incidence of diabetes mellitus. Methods Fasting values for serum insulin and blood glucose were obtained in 1,212 nondiabetic women 38 to 60 years of age at the 1968 baseline. Risk of dementia was assessed by Cox proportional hazard regression with adjustment for insulin, glucose, and other covariates and, in a second model, after censoring for incident cases of diabetes mellitus. Incident diabetes mellitus was considered as a third endpoint for comparison with dementia. Results Over 34 years, we observed 142 incident cases of dementia. The low tertile of insulin displayed excess risk for dementia (hazard ratio [HR] 2.34, 95% confidence interval [CI] 1.52–3.58) compared to the medium tertile, but the high tertile of insulin did not (HR 1.28, 95% CI 0.81–2.03). These associations were also seen for dementia without diabetes comorbidity. In contrast, high but not low insulin predicted incident diabetes mellitus (115 cases) (HR 1.70, 95% CI 1.08–2.68 and HR 0.76, 95% CI 0.43–1.37, respectively). Conclusion A previous study reported a U-shaped association between fasting insulin and dementia in a 5-year follow-up of elderly men. Our results confirmed a nonlinear association in a female population, with high risk at low insulin values that was not attributable to preclinical dementia or impaired insulin secretion. This condition suggests a new pathway to dementia, which differs from the metabolic pathway involving diabetes mellitus.
Collapse
Affiliation(s)
- Kirsten Mehlig
- From the Institutes of Medicine (K.M., L.L., D.S.T., C.B., L.L.) and Neuroscience and Physiology (M.W., H.Z., I.S.), University of Gothenburg, Sweden; Institute of Basic Medical Sciences (D.S.T.), University of Oslo, Norway; UCL Institute of Neurology (H.Z.), Queen Square; UK Dementia Research Institute (H.Z.), London; and Clinical Neurochemistry Laboratory (H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden.
| | - Leif Lapidus
- From the Institutes of Medicine (K.M., L.L., D.S.T., C.B., L.L.) and Neuroscience and Physiology (M.W., H.Z., I.S.), University of Gothenburg, Sweden; Institute of Basic Medical Sciences (D.S.T.), University of Oslo, Norway; UCL Institute of Neurology (H.Z.), Queen Square; UK Dementia Research Institute (H.Z.), London; and Clinical Neurochemistry Laboratory (H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden
| | - Dag S Thelle
- From the Institutes of Medicine (K.M., L.L., D.S.T., C.B., L.L.) and Neuroscience and Physiology (M.W., H.Z., I.S.), University of Gothenburg, Sweden; Institute of Basic Medical Sciences (D.S.T.), University of Oslo, Norway; UCL Institute of Neurology (H.Z.), Queen Square; UK Dementia Research Institute (H.Z.), London; and Clinical Neurochemistry Laboratory (H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden
| | - Margda Waern
- From the Institutes of Medicine (K.M., L.L., D.S.T., C.B., L.L.) and Neuroscience and Physiology (M.W., H.Z., I.S.), University of Gothenburg, Sweden; Institute of Basic Medical Sciences (D.S.T.), University of Oslo, Norway; UCL Institute of Neurology (H.Z.), Queen Square; UK Dementia Research Institute (H.Z.), London; and Clinical Neurochemistry Laboratory (H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- From the Institutes of Medicine (K.M., L.L., D.S.T., C.B., L.L.) and Neuroscience and Physiology (M.W., H.Z., I.S.), University of Gothenburg, Sweden; Institute of Basic Medical Sciences (D.S.T.), University of Oslo, Norway; UCL Institute of Neurology (H.Z.), Queen Square; UK Dementia Research Institute (H.Z.), London; and Clinical Neurochemistry Laboratory (H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden
| | - Cecilia Björkelund
- From the Institutes of Medicine (K.M., L.L., D.S.T., C.B., L.L.) and Neuroscience and Physiology (M.W., H.Z., I.S.), University of Gothenburg, Sweden; Institute of Basic Medical Sciences (D.S.T.), University of Oslo, Norway; UCL Institute of Neurology (H.Z.), Queen Square; UK Dementia Research Institute (H.Z.), London; and Clinical Neurochemistry Laboratory (H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ingmar Skoog
- From the Institutes of Medicine (K.M., L.L., D.S.T., C.B., L.L.) and Neuroscience and Physiology (M.W., H.Z., I.S.), University of Gothenburg, Sweden; Institute of Basic Medical Sciences (D.S.T.), University of Oslo, Norway; UCL Institute of Neurology (H.Z.), Queen Square; UK Dementia Research Institute (H.Z.), London; and Clinical Neurochemistry Laboratory (H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden
| | - Lauren Lissner
- From the Institutes of Medicine (K.M., L.L., D.S.T., C.B., L.L.) and Neuroscience and Physiology (M.W., H.Z., I.S.), University of Gothenburg, Sweden; Institute of Basic Medical Sciences (D.S.T.), University of Oslo, Norway; UCL Institute of Neurology (H.Z.), Queen Square; UK Dementia Research Institute (H.Z.), London; and Clinical Neurochemistry Laboratory (H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
262
|
Duarte A, Santos M, Oliveira C, Moreira P. Brain insulin signalling, glucose metabolism and females' reproductive aging: A dangerous triad in Alzheimer's disease. Neuropharmacology 2018; 136:223-242. [PMID: 29471055 DOI: 10.1016/j.neuropharm.2018.01.044] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/22/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
|
263
|
Cairns K, McCarvill T, Ruzickova M, Calkin CV. Course of bipolar illness worsens after onset of insulin resistance. J Psychiatr Res 2018; 102:34-37. [PMID: 29579625 DOI: 10.1016/j.jpsychires.2018.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/12/2018] [Accepted: 03/13/2018] [Indexed: 12/25/2022]
Abstract
Cross-sectional studies indicate that comorbid insulin resistance (IR) and type 2 diabetes are associated with a more severe course of bipolar disorder (BD); however, this relationship has not previously been assessed longitudinally. To address this, we reviewed health records of a case series of six patients with BD and comorbid IR. Severity and length of affective episodes (both mania and depression) over the lifetime were recorded using the Affective Morbidity Index; these data were obtained from ongoing prospective follow-up and from detailed retrospective chart reviews. All six patients with a previously episodic, relapsing-remitting course of illness experienced a worsening of morbidity after the onset of laboratory-demonstrated IR. These results suggest that IR may be a potential testable, modifiable factor in the progression of BD from a treatment responsive (episodic) to a non-responsive (chronic) course of illness.
Collapse
Affiliation(s)
- Kathleen Cairns
- Nova Scotia Health Authority, Abbie J. Lane Memorial Building, Room 3089, 5909 Veterans' Memorial Lane, Halifax, NS, B3H 2E2, Canada
| | - Terrence McCarvill
- Department of Psychiatry, Dalhousie University, Abbie J. Lane Memorial Building, Room 3094, 5909 Veterans' Memorial Lane, Halifax, NS, B3H 2E2, Canada; Operational Trauma/Stress Support Centre, Suite 210, 100 Eileen Stubbs Avenue, Dartmouth, NS, B3B 1Y6, Canada
| | - Martina Ruzickova
- Department of Psychiatry, Dalhousie University, Abbie J. Lane Memorial Building, Room 3094, 5909 Veterans' Memorial Lane, Halifax, NS, B3H 2E2, Canada
| | - Cynthia V Calkin
- Department of Psychiatry, Dalhousie University, Abbie J. Lane Memorial Building, Room 3094, 5909 Veterans' Memorial Lane, Halifax, NS, B3H 2E2, Canada; Department of Medical Neuroscience, Dalhousie University, Abbie J. Lane Memorial Building, Room 3095, 5909 Veterans' Memorial Lane, Halifax, NS, B3H 2E2, Canada.
| |
Collapse
|
264
|
Sachdeva AK, Dharavath RN, Chopra K. Time-response studies on development of cognitive deficits in an experimental model of insulin resistance. Clin Nutr 2018; 38:1447-1456. [PMID: 30037709 DOI: 10.1016/j.clnu.2018.06.966] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 05/30/2018] [Accepted: 06/19/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Alzheimer's disease is suggested to be primarily metabolic, mainly characterized by brain insulin resistance. Chronic fructose feeding results in hippocampal insulin resistance. However, variable opinion exists regarding the concentration and duration of fructose feeding to trigger insulin resistance and resultant cognitive insults. Therefore this study was planned to construct a time-response curve of the appearance of fructose-induced insulin resistance and memory insufficiencies in rats over a period of 24 weeks. Further, Pearson's correlations were drawn between indices of insulin resistance and markers of memory deficits at various time points. METHODS Male Wistar rats (6 weeks old; 155 ± 5 g) were fed with 15% fructose in normal drinking water for a period of 24 weeks. Body weight, food and water intake were weekly monitored. Fasting blood glucose, glycosylated hemoglobin (HbA1C), lipid profiling, plasma insulin, HOMA-IR index, and systolic blood pressure were estimated to confirm the manifestation of insulin resistance. Cognitive derangements were evaluated by Elevated plus maze and Morris water maze at different time points during the study. RESULTS Most of the parameters including insulin resistance became evident at the 7th week and continued until the end of study (24th week) whereas cognitive insufficiency became significantly distinct at the 20th, 22nd and 24th week. Significantly increased serum nitro-oxidative stress, inflammatory cytokines and serum homocysteine levels were intensely connected with fructose-induced neuronal deficits. CONCLUSIONS The construction of time response study reveals that the hallmark characteristics of insulin resistance appear from the 7th week of fructose feeding whereas the cognitive dysfunction appears on the 20th week and both persist till the end of the study. Fructose-induced oxidative stress and neuroinflammation plausibly impair neuronal signaling and synaptic plasticity.
Collapse
Affiliation(s)
- Anand Kamal Sachdeva
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India
| | - Ravinder Naik Dharavath
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India
| | - Kanwaljit Chopra
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India.
| |
Collapse
|
265
|
Rodriguez‐Otormin F, Duro‐Castano A, Conejos‐Sánchez I, Vicent MJ. Envisioning the future of polymer therapeutics for brain disorders. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 11:e1532. [DOI: 10.1002/wnan.1532] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/24/2018] [Accepted: 05/09/2018] [Indexed: 01/09/2023]
Affiliation(s)
| | - Aroa Duro‐Castano
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Valencia Spain
| | | | - María J. Vicent
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Valencia Spain
| |
Collapse
|
266
|
Kabat GC, Rohan TE. Adiposity at different periods of life and risk of adult glioma in a cohort of postmenopausal women. Cancer Epidemiol 2018; 54:71-74. [PMID: 29655085 DOI: 10.1016/j.canep.2018.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 02/28/2018] [Accepted: 03/14/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND Little is known about risk factors for adult glioma. Adiposity has received some attention as a possible risk factor. METHODS We examined the association of body mass index (BMI), waist circumference (WC) and waist-to-hip ratio (WHR), measured at enrollment, as well as self-reported weight earlier in life, with risk of glioma in a large cohort of postmenopausal women. Over 18 years of follow-up, 217 glioma cases were ascertained, including 164 glioblastomas. Cox proportional hazards models were used to estimate hazard ratios and 95% confidence intervals. RESULTS There was a modest, non-significant trend toward increasing risk of glioma and glioblastoma with increasing measured BMI and WHR. No trend was seen for WC. Self-reported BMI earlier in life showed no association with risk. CONCLUSIONS Our weak findings regarding the association of adiposity measures with risk of glioma are in agreement the results of several large cohort studies. In view of the available evidence, adiposity is unlikely to represent an important risk factor for glioma.
Collapse
Affiliation(s)
- Geoffrey C Kabat
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| |
Collapse
|
267
|
Shahriyary L, Riazi G, Lornejad MR, Ghezlou M, Bigdeli B, Delavari B, Mamashli F, Abbasi S, Davoodi J, Saboury AA. Effect of glycated insulin on the blood-brain barrier permeability: An in vitro study. Arch Biochem Biophys 2018; 647:54-66. [DOI: 10.1016/j.abb.2018.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/24/2018] [Accepted: 02/07/2018] [Indexed: 02/08/2023]
|
268
|
Nedelcovych MT, Gadiano AJ, Wu Y, Manning AA, Thomas AG, Khuder SS, Yoo SW, Xu J, McArthur JC, Haughey NJ, Volsky DJ, Rais R, Slusher BS. Pharmacokinetics of Intranasal versus Subcutaneous Insulin in the Mouse. ACS Chem Neurosci 2018; 9:809-816. [PMID: 29257872 PMCID: PMC5906198 DOI: 10.1021/acschemneuro.7b00434] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Insulin delivery to the brain has emerged as an important therapeutic target for cognitive disorders associated with abnormal brain energy metabolism. Although insulin is transported across the blood-brain barrier, peripheral routes of administration are problematic due to systemic effects of insulin on blood glucose. Intranasal (IN) administration is being investigated as an alternative route. We conducted a head-to-head comparison of subcutaneous (SC) and IN insulin, assessing plasma and brain pharmacokinetics and blood glucose levels in the mouse. SC insulin (2.4 IU) achieved therapeutically relevant concentrations in the brain (AUCbrain = 2537 h·μIU/mL) but dramatically increased plasma insulin (AUCplasma = 520 351 h·*μIU/mL), resulting in severe hypoglycemia and in some cases death. IN administration of the same dose resulted in similar insulin levels in the brain (AUCbrain = 3442 h·μIU/mL) but substantially lower plasma concentrations (AUCplasma = 354 h·μIU/mL), amounting to a ∼ 2000-fold increase in the AUCbrain:plasma ratio relative to SC. IN dosing also had no significant effect on blood glucose. When administered daily for 9 days, IN insulin increased brain glucose and energy metabolite concentrations (e.g., adenosine triphosphate and phosphocreatine) without causing overt toxicity, suggesting that IN insulin may be a safe therapeutic option for cognitively impaired patients.
Collapse
Affiliation(s)
- Michael T. Nedelcovych
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Alexandra J. Gadiano
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Arena A. Manning
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Saja S. Khuder
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neuroimmunology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Seung-Wan Yoo
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Jiadi Xu
- Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Justin C. McArthur
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Norman J. Haughey
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - David J. Volsky
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
269
|
Yulyaningsih E, Rudenko IA, Valdearcos M, Dahlén E, Vagena E, Chan A, Alvarez-Buylla A, Vaisse C, Koliwad SK, Xu AW. Acute Lesioning and Rapid Repair of Hypothalamic Neurons outside the Blood-Brain Barrier. Cell Rep 2018; 19:2257-2271. [PMID: 28614713 DOI: 10.1016/j.celrep.2017.05.060] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/28/2017] [Accepted: 05/17/2017] [Indexed: 11/27/2022] Open
Abstract
Neurons expressing agouti-related protein (AgRP) are essential for feeding. The majority of these neurons are located outside the blood-brain barrier (BBB), allowing them to directly sense circulating metabolic factors. Here, we show that, in adult mice, AgRP neurons outside the BBB (AgRPOBBB) were rapidly ablated by peripheral administration of monosodium glutamate (MSG), whereas AgRP neurons inside the BBB and most proopiomelanocortin (POMC) neurons were spared. MSG treatment induced proliferation of tanycytes, the putative hypothalamic neural progenitor cells, but the newly proliferated tanycytes did not become neurons. Intriguingly, AgRPOBBB neuronal number increased within a week after MSG treatment, and newly emerging AgRP neurons were derived from post-mitotic cells, including some from the Pomc-expressing cell lineage. Our study reveals that the lack of protection by the BBB renders AgRPOBBB vulnerable to lesioning by circulating toxins but that the rapid re-emergence of AgRPOBBB is part of a reparative process to maintain energy balance.
Collapse
Affiliation(s)
- Ernie Yulyaningsih
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ivan A Rudenko
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Martin Valdearcos
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Emma Dahlén
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eirini Vagena
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alvin Chan
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arturo Alvarez-Buylla
- Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christian Vaisse
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Suneil K Koliwad
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Allison W Xu
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
270
|
Erickson MA, Banks WA. Neuroimmune Axes of the Blood-Brain Barriers and Blood-Brain Interfaces: Bases for Physiological Regulation, Disease States, and Pharmacological Interventions. Pharmacol Rev 2018; 70:278-314. [PMID: 29496890 PMCID: PMC5833009 DOI: 10.1124/pr.117.014647] [Citation(s) in RCA: 216] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Central nervous system (CNS) barriers predominantly mediate the immune-privileged status of the brain, and are also important regulators of neuroimmune communication. It is increasingly appreciated that communication between the brain and immune system contributes to physiologic processes, adaptive responses, and disease states. In this review, we discuss the highly specialized features of brain barriers that regulate neuroimmune communication in health and disease. In section I, we discuss the concept of immune privilege, provide working definitions of brain barriers, and outline the historical work that contributed to the understanding of CNS barrier functions. In section II, we discuss the unique anatomic, cellular, and molecular characteristics of the vascular blood-brain barrier (BBB), blood-cerebrospinal fluid barrier, and tanycytic barriers that confer their functions as neuroimmune interfaces. In section III, we consider BBB-mediated neuroimmune functions and interactions categorized as five neuroimmune axes: disruption, responses to immune stimuli, uptake and transport of immunoactive substances, immune cell trafficking, and secretions of immunoactive substances. In section IV, we discuss neuroimmune functions of CNS barriers in physiologic and disease states, as well as pharmacological interventions for CNS diseases. Throughout this review, we highlight many recent advances that have contributed to the modern understanding of CNS barriers and their interface functions.
Collapse
Affiliation(s)
- Michelle A Erickson
- Geriatric Research and Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington; and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - William A Banks
- Geriatric Research and Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington; and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
271
|
McEwen BS. Redefining neuroendocrinology: Epigenetics of brain-body communication over the life course. Front Neuroendocrinol 2018; 49:8-30. [PMID: 29132949 DOI: 10.1016/j.yfrne.2017.11.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 11/01/2017] [Accepted: 11/04/2017] [Indexed: 12/15/2022]
Abstract
The brain is the central organ of stress and adaptation to stress that perceives and determines what is threatening, as well as the behavioral and physiological responses to the stressor, and it does so somewhat differently in males and females. The expression of steroid hormone receptors throughout the brain has broadened the definition of 'neuroendocrinology' to include the reciprocal communication between the entire brain and body via hormonal and neural pathways. Mediated in part via systemic hormonal influences, the adult and developing brain possess remarkable structural and functional plasticity in response to stress, including neuronal replacement, dendritic remodeling, and synapse turnover. This article is both an account of an emerging field elucidating brain-body interactions at multiple levels, from molecules to social organization, as well as a personal account of my laboratory's role and, most importantly, the roles of trainees and colleagues, along with my involvement in interdisciplinary groups working on this topic.
Collapse
Affiliation(s)
- Bruce S McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Ave, New York, NY 10065, USA. http://www.rockefeller.edu/labheads/mcewen/mcewen-lab.php
| |
Collapse
|
272
|
Banks WA, Farr SA, Salameh TS, Niehoff ML, Rhea EM, Morley JE, Hanson AJ, Hansen KM, Craft S. Triglycerides cross the blood-brain barrier and induce central leptin and insulin receptor resistance. Int J Obes (Lond) 2018; 42:391-397. [PMID: 28990588 PMCID: PMC5880581 DOI: 10.1038/ijo.2017.231] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 08/21/2017] [Accepted: 09/03/2017] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Resistance at the brain receptors for leptin and insulin has been associated with increased feeding, obesity and cognitive impairments. The causal agent for central resistance is unknown but could be derived from the blood. Here we postulate whether hypertriglyceridemia, the major dyslipidemia of the metabolic syndrome, could underlie central leptin and insulin resistance. DESIGN We used radioactively labeled triglycerides to measure blood-brain barrier (BBB) penetration, western blots to measure receptor activation, and feeding and cognitive tests to assess behavioral endpoints. RESULTS Human CSF was determined to contain triglycerides, a finding previously unclear. The radioactive triglyceride triolein readily crossed the BBB and centrally administered triolein and peripherally administered lipids induced in vivo leptin and/or insulin resistance at hypothalamic receptors. Central triolein blocked the satiety effect of centrally administered leptin. Decreasing serum triglycerides with gemfibrozil improved both learning and memory inversely proportionate to triglyceride levels. CONCLUSIONS Triglycerides cross the blood-brain barrier rapidly, are found in human cerebrospinal fluid, and induce central leptin and insulin receptor resistance, decreasing satiety and cognition.
Collapse
Affiliation(s)
- W A Banks
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - S A Farr
- Research and Development, John Cochran Veterans Affairs Medical Center, St Louis, MO, USA
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, MO, USA
| | - T S Salameh
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - M L Niehoff
- Research and Development, John Cochran Veterans Affairs Medical Center, St Louis, MO, USA
| | - E M Rhea
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - J E Morley
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, MO, USA
| | - A J Hanson
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - K M Hansen
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - S Craft
- Sticht Center on Aging, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
273
|
Pratchayasakul W, Thongnak LO, Chattipakorn K, Lungaphin A, Pongchaidecha A, Satjaritanun P, Jaiwongkam T, Kerdphoo S, Chattipakorn SC. Atorvastatin and insulin equally mitigate brain pathology in diabetic rats. Toxicol Appl Pharmacol 2018; 342:79-85. [PMID: 29391240 DOI: 10.1016/j.taap.2018.01.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 01/15/2018] [Accepted: 01/28/2018] [Indexed: 01/08/2023]
Abstract
Although insulin and atorvastatin have been shown to exert glycemic control and could improve brain function, the effects of atorvastatin or insulin as well as the combination of atorvastatin plus insulin on brain pathology in diabetes mellitus type 1 (T1DM) are unclear. Therefore, this study investigated the effect of atorvastatin, insulin or combined drugs on brain pathology in streptozotocin-induced diabetic rats. Thirty-six male rats were divided into two groups, a control group (n = 12) and a diabetic or experimental group (n = 24). Diabetic rats were further divided into four groups (n = 6/group) and the groups received either a vehicle (normal saline), atorvastatin (10 mg/kg/day), insulin (4 U/day) or a combination of the drugs for 4 weeks. The control group rats were divided into two groups (n = 6/group) to receive either just the vehicle or atorvastatin for 4 weeks. We found that streptozotocin-induced diabetic rats developed hyperglycemia, showing evidence of increased brain oxidative stress, impaired brain mitochondrial function, increased brain apoptosis, increased tau protein expression, increased phosphorylation of tau protein expression and amyloid beta levels, and decreased dendritic spine density. Although atorvastatin and insulin therapies led to an equal reduction in plasma glucose level in these diabetic rats, the combined drug therapy showed the greatest efficacy in decreasing plasma glucose level. Interestingly, atorvastatin, insulin and the combined drugs equally mitigated brain pathology. Our findings indicate that the combined drug therapy showed the greatest efficacy in improving metabolic parameters. However, atorvastatin, insulin and the combined drug therapy shared a similar efficacy in preventing brain damage in T1DM rats.
Collapse
Affiliation(s)
- Wasana Pratchayasakul
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - La-Ongdao Thongnak
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kenneth Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Anusorn Lungaphin
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Anchalee Pongchaidecha
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pattarapong Satjaritanun
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thidarat Jaiwongkam
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasiwan Kerdphoo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
274
|
The Biased G-Protein-Coupled Receptor Agonism Bridges the Gap between the Insulin Receptor and the Metabolic Syndrome. Int J Mol Sci 2018; 19:ijms19020575. [PMID: 29462993 PMCID: PMC5855797 DOI: 10.3390/ijms19020575] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 02/11/2018] [Accepted: 02/15/2018] [Indexed: 12/14/2022] Open
Abstract
Insulin signaling, as mediated through the insulin receptor (IR), plays a critical role in metabolism. Aberrations in this signaling cascade lead to several pathologies, the majority of which are classified under the umbrella term "metabolic syndrome". Although many of these pathologies are associated with insulin resistance, the exact mechanisms are not well understood. One area of current interest is the possibility of G-protein-coupled receptors (GPCRs) influencing or regulating IR signaling. This concept is particularly significant, because GPCRs have been shown to participate in cross-talk with the IR. More importantly, GPCR signaling has also been shown to preferentially regulate specific downstream signaling targets through GPCR agonist bias. A novel study recently demonstrated that this GPCR-biased agonism influences the activity of the IR without the presence of insulin. Although GPCR-IR cross-talk has previously been established, the notion that GPCRs can regulate the activation of the IR is particularly significant in relation to metabolic syndrome and other pathologies that develop as a result of alterations in IR signaling. As such, we aim to provide an overview of the physiological and pathophysiological roles of the IR within metabolic syndrome and its related pathologies, including cardiovascular health, gut microflora composition, gastrointestinal tract functioning, polycystic ovarian syndrome, pancreatic cancer, and neurodegenerative disorders. Furthermore, we propose that the GPCR-biased agonism may perhaps mediate some of the downstream signaling effects that further exacerbate these diseases for which the mechanisms are currently not well understood.
Collapse
|
275
|
Abstract
Chronic diseases are defined diseases whose symptoms last for at least six months and tend to worsen over time. In Europe, they cause at least 86% of deaths. In this speculative unifying model I set a new hypothesis for the etiology of the majority of chronic diseases. The main aim is to put order and observe our organism in a systemic way, connecting pathologies we now see as disconnected phenomena, with the conceptual frameworks of complex systems and network medicine. Chronic diseases could be caused by a first unsolved acute infection. In case the pathogen cannot be completely eliminated, it becomes a persistent infectious. After the acute episode, some mild symptoms will occur and probably disappear; the chronic disease will remain latent over time. It will manifest even after years or decades, in the presence of another acute infection, a particular stress, trauma, or another event. The presence of the persistent infectious elicits changes in the immune and systemic regulation, and these processes degenerate over time. They will assume their rules and patterns, being independent from the initial stimulus. The key to understand the dynamics and individuality of chronic diseases is the immune system and its networks. The immune mechanisms that can lead to the persistent response are mainly the switch from the Th1 to the Th2 immunity and the molecular mimicry. The first persistent infectious will also modify the susceptibility to other pathogens, facilitating new infections and new consequent persistent infectious. From the immune point of view, our organism is divided into three compartments: the outer one, which comprehend all the surfaces in contact with the environment, the intermediate one, which comprehend the internal organs and tissues, and the innermost one, comprehending the Central Nervous System and the adluminal compartment of the seminiferous tubule. The immune key-role is played respectively by the mucosa-associated lymphoid tissue, the endothelium, the blood-brain barrier and blood-testis barrier. The chronic diseases follow a progressive scheme, involving the three compartments from the outer to the innermost one. The primer microorganism at the origin of the majority of diseases could be streptococcus, or staphylococcus. Both cause acute in children, with a great variability of responses and symptoms, and both cause molecular mimicry. This model can be tested and proved in more ways, I propose here some of them. It could pave the way to a radical change in our comprehension and therapeutic approaches to chronic diseases.
Collapse
|
276
|
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: 971] [Impact Index Per Article: 138.7] [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
|
277
|
Raben AT, Marshe VS, Chintoh A, Gorbovskaya I, Müller DJ, Hahn MK. The Complex Relationship between Antipsychotic-Induced Weight Gain and Therapeutic Benefits: A Systematic Review and Implications for Treatment. Front Neurosci 2018; 11:741. [PMID: 29403343 PMCID: PMC5786866 DOI: 10.3389/fnins.2017.00741] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 12/20/2017] [Indexed: 12/15/2022] Open
Abstract
Background: Antipsychotic-induced weight gain (AIWG) and other adverse metabolic effects represent serious side effects faced by many patients with psychosis that can lead to numerous comorbidities and which reduce the lifespan. While the pathophysiology of AIWG remains poorly understood, numerous studies have reported a positive association between AIWG and the therapeutic benefit of antipsychotic medications. Objectives: To review the literature to (1) determine if AIWG is consistently associated with therapeutic benefit and (2) investigate which variables may mediate such an association. Data Sources: MEDLINE, Google Scholar, Cochrane Database and PsycINFO databases were searched for articles containing all the following exploded MESH terms: schizophrenia [AND] antipsychotic agents/neuroleptics [AND] (weight gain [OR] lipids [OR] insulin [OR] leptin) [AND] treatment outcome. Results were limited to full-text, English journal articles. Results: Our literature search uncovered 31 independent studies which investigated an AIWG-therapeutic benefit association with a total of 6063 enrolled individuals diagnosed with schizophrenia or another serious mental illness receiving antipsychotic medications. Twenty-two studies found a positive association while, 10 studies found no association and one study reported a negative association. Study variables including medication compliance, sex, ethnicity, or prior antipsychotic exposure did not appear to consistently affect the AIWG-therapeutic benefit relationship. In contrast, there was some evidence that controlling for baseline BMI/psychopathology, duration of treatment and specific agent studied [i.e., olanzapine (OLZ) or clozapine (CLZ)] strengthened the relationship between AIWG and therapeutic benefit. Limitations: There were limitations of the reviewed studies in that many had small sample sizes, and/or were retrospective. The heterogeneity of the studies also made comparisons difficult and publication bias was not controlled for. Conclusions: An AIWG-therapeutic benefit association may exist and is most likely to be observed in OLZ and CLZ-treated patients. The clinical meaningfulness of this association remains unclear and weight gain and other metabolic comorbidities should be identified and treated to the same targets as the general population. Further research should continue to explore the links between therapeutic benefit and metabolic health with emphasis on both pre-clinical work and well-designed prospective clinical trials examining metabolic parameters associated, but also occurring independently to AIWG.
Collapse
Affiliation(s)
- Alex T Raben
- Schizophrenia Program, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Victoria S Marshe
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Araba Chintoh
- Schizophrenia Program, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ilona Gorbovskaya
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Daniel J Müller
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Margaret K Hahn
- Schizophrenia Program, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
278
|
Miranda-Martínez A, Mercado-Gómez OF, Arriaga-Ávila V, Guevara-Guzmán R. Distribution of Adiponectin Receptors 1 and 2 in the Rat Olfactory Bulb and the Effect of Adiponectin Injection on Insulin Receptor Expression. Int J Endocrinol 2017; 2017:4892609. [PMID: 29463982 PMCID: PMC5804105 DOI: 10.1155/2017/4892609] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 11/08/2017] [Accepted: 11/14/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Adiponectin (APN) is an adipocyte-derived hormone that has peripheral beneficial effects. Although its receptors AdipoR1 and AdipoR2 are expressed in the brain, their function in neurons is poorly understood. The aims of this work were to describe the distribution of APN receptors in the olfactory bulb (OB) as well as the possible effects of APN injection on the insulin receptor (InsR) content and Akt kinase. METHOD We performed the double immunofluorescence technique to describe the distribution of AdipoRs and the cellular type they were expressing. mRNA transcript and protein content were assessed by RT-PCR and Western blot, respectively. APN injection was performed to analyze its possible effect on the insulin pathway. RESULTS We found that AdipoRs were localized in all cell layers and in both neurons and astrocytes. We observed the presence of mRNA transcripts and immunoblot analysis confirmed the protein on the intact OB; APN injection in the OB resulted in a slight decrease of the total InsR and Akt phosphorylation and a reduction of phopho-InsR content. CONCLUSIONS These data demonstrated that AdipoRs are expressed in OB regions, and APN injection could act as an insulin pathway modulator in the OB and thus possibly contribute to olfaction physiology.
Collapse
Affiliation(s)
- Alfredo Miranda-Martínez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Del. Coyoacán, 04510 Ciudad de México, Mexico
- Programa de Doctorado en Ciencias Biológicas, Coordinación del Posgrado en Ciencias Biológicas, Edificio B, 1° Piso. Circuito de Posgrados, Ciudad Universitaria, Del. Coyoacán, 04510 Ciudad de México, Mexico
| | - Octavio Fabián Mercado-Gómez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Del. Coyoacán, 04510 Ciudad de México, Mexico
| | - Virginia Arriaga-Ávila
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Del. Coyoacán, 04510 Ciudad de México, Mexico
| | - Rosalinda Guevara-Guzmán
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Del. Coyoacán, 04510 Ciudad de México, Mexico
| |
Collapse
|
279
|
Microbial Regulation of Glucose Metabolism and Insulin Resistance. Genes (Basel) 2017; 9:genes9010010. [PMID: 29286343 PMCID: PMC5793163 DOI: 10.3390/genes9010010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes is a combined disease, resulting from a hyperglycemia and peripheral and hepatic insulin resistance. Recent data suggest that the gut microbiota is involved in diabetes development, altering metabolic processes including glucose and fatty acid metabolism. Thus, type 2 diabetes patients show a microbial dysbiosis, with reduced butyrate-producing bacteria and elevated potential pathogens compared to metabolically healthy individuals. Furthermore, probiotics are a known tool to modulate the microbiota, having a therapeutic potential. Current literature will be discussed to elucidate the complex interaction of gut microbiota, intestinal permeability and inflammation leading to peripheral and hepatic insulin resistance. Therefore, this review aims to generate a deeper understanding of the underlying mechanism of potential microbial strains, which can be used as probiotics.
Collapse
|
280
|
Nilsson M, Gjedde A, Brock B, Gejl M, Rungby J. The effects of incretin hormones on cerebral glucose metabolism in health and disease. Neuropharmacology 2017; 136:243-250. [PMID: 29274367 DOI: 10.1016/j.neuropharm.2017.12.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 12/25/2022]
Abstract
Incretin hormones, notably glucagon-like peptide-1 (GLP-1), are gluco-regulatory hormones with pleiotropic effects also in the central nervous system. Apart from a local production of GLP-1, systemic administration of the hormone has been shown to influence a number of cerebral pathologies, including neuroinflammation. Given the brains massive dependence on glucose as its major fuel, we here review the mechanistics of cerebral glucose transport and metabolism, focusing on the deleterious effects of both hypo- and hyperglycaemia. GLP-1, when administered as long-acting analogues or intravenously, appears to decrease transport of glucose in normoglycaemic conditions, without affecting the total cerebral glucose content. During hypoglycaemia this effect seems abated, whereas during hyperglycaemia GLP-1 regulates cerebral glucose metabolism towards stable levels resembling normoglycaemia. In Alzheimer's disease, a 6-month intervention with GLP-1 maintained cerebral glucose levels at baseline levels, contrasting the decline otherwise seen in Alzheimer's. Kinetic studies suggest blood-brain barrier (BBB) glucose transport as the key player in GLP-1 mediated effects on cerebral glucose metabolism. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Malin Nilsson
- Department of Endocrinology, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Albert Gjedde
- Department of Neuroscience, Panum Institute, University of Copenhagen, Copenhagen, Denmark; Departments of Clinical Research, and Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Michael Gejl
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Jørgen Rungby
- Department of Endocrinology, Bispebjerg University Hospital, Copenhagen, Denmark.
| |
Collapse
|
281
|
Insulin-mediated synaptic plasticity in the CNS: Anatomical, functional and temporal contexts. Neuropharmacology 2017; 136:182-191. [PMID: 29217283 PMCID: PMC5988909 DOI: 10.1016/j.neuropharm.2017.12.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 12/17/2022]
Abstract
For decades the brain was erroneously considered an insulin insensitive organ. Although gaps in our knowledge base remain, conceptual frameworks are starting to emerge to provide insight into the mechanisms through which insulin facilitates critical brain functions like metabolism, cognition, and motivated behaviors. These diverse physiological and behavioral activities highlight the region-specific activities of insulin in the CNS; that is, there is an anatomical context to the activities of insulin in the CNS. Similarly, there is also a temporal context to the activities of insulin in the CNS. Indeed, brain insulin receptor activity can be conceptualized as a continuum in which insulin promotes neuroplasticity from development into adulthood where it is an integral part of healthy brain function. Unfortunately, brain insulin resistance likely contributes to neuroplasticity deficits in obesity and type 2 diabetes mellitus (T2DM). This neuroplasticity continuum can be conceptualized by the mechanisms through which insulin promotes cognitive function through its actions in brain regions like the hippocampus, as well as the ability of insulin to modulate motivated behaviors through actions in brain regions like the nucleus accumbens and the ventral tegmental area. Thus, the goals of this review are to highlight these anatomical, temporal, and functional contexts of insulin activity in these brain regions, and to identify potentially critical time points along this continuum where the transition from enhancement of neuroplasticity to impairment may take place.
Collapse
|
282
|
Ladyman SR, Grattan DR. Region-Specific Suppression of Hypothalamic Responses to Insulin To Adapt to Elevated Maternal Insulin Secretion During Pregnancy. Endocrinology 2017; 158:4257-4269. [PMID: 29029017 DOI: 10.1210/en.2017-00600] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/22/2017] [Indexed: 01/22/2023]
Abstract
As part of the adaptation of maternal glucose regulation during pregnancy to ensure glucose provision to the fetus, maternal insulin concentrations become elevated. However, increased central actions of insulin, such as suppression of appetite, would be maladaptive during pregnancy. We hypothesized that central nervous system targets of insulin become less responsive during pregnancy to prevent overstimulation by the increased circulating insulin concentrations. To test this hypothesis, we have measured insulin-induced phosphorylation of Akt (pAkt) in specific hypothalamic nuclei as an index of hypothalamic insulin responsiveness. Despite higher endogenous insulin concentrations following feeding, arcuate nucleus pAkt levels were significantly lower in the pregnant group compared with the nonpregnant group. In response to an intracerebroventricular injection of insulin, insulin-induced pAkt was significantly reduced in the arcuate nucleus and ventromedial nucleus of pregnant rats compared with nonpregnant rats. Similar levels of insulin receptor β and PTEN, a negative regulator of the phosphoinositide 3-kinase/Akt pathway, were detected in hypothalamic areas of nonpregnant and pregnant rats. In the ventromedial nucleus, however, levels of phosphorylated PTEN were significantly lower in pregnancy, suggesting that reduced inactivation of PTEN may contribute to the attenuated insulin signaling in this area during pregnancy. In conclusion, these results demonstrate region-specific changes in responsiveness to insulin in the hypothalamus during pregnancy that may represent an adaptive response to minimize the impact of elevated circulating insulin on the maternal brain.
Collapse
Affiliation(s)
- Sharon R Ladyman
- Centre for Neuroendocrinology, University of Otago, New Zealand
- Department of Anatomy, University of Otago, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| | - David R Grattan
- Centre for Neuroendocrinology, University of Otago, New Zealand
- Department of Anatomy, University of Otago, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| |
Collapse
|
283
|
Markowicz-Piasecka M, Sikora J, Szydłowska A, Skupień A, Mikiciuk-Olasik E, Huttunen KM. Metformin - a Future Therapy for Neurodegenerative Diseases : Theme: Drug Discovery, Development and Delivery in Alzheimer's Disease Guest Editor: Davide Brambilla. Pharm Res 2017; 34:2614-2627. [PMID: 28589443 PMCID: PMC5736777 DOI: 10.1007/s11095-017-2199-y] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 05/30/2017] [Indexed: 12/13/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a complex, chronic and progressive metabolic disease, which is characterized by relative insulin deficiency, insulin resistance, and high glucose levels in blood. Esteemed published articles and epidemiological data exhibit an increased risk of developing Alzheimer's disease (AD) in diabetic pateints. Metformin is the most frequently used oral anti-diabetic drug, which apart from hypoglycaemic activity, improves serum lipid profiles, positively influences the process of haemostasis, and possesses anti-inflammatory properties. Recently, scientists have put their efforts in establishing metformin's role in the treatment of neurodegenerative diseases, such as AD, amnestic mild cognitive impairment and Parkinson's disease. Results of several clinical studies confirm that long term use of metformin in diabetic patients contributes to better cognitive function, compared to participants using other anti-diabetic drugs. The exact mechanism of metformin's advantageous activity in AD is not fully understood, but scientists claim that activation of AMPK-dependent pathways in human neural stem cells might be responsible for the neuroprotective activity of metformin. Metformin was also found to markedly decease Beta-secretase 1 (BACE1) protein expression and activity in cell culture models and in vivo, thereby reducing BACE1 cleavage products and the production of Aβ (β-amyloid). Furthermore, there is also some evidence that metformin decreases the activity of acetylcholinesterase (AChE), which is responsible for the degradation of acetylcholine (Ach), a neurotransmitter involved in the process of learning and memory. In regard to the beneficial effects of metformin, its anti-inflammatory and anti-oxidative properties cannot be omitted. Numerous in vitro and in vivo studies have confirmed that metformin ameliorates oxidative damage.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry Drug Analysis and Radiopharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Joanna Sikora
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry Drug Analysis and Radiopharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Aleksandra Szydłowska
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry Drug Analysis and Radiopharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Agata Skupień
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry Drug Analysis and Radiopharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Elżbieta Mikiciuk-Olasik
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul., Muszyńskiego 1, 90-151, Lodz, Poland
| | - Kristiina M. Huttunen
- School Of Pharmacy, Faculty of Health Sciences, University of Eastern Finland,, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland
| |
Collapse
|
284
|
The Role of Glucagon-Like Peptide 1 (GLP1) in Type 3 Diabetes: GLP-1 Controls Insulin Resistance, Neuroinflammation and Neurogenesis in the Brain. Int J Mol Sci 2017; 18:ijms18112493. [PMID: 29165354 PMCID: PMC5713459 DOI: 10.3390/ijms18112493] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease (AD), characterized by the aggregation of amyloid-β (Aβ) protein and neuroinflammation, is the most common neurodegenerative disease globally. Previous studies have reported that some AD patients show impaired glucose utilization in brain, leading to cognitive decline. Recently, diabetes-induced dementia has been called "type 3 diabetes", based on features in common with those of type 2 diabetes and the progression of AD. Impaired glucose uptake and insulin resistance in the brain are important issues in type 3 diabetes, because these problems ultimately aggravate memory dysfunction in the brain. Glucagon-like peptide 1 (GLP-1) has been known to act as a critical controller of the glucose metabolism. Several studies have demonstrated that GLP-1 alleviates learning and memory dysfunction by enhancing the regulation of glucose in the AD brain. However, the specific actions of GLP-1 in the AD brain are not fully understood. Here, we review evidences related to the role of GLP-1 in type 3 diabetes.
Collapse
|
285
|
Jia JJ, Zeng XS, Song XQ, Zhang PP, Chen L. Diabetes Mellitus and Alzheimer's Disease: The Protection of Epigallocatechin-3-gallate in Streptozotocin Injection-Induced Models. Front Pharmacol 2017; 8:834. [PMID: 29209211 PMCID: PMC5702501 DOI: 10.3389/fphar.2017.00834] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 11/01/2017] [Indexed: 12/27/2022] Open
Abstract
Diabetes mellitus is considered as a risk factor of Alzheimer's disease (AD), the front runner of neurodegenerative disorders. Streptozotocin (STZ) is a toxin for pancreatic β-cell, which can construct a model of insulin deficient diabetes through intraperitoneal or intravenous injection. A model generated by intracerebroventricular STZ (icv-STZ) also shows numerous aspects of sporadic AD. The protective roles of tea polyphenols epigallocatechin-3-gallate (EGCG) on both two diseases were researched by some scientists. This review highlights the link between diabetes and AD and recent studies on STZ injection-induced models, and also discusses the protection of EGCG to clarify its treatment in STZ-induced diabetes and AD.
Collapse
Affiliation(s)
- Jin-Jing Jia
- College of Life Sciences, Xinyang Normal University, Xinyang, China
- Institute for Conservation and Utilization of Agro-bioresources in Dabie Mountains, Xinyang Normal University, Xinyang, China
- Henan Key Laboratory of Tea Biology, Xinyang Normal University, Xinyang, China
| | - Xian-Si Zeng
- College of Life Sciences, Xinyang Normal University, Xinyang, China
- Institute for Conservation and Utilization of Agro-bioresources in Dabie Mountains, Xinyang Normal University, Xinyang, China
- Henan Key Laboratory of Tea Biology, Xinyang Normal University, Xinyang, China
| | - Xin-Qiang Song
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Peng-Peng Zhang
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Lei Chen
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| |
Collapse
|
286
|
Jia JJ, Zeng XS, Song XQ, Zhang PP, Chen L. Diabetes Mellitus and Alzheimer's Disease: The Protection of Epigallocatechin-3-gallate in Streptozotocin Injection-Induced Models. Front Pharmacol 2017; 8:834. [PMID: 29209211 DOI: 10.3389/fphar.2017.00834if:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 11/01/2017] [Indexed: 11/16/2024] Open
Abstract
Diabetes mellitus is considered as a risk factor of Alzheimer's disease (AD), the front runner of neurodegenerative disorders. Streptozotocin (STZ) is a toxin for pancreatic β-cell, which can construct a model of insulin deficient diabetes through intraperitoneal or intravenous injection. A model generated by intracerebroventricular STZ (icv-STZ) also shows numerous aspects of sporadic AD. The protective roles of tea polyphenols epigallocatechin-3-gallate (EGCG) on both two diseases were researched by some scientists. This review highlights the link between diabetes and AD and recent studies on STZ injection-induced models, and also discusses the protection of EGCG to clarify its treatment in STZ-induced diabetes and AD.
Collapse
Affiliation(s)
- Jin-Jing Jia
- College of Life Sciences, Xinyang Normal University, Xinyang, China
- Institute for Conservation and Utilization of Agro-bioresources in Dabie Mountains, Xinyang Normal University, Xinyang, China
- Henan Key Laboratory of Tea Biology, Xinyang Normal University, Xinyang, China
| | - Xian-Si Zeng
- College of Life Sciences, Xinyang Normal University, Xinyang, China
- Institute for Conservation and Utilization of Agro-bioresources in Dabie Mountains, Xinyang Normal University, Xinyang, China
- Henan Key Laboratory of Tea Biology, Xinyang Normal University, Xinyang, China
| | - Xin-Qiang Song
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Peng-Peng Zhang
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Lei Chen
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| |
Collapse
|
287
|
Tsou YH, Zhang XQ, Zhu H, Syed S, Xu X. Drug Delivery to the Brain across the Blood-Brain Barrier Using Nanomaterials. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1701921. [PMID: 29045030 DOI: 10.1002/smll.201701921] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/09/2017] [Indexed: 05/24/2023]
Abstract
A major obstacle facing brain diseases such as Alzheimer's disease, multiple sclerosis, brain tumors, and strokes is the blood-brain barrier (BBB). The BBB prevents the passage of certain molecules and pathogens from the circulatory system into the brain. Therefore, it is nearly impossible for therapeutic drugs to target the diseased cells without the assistance of carriers. Nanotechnology is an area of growing public interest; nanocarriers, such as polymer-based, lipid-based, and inorganic-based nanoparticles can be engineered in different sizes, shapes, and surface charges, and they can be modified with functional groups to enhance their penetration and targeting capabilities. Hence, understanding the interaction between nanomaterials and the BBB is crucial. In this Review, the components and properties of the BBB are revisited and the types of nanocarriers that are most commonly used for brain drug delivery are discussed. The properties of the nanocarriers and the factors that affect drug delivery across the BBB are elaborated upon in this review. Additionally, the most recent developments of nanoformulations and nonconventional drug delivery strategies are highlighted. Finally, challenges and considerations for the development of brain targeting nanomedicines are discussed. The overall objective is to broaden the understanding of the design and to develop nanomedicines for the treatment of brain diseases.
Collapse
Affiliation(s)
- Yung-Hao Tsou
- Department of Chemical Biological, and Pharmaceutical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Xue-Qing Zhang
- Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai, 200240, China
| | - He Zhu
- Department of Chemical Biological, and Pharmaceutical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Sahla Syed
- Department of Chemical Biological, and Pharmaceutical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Xiaoyang Xu
- Department of Chemical Biological, and Pharmaceutical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| |
Collapse
|
288
|
Geijselaers SLC, Sep SJS, Claessens D, Schram MT, van Boxtel MPJ, Henry RMA, Verhey FRJ, Kroon AA, Dagnelie PC, Schalkwijk CG, van der Kallen CJH, Biessels GJ, Stehouwer CDA. The Role of Hyperglycemia, Insulin Resistance, and Blood Pressure in Diabetes-Associated Differences in Cognitive Performance-The Maastricht Study. Diabetes Care 2017; 40:1537-1547. [PMID: 28842522 DOI: 10.2337/dc17-0330] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 07/23/2017] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To study to what extent differences in cognitive performance between individuals with different glucose metabolism status are potentially attributable to hyperglycemia, insulin resistance, and blood pressure-related variables. RESEARCH DESIGN AND METHODS We used cross-sectional data from 2,531 participants from the Maastricht Study (mean age ± SD, 60 ± 8 years; 52% men; n = 666 with type 2 diabetes), all of whom completed a neuropsychological test battery. Hyperglycemia was assessed by a composite index of fasting glucose, postload glucose, glycated hemoglobin (HbA1c), and tissue advanced glycation end products; insulin resistance by the HOMA of insulin resistance index; and blood pressure-related variables included 24-h ambulatory pressures, their weighted SDs, and the use of antihypertensive medication. Linear regression analyses were used to estimate mediating effects. RESULTS After adjustment for age, sex, and education, individuals with type 2 diabetes, compared with those with normal glucose metabolism, performed worse in all cognitive domains (mean differences in composite z scores for memory -0.087, processing speed -0.196, executive function and attention -0.182; P values <0.032), whereas individuals with prediabetes did not. Diabetes-associated differences in processing speed and executive function and attention were largely explained by hyperglycemia (mediating effect 79.6% [bootstrapped 95% CI 36.6; 123.4] and 50.3% [0.6; 101.2], respectively) and, for processing speed, to a lesser extent by blood pressure-related variables (17.7% [5.6; 30.1]), but not by insulin resistance. None of the factors explained the differences in memory function. CONCLUSIONS Our cross-sectional data suggest that early glycemic and blood pressure control, perhaps even in the prediabetic stage, may be promising therapeutic targets for the prevention of diabetes-associated decrements in cognitive performance.
Collapse
Affiliation(s)
- Stefan L C Geijselaers
- Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, the Netherlands.,School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands.,Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Simone J S Sep
- Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, the Netherlands.,School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Danny Claessens
- Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Miranda T Schram
- Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, the Netherlands.,School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Martin P J van Boxtel
- Department of Psychiatry and Neuropsychology and School for Mental Health and Neuroscience, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Ronald M A Henry
- Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, the Netherlands.,School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Frans R J Verhey
- Department of Psychiatry and Neuropsychology and School for Mental Health and Neuroscience, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Abraham A Kroon
- Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, the Netherlands.,School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Pieter C Dagnelie
- School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands.,School for Public Health and Primary Care (CAPHRI), Maastricht University, Maastricht, the Netherlands.,Department of Epidemiology, Maastricht University, Maastricht, the Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, the Netherlands.,School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Carla J H van der Kallen
- Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, the Netherlands.,School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Geert Jan Biessels
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, the Netherlands .,School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
289
|
Neth BJ, Craft S. Insulin Resistance and Alzheimer's Disease: Bioenergetic Linkages. Front Aging Neurosci 2017; 9:345. [PMID: 29163128 PMCID: PMC5671587 DOI: 10.3389/fnagi.2017.00345] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/13/2017] [Indexed: 12/14/2022] Open
Abstract
Metabolic dysfunction is a well-established feature of Alzheimer's disease (AD), evidenced by brain glucose hypometabolism that can be observed potentially decades prior to the development of AD symptoms. Furthermore, there is mounting support for an association between metabolic disease and the development of AD and related dementias. Individuals with insulin resistance, type 2 diabetes mellitus (T2D), hyperlipidemia, obesity, or other metabolic disease may have increased risk for the development of AD and similar conditions, such as vascular dementia. This association may in part be due to the systemic mitochondrial dysfunction that is common to these pathologies. Accumulating evidence suggests that mitochondrial dysfunction is a significant feature of AD and may play a fundamental role in its pathogenesis. In fact, aging itself presents a unique challenge due to inherent mitochondrial dysfunction and prevalence of chronic metabolic disease. Despite the progress made in understanding the pathogenesis of AD and in the development of potential therapies, at present we remain without a disease-modifying treatment. In this review, we will discuss insulin resistance as a contributing factor to the pathogenesis of AD, as well as the metabolic and bioenergetic disruptions linking insulin resistance and AD. We will also focus on potential neuroimaging tools for the study of the metabolic dysfunction commonly seen in AD with hopes of developing therapeutic and preventative targets.
Collapse
Affiliation(s)
- Bryan J Neth
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
290
|
Beirami E, Oryan S, Seyedhosseini Tamijani SM, Ahmadiani A, Dargahi L. Intranasal insulin treatment restores cognitive deficits and insulin signaling impairment induced by repeated methamphetamine exposure. J Cell Biochem 2017; 119:2345-2355. [PMID: 28884876 DOI: 10.1002/jcb.26398] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/30/2017] [Indexed: 01/03/2023]
Abstract
Long-term use of methamphetamine (MA) causes a broad range of cognitive deficits. Recently, it has been reported insulin signaling and mitochondrial biogenesis are involved in cognitive processes. This study aimed to examine whether MA induces cognitive deficits concomitant with insulin signaling impairment and mitochondrial dysfunctions and also intranasal (IN) insulin treatment can reverse cognitive deficits caused by MA. Rats were repeatedly treated with increasing doses of MA (1-10 mg/kg) twice a day for 10 days, and their cognitive functions were assessed using Y-maze, novel object recognition and passive avoidance tasks. The expression of components involved in insulin signaling (IR/IRS2/PI3K/Akt/GSK3β) and mitochondrial biogenesis (PGC-1α, NRF1, and TFAM) was measured in the hippocampus. Therapeutic effects of IN insulin delivery (0.5- IU/day, for 7 days after MA discontinuation) were also investigated in MA-treated animals. Our results showed that repeated MA exposure induced cognitive deficits, and led to insulin signaling impairment and mitochondrial dysfunction. Interestingly, IN insulin treatment reduced MA-induced cognitive impairments possibly through activating insulin signaling, particularly PI3K/Akt/GSK3β pathway, and mitochondrial biogenesis. Thus, insulin and insulin signaling pathway can be considered as useful targets for the treatment of abnormalities associated with MA abuse.
Collapse
Affiliation(s)
- Elmira Beirami
- Faculty of Biological Sciences, Department of Animal Biology, Kharazmi University, Tehran, Iran
| | - Shahrbanoo Oryan
- Faculty of Biological Sciences, Department of Animal Biology, Kharazmi University, Tehran, Iran
| | | | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
291
|
Pousti F, Ahmadi R, Mirahmadi F, Hosseinmardi N, Rohampour K. Adiponectin modulates synaptic plasticity in hippocampal dentate gyrus. Neurosci Lett 2017; 662:227-232. [PMID: 29079430 DOI: 10.1016/j.neulet.2017.10.042] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 10/17/2017] [Accepted: 10/20/2017] [Indexed: 12/15/2022]
Abstract
Recent studies have suggested the involvement of some metabolic hormones in memory formation and synaptic plasticity. Insulin dysfunction is known as an essential process in the pathogenesis of sporadic Alzheimer's disease (AD). In this study we examined whether adiponectin (ADN), as an insulin-sensitizing adipokine, could affect hippocampal synaptic plasticity. Field potential recordings were performed on intracerebroventricular (icv) cannulated urethane anesthetized rats. After baseline recording from dentate gyrus (DG) and 10min prior to high/low frequency stimulation (HFS/LFS), 10μl icv ADN (600nm) were injected. The slope of field excitatory postsynaptic potentials (fEPSP) and the amplitude of population spikes (PS) were recorded in response to perforanth path (PP) stimulation. Paired pulse stimuli and ADN injection without any stimulation protocols were also evaluated. Application of ADN before HFS increased PS amplitude recorded in DG significantly (P≤0.05) in comparison to HFS only group. ADN suppressed the potency of LFS to induce long-term depression (LTD), causing a significant difference between fEPSP slope (P≤0.05) and PS amplitude (P≤0.01) between ADN+LFS and ADN group. Paired pulse stimuli applied at 20ms intervals showed more paired pulse facilitation (PPF), when applied after ADN (P≤0.05). ADN induced a chemical long-term potentiation (LTP) in which fEPSP slope and PS amplitude increased significantly (P≤0.01 and P≤0.05, respectively). It is concluded that ADN is able to potentiate the HFS-induced LTP and suppress LFS-induced LTD. ADN caused a chemical LTP, when applied without any tetanic protocol. ADN may enhance the presynaptic release probability.
Collapse
Affiliation(s)
- Farideh Pousti
- Department of Biology, Islamic Azad University, Qom Branch, Qom, Iran
| | - Ramesh Ahmadi
- Department of Biology, Islamic Azad University, Qom Branch, Qom, Iran
| | - Fatemeh Mirahmadi
- Department of Biology, Islamic Azad University, Qom Branch, Qom, Iran
| | - Narges Hosseinmardi
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kambiz Rohampour
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| |
Collapse
|
292
|
Belfiore A, Malaguarnera R, Vella V, Lawrence MC, Sciacca L, Frasca F, Morrione A, Vigneri R. Insulin Receptor Isoforms in Physiology and Disease: An Updated View. Endocr Rev 2017; 38:379-431. [PMID: 28973479 PMCID: PMC5629070 DOI: 10.1210/er.2017-00073] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/13/2017] [Indexed: 02/08/2023]
Abstract
The insulin receptor (IR) gene undergoes differential splicing that generates two IR isoforms, IR-A and IR-B. The physiological roles of IR isoforms are incompletely understood and appear to be determined by their different binding affinities for insulin-like growth factors (IGFs), particularly for IGF-2. Predominant roles of IR-A in prenatal growth and development and of IR-B in metabolic regulation are well established. However, emerging evidence indicates that the differential expression of IR isoforms may also help explain the diversification of insulin and IGF signaling and actions in various organs and tissues by involving not only different ligand-binding affinities but also different membrane partitioning and trafficking and possibly different abilities to interact with a variety of molecular partners. Of note, dysregulation of the IR-A/IR-B ratio is associated with insulin resistance, aging, and increased proliferative activity of normal and neoplastic tissues and appears to sustain detrimental effects. This review discusses novel information that has generated remarkable progress in our understanding of the physiology of IR isoforms and their role in disease. We also focus on novel IR ligands and modulators that should now be considered as an important strategy for better and safer treatment of diabetes and cancer and possibly other IR-related diseases.
Collapse
Affiliation(s)
- Antonino Belfiore
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Roberta Malaguarnera
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Veronica Vella
- School of Human and Social Sciences, University Kore of Enna, via della Cooperazione, 94100 Enna, Italy
| | - Michael C. Lawrence
- Structural Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Laura Sciacca
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Francesco Frasca
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Andrea Morrione
- Department of Urology and Biology of Prostate Cancer Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Riccardo Vigneri
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| |
Collapse
|
293
|
Nash AI. Crosstalk between insulin and dopamine signaling: A basis for the metabolic effects of antipsychotic drugs. J Chem Neuroanat 2017; 83-84:59-68. [DOI: 10.1016/j.jchemneu.2016.07.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/14/2016] [Accepted: 07/27/2016] [Indexed: 12/21/2022]
|
294
|
Engin AB, Engin ED, Karakus R, Aral A, Gulbahar O, Engin A. N-Methyl-D aspartate receptor-mediated effect on glucose transporter-3 levels of high glucose exposed-SH-SY5Y dopaminergic neurons. Food Chem Toxicol 2017; 109:465-471. [PMID: 28951307 DOI: 10.1016/j.fct.2017.09.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/16/2017] [Accepted: 09/20/2017] [Indexed: 11/29/2022]
Abstract
High glucose and insulin lead to neuronal insulin resistance. Glucose transport into the neurons is achieved by regulatory induction of surface glucose transporter-3 (GLUT3) instead of the insulin. N-methyl-D aspartate (NMDA) receptor activity increases GLUT3 expression. This study explored whether an endogenous NMDA receptor antagonist, kynurenic acid (KynA) affects the neuronal cell viability at high glucose concentrations. SH-SY5Y neuroblastoma cells were exposed to 150-250 mg/dL glucose and 40 μU/mL insulin. In KynA and N-nitro-l-arginine methyl ester (L-NAME) supplemented cultures, oxidative stress, mitochondrial metabolic activity (MTT), nitric oxide as nitrite+nitrate (NOx) and GLUT3 were determined at the end of 24 and 48-h incubation periods. Viable cells were counted by trypan blue dye. High glucose-exposed SH-SY5Y cells showed two-times more GLUT3 expression at second 24-h period. While GLUT3-stimulated glucose transport and oxidative stress was increased, total mitochondrial metabolic activity was significantly reduced. Insulin supplementation to high glucose decreased NOx synthesis and GLUT3 levels, in contrast oxidative stress increased three-fold. KynA significantly reduced oxidative stress, and increased MTT by regulating NOx production and GLUT3 expression. KynA is a noteworthy compound, as an endogenous, specific NMDA receptor antagonist; it significantly reduces oxidative stress, while increasing cell viability at high glucose and insulin concentrations.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Gazi University, Faculty of Pharmacy, Department of Toxicology, Hipodrom, Ankara, Turkey.
| | - Evren Doruk Engin
- Ankara University, Biotechnology Institute, Tandogan, Ankara, Turkey
| | - Resul Karakus
- Gazi University, Faculty of Medicine, Department of Immunology, Besevler, Ankara, Turkey
| | - Arzu Aral
- Gazi University, Faculty of Medicine, Department of Immunology, Besevler, Ankara, Turkey
| | - Ozlem Gulbahar
- Gazi University, Faculty of Medicine, Department of Biochemistry, Besevler, Ankara, Turkey
| | - Atilla Engin
- Gazi University, Faculty of Medicine, Department of General Surgery, Besevler, Ankara, Turkey
| |
Collapse
|
295
|
Endothelial insulin receptors differentially control insulin signaling kinetics in peripheral tissues and brain of mice. Proc Natl Acad Sci U S A 2017; 114:E8478-E8487. [PMID: 28923931 DOI: 10.1073/pnas.1710625114] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Insulin receptors (IRs) on endothelial cells may have a role in the regulation of transport of circulating insulin to its target tissues; however, how this impacts on insulin action in vivo is unclear. Using mice with endothelial-specific inactivation of the IR gene (EndoIRKO), we find that in response to systemic insulin stimulation, loss of endothelial IRs caused delayed onset of insulin signaling in skeletal muscle, brown fat, hypothalamus, hippocampus, and prefrontal cortex but not in liver or olfactory bulb. At the level of the brain, the delay of insulin signaling was associated with decreased levels of hypothalamic proopiomelanocortin, leading to increased food intake and obesity accompanied with hyperinsulinemia and hyperleptinemia. The loss of endothelial IRs also resulted in a delay in the acute hypoglycemic effect of systemic insulin administration and impaired glucose tolerance. In high-fat diet-treated mice, knockout of the endothelial IRs accelerated development of systemic insulin resistance but not food intake and obesity. Thus, IRs on endothelial cells have an important role in transendothelial insulin delivery in vivo which differentially regulates the kinetics of insulin signaling and insulin action in peripheral target tissues and different brain regions. Loss of this function predisposes animals to systemic insulin resistance, overeating, and obesity.
Collapse
|
296
|
Woods SC, May AA, Liu M, Tso P, Begg DP. Using the cerebrospinal fluid to understand ingestive behavior. Physiol Behav 2017; 178:172-178. [PMID: 27923718 PMCID: PMC5944842 DOI: 10.1016/j.physbeh.2016.11.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 01/08/2023]
Abstract
The cerebrospinal fluid (CSF) offers a window into the workings of the brain and blood-brain barrier (BBB). Molecules that enter into the central nervous system (CNS) by passive diffusion or receptor-mediated transport through the choroid plexus often appear in the CSF prior to acting within the brain. Other molecules enter the CNS by passing through the BBB into the brain's interstitial fluid prior to appearing in the CSF. This pattern is also often observed for molecules synthesized by neurons or glia within the CNS. The CSF is therefore an important conduit for the entry and clearance of molecules into/from the CNS and thereby constitutes an important window onto brain activity and barrier function. Assessing the CSF basally, under experimental conditions, or in the context of challenges or metabolic diseases can provide powerful insights about brain function. Here, we review important findings made by our labs, as influenced by the late Randall Sakai, by interrogating the CSF.
Collapse
Affiliation(s)
- Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Aaron A May
- Department of Pathology and Molecular Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Min Liu
- Department of Pathology and Molecular Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Patrick Tso
- Department of Pathology and Molecular Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Denovan P Begg
- School of Psychology, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
297
|
Song J, Whitcomb DJ, Kim BC. The role of melatonin in the onset and progression of type 3 diabetes. Mol Brain 2017; 10:35. [PMID: 28764741 PMCID: PMC5539639 DOI: 10.1186/s13041-017-0315-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 07/12/2017] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is defined by the excessive accumulation of toxic peptides, such as beta amyloid (Aβ) plaques and intracellular neurofibrillary tangles (NFT). The risk factors associated with AD include genetic mutations, aging, insulin resistance, and oxidative stress. To date, several studies that have demonstrated an association between AD and diabetes have revealed that the common risk factors include insulin resistance, sleep disturbances, blood brain barrier (BBB) disruption, and altered glucose homeostasis. Many researchers have discovered that there are mechanisms common to both diabetes and AD. AD that results from insulin resistance in the brain is termed “type 3 diabetes”. Melatonin synthesized by the pineal gland is known to contribute to circadian rhythms, insulin resistance, protection of the BBB, and cell survival mechanisms. Here, we review the relationship between melatonin and type 3 diabetes, and suggest that melatonin might regulate the risk factors for type 3 diabetes. We suggest that melatonin is crucial for attenuating the onset of type 3 diabetes by intervening in Aβ accumulation, insulin resistance, glucose metabolism, and BBB permeability.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, 61469, South Korea
| | - Daniel J Whitcomb
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, Faculty of Healthy Sciences, University of Bristol, Whitson street, Bristol, BS1 3NY, UK
| | - Byeong C Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea.
| |
Collapse
|
298
|
The diabetic brain and cognition. J Neural Transm (Vienna) 2017; 124:1431-1454. [PMID: 28766040 DOI: 10.1007/s00702-017-1763-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/13/2017] [Indexed: 12/20/2022]
Abstract
The prevalence of both Alzheimer's disease (AD) and vascular dementia (VaD) is increasing with the aging of the population. Studies from the last several years have shown that people with diabetes have an increased risk for dementia and cognitive impairment. Therefore, the authors of this consensus review tried to elaborate on the role of diabetes, especially diabetes type 2 (T2DM) in both AD and VaD. Based on the clinical and experimental work of scientists from 18 countries participating in the International Congress on Vascular Disorders and on literature search using PUBMED, it can be concluded that T2DM is a risk factor for both, AD and VaD, based on a pathology of glucose utilization. This pathology is the consequence of a disturbance of insulin-related mechanisms leading to brain insulin resistance. Although the underlying pathological mechanisms for AD and VaD are different in many aspects, the contribution of T2DM and insulin resistant brain state (IRBS) to cerebrovascular disturbances in both disorders cannot be neglected. Therefore, early diagnosis of metabolic parameters including those relevant for T2DM is required. Moreover, it is possible that therapeutic options utilized today for diabetes treatment may also have an effect on the risk for dementia. T2DM/IRBS contribute to pathological processes in AD and VaD.
Collapse
|
299
|
Insulin Treatment Prevents Neuroinflammation and Neuronal Injury with Restored Neurobehavioral Function in Models of HIV/AIDS Neurodegeneration. J Neurosci 2017; 36:10683-10695. [PMID: 27733618 DOI: 10.1523/jneurosci.1287-16.2016] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/26/2016] [Indexed: 12/17/2022] Open
Abstract
HIV-1 infection of the brain causes the neurodegenerative syndrome HIV-associated neurocognitive disorders (HAND), for which there is no specific treatment. Herein, we investigated the actions of insulin using ex vivo and in vivo models of HAND. Increased neuroinflammatory gene expression was observed in brains from patients with HIV/AIDS. The insulin receptor was detected on both neurons and glia, but its expression was unaffected by HIV-1 infection. Insulin treatment of HIV-infected primary human microglia suppressed supernatant HIV-1 p24 levels, reduced CXCL10 and IL-6 transcript levels, and induced peroxisome proliferator-activated receptor gamma (PPAR-γ) expression. Insulin treatment of primary human neurons prevented HIV-1 Vpr-mediated cell process retraction and death. In feline immunodeficiency virus (FIV) infected cats, daily intranasal insulin treatment (20.0 IU/200 μl for 6 weeks) reduced CXCL10, IL-6, and FIV RNA detection in brain, although PPAR-γ in glia was increased compared with PBS-treated FIV+ control animals. These molecular changes were accompanied by diminished glial activation in cerebral cortex and white matter of insulin-treated FIV+ animals, with associated preservation of cortical neurons. Neuronal counts in parietal cortex, striatum, and hippocampus were higher in the FIV+/insulin-treated group compared with the FIV+/PBS-treated group. Moreover, intranasal insulin treatment improved neurobehavioral performance, including both memory and motor functions, in FIV+ animals. Therefore, insulin exerted ex vivo and in vivo antiviral, anti-inflammatory, and neuroprotective effects in models of HAND, representing a new therapeutic option for patients with inflammatory or infectious neurodegenerative disorders including HAND. SIGNIFICANCE STATEMENT HIV-associated neurocognitive disorders (HAND) represent a spectrum disorder of neurocognitive dysfunctions resulting from HIV-1 infection. Although the exact mechanisms causing HAND are unknown, productive HIV-1 infection in the brain with associated neuroinflammation is a potential pathogenic mechanism resulting in neuronal damage and death. We report that, in HIV-infected microglia cultures, insulin treatment led to reduced viral replication and inflammatory gene expression. In addition, intranasal insulin treatment of experimentally feline immunodeficiency virus-infected animals resulted in improved motor and memory performances. We show that insulin restored expression of the nuclear receptor peroxisome proliferator-activated receptor gamma (PPAR-γ), which is suppressed by HIV-1 replication. Our findings indicate a unique function for insulin in improving neurological outcomes in lentiviral infections, implicating insulin as a therapeutic intervention for HAND.
Collapse
|
300
|
Lee LC, Goh MQL, Koo EH. Transcriptional regulation of APP by apoE: To boldly go where no isoform has gone before: ApoE, APP transcription and AD: Hypothesised mechanisms and existing knowledge gaps. Bioessays 2017; 39. [PMID: 28731260 DOI: 10.1002/bies.201700062] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia that gradually disrupts the brain network to impair memory, language and cognition. While the amyloid hypothesis remains the leading proposed mechanism to explain AD pathophysiology, anti-amyloid therapeutic strategies have yet to translate into useful therapies, suggesting that amyloid β-protein and its precursor, the amyloid precursor protein (APP) are but a part of the disease cascade. Further, risk of AD can be modulated by a number of factors, the most impactful being the ɛ4 isoform of apolipoprotein E (apoE). A recent study reported a novel isoform-dependent transcriptional regulation of APP by apoE. These interesting new results add to the myriad of mechanisms that have been proposed to explain how apoE4 enhances AD risk, highlighting the complexities of not only apoE and AD pathophysiology, but also of disease itself. Also see the video abstract here: https://youtu.be/yd14MBdPkCY.
Collapse
Affiliation(s)
- Liying Corinne Lee
- Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Michele Q L Goh
- Department of Medicine, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Edward H Koo
- Department of Medicine, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore.,Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|