351
|
Fang F, Gao Y, Wang T, Chen D, Liu J, Qian W, Cheng J, Gao R, Wang J, Xiao H. Insulin signaling disruption in male mice due to perinatal bisphenol A exposure: Role of insulin signaling in the brain. Toxicol Lett 2016; 245:59-67. [PMID: 26779933 DOI: 10.1016/j.toxlet.2016.01.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 01/10/2016] [Accepted: 01/11/2016] [Indexed: 12/16/2022]
Abstract
Bisphenol A (BPA), an environmental estrogenic endocrine disruptor, is widely used for producing polycarbonate plastics and epoxy resins. Available data have shown that perinatal exposure to BPA contributes to peripheral insulin resistance, while in the present study, we aimed to investigate the effects of perinatal BPA exposure on insulin signaling and glucose transport in the cortex of offspring mice. The pregnant mice were administrated either vehicle or BPA (100 μg/kg/day) at three perinatal stages. Stage I: from day 6 of gestation until parturition (P6-PND0 fetus exposure); Stage II: from lactation until delactation (PND0-PND21 newborn exposure) and Stage III: from day 6 of pregnancy until delactation (P6-PND21 fetus and newborn exposure). At 8 months of age for the offspring mice, the insulin signaling pathways and glucose transporters (GLUTs) were detected. Our data indicated that the insulin signaling including insulin, phosphorylated insulin receptor (IR), phosphorylated protein kinase B (p-AKT), phosphorylated glycogen synthase kinase 3β (p-GSK3β) and phosphorylated extracellular signal regulated protein kinase (p-ERK) were significantly decreased in the brain. In parallel, GLUTs (GLUT1/3/4) were obviously decreased as well in BPA-treated group in mice brain. Noteworthily, the phosphorylated tau (p-tau) and amyloid precursor protein (APP) were markedly up-regulated in all BPA-treated groups. These results, taken together, suggest the adverse effects of BPA on insulin signaling and GLUTs, which might subsequently contribute to the increment of p-tau and APP in the brain of adult offspring. Therefore, perinatal BPA exposure might be a risk factor for the long-term neurodegenerative changes in offspring male mice.
Collapse
Affiliation(s)
- Fangfang Fang
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, China
| | - Yue Gao
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, China
| | - Tingwei Wang
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, China
| | - Donglong Chen
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, China
| | - Jingli Liu
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, China; Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210000, China
| | - Wenyi Qian
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, China
| | - Jie Cheng
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, China
| | - Rong Gao
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, China
| | - Jun Wang
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, China.
| | - Hang Xiao
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, China.
| |
Collapse
|
352
|
Pang Y, Lin S, Wright C, Shen J, Carter K, Bhatt A, Fan LW. Intranasal insulin protects against substantia nigra dopaminergic neuronal loss and alleviates motor deficits induced by 6-OHDA in rats. Neuroscience 2016; 318:157-65. [PMID: 26777890 DOI: 10.1016/j.neuroscience.2016.01.020] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/22/2015] [Accepted: 01/08/2016] [Indexed: 01/04/2023]
Abstract
Protection of substantia nigra (SN) dopaminergic (DA) neurons by neurotrophic factors (NTFs) is one of the promising strategies in Parkinson's disease (PD) therapy. A major clinical challenge for NTF-based therapy is that NTFs need to be delivered into the brain via invasive means, which often shows limited delivery efficiency. The nose to brain pathway is a non-invasive brain drug delivery approach developed in recent years. Of particular interest is the finding that intranasal insulin improves cognitive functions in Alzheimer's patients. In vitro, insulin has been shown to protect neurons against various insults. Therefore, the current study was designed to test whether intranasal insulin could afford neuroprotection in the 6-hydroxydopamine (6-OHDA)-based rat PD model. 6-OHDA was injected into the right side of striatum to induce a progressive DA neuronal lesion in the ipsilateral SN pars compact (SNc). Recombinant human insulin was applied intranasally to rats starting from 24h post lesion, once per day, for 2 weeks. A battery of motor behavioral tests was conducted on day 8 and 15. The number of DA neurons in the SNc was estimated by stereological counting. Our results showed that 6-OHDA injection led to significant motor deficits and 53% of DA neuron loss in the ipsilateral side of injection. Treatment with insulin significantly ameliorated 6-OHDA-induced motor impairments, as shown by improved locomotor activity, tapered/ledged beam-walking performance, vibrissa-elicited forelimb-placing, initial steps, as well as methamphetamine-induced rotational behavior. Consistent with behavioral improvements, insulin treatment provided a potent protection of DA neurons in the SNc against 6-OHDA neurotoxicity, as shown by a 74.8% increase in tyrosine hydroxylase (TH)-positive neurons compared to the vehicle group. Intranasal insulin treatment did not affect body weight and blood glucose levels. In conclusion, our study showed that intranasal insulin provided strong neuroprotection in the 6-OHDA rat PD model, suggesting that insulin signaling may be a novel therapeutic target in broad neurodegenerative disorders.
Collapse
Affiliation(s)
- Y Pang
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States.
| | - S Lin
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - C Wright
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - J Shen
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - K Carter
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - A Bhatt
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - L-W Fan
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| |
Collapse
|
353
|
Kealy J, Campbell M. The Blood-Brain Barrier in Glioblastoma: Pathology and Therapeutic Implications. RESISTANCE TO TARGETED ANTI-CANCER THERAPEUTICS 2016. [DOI: 10.1007/978-3-319-46505-0_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
354
|
Lazard D, Vardi P, Bloch K. Anti-diabetic and neuroprotective effects of pancreatic islet transplantation into the central nervous system. Diabetes Metab Res Rev 2016; 32:11-20. [PMID: 25708430 DOI: 10.1002/dmrr.2644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/19/2015] [Indexed: 12/19/2022]
Abstract
During the last decades, the central nervous system (CNS) was intensively tested as a site for islet transplantation in different animal models of diabetes. Immunoprivilege properties of intracranial and intrathecal sites were found to delay and reduce rejection of transplanted allo-islets and xeno-islets, especially in the form of dispersed single cells. Insulin released from islets grafted in CNS was shown to cross the blood-brain barrier and to act as a regulator of peripheral glucose metabolism. In diabetic animals, sufficient nutrition and oxygen supply to islets grafted in the CNS provide adequate insulin response to increase glucose level resulting in rapid normoglycemia. In addition to insulin, pancreatic islets produce and secrete several other hormones, as well as neurotrophic and angiogenic factors with potential neuroprotective properties. Recent experimental studies and clinical trials provide a strong support for delivery of islet-derived macromolecules to CNS as a promising strategy to treat various brain disorders. This review article focuses mainly on analysis of current status of intracranial and intrathecal islet transplantations for treatment of experimental diabetes and discusses the possible neuroprotective properties of grafted islets into CNS as a novel therapeutic approach to brain disorders with cognitive dysfunctions characterized by impaired brain insulin signalling. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier
- Brain
- Central Nervous System
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/surgery
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/surgery
- Diabetic Neuropathies/prevention & control
- Disease Models, Animal
- Humans
- Hyperglycemia/prevention & control
- Hypoglycemia/prevention & control
- Insulin/metabolism
- Insulin Resistance
- Insulin Secretion
- Islets of Langerhans Transplantation/adverse effects
- Spinal Cord
- Subarachnoid Space
- Transplantation, Heterologous/adverse effects
- Transplantation, Heterotopic/adverse effects
Collapse
Affiliation(s)
- Daniel Lazard
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Pnina Vardi
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Konstantin Bloch
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| |
Collapse
|
355
|
Hughes TM, Craft S. The role of insulin in the vascular contributions to age-related dementia. Biochim Biophys Acta Mol Basis Dis 2015; 1862:983-91. [PMID: 26657615 DOI: 10.1016/j.bbadis.2015.11.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/17/2015] [Accepted: 11/29/2015] [Indexed: 01/12/2023]
Abstract
In addition to its well-known role in energy metabolism in the body, insulin is a vasoactive hormone that regulates peripheral and cerebral blood flow and neuronal function. Vascular and metabolic dysfunctions are emerging risk factors for Alzheimer's disease (AD) and age-related dementias, and recent evidence suggests that the two pathways are constitutive and interrelated. As a result, an emphasis on correcting metabolic disorders is emerging as an important strategy in the treatment and prevention of age-related cognitive impairment and AD. We review the evidence regarding the unique and interactive effects of vascular and metabolic disorders in pathological brain aging, with special consideration of the role of insulin dysregulation in promoting AD pathologic processes and vascular brain injury. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Timothy M Hughes
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Wake Forest University, USA
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Wake Forest University, USA.
| |
Collapse
|
356
|
Picone P, Ditta LA, Sabatino MA, Militello V, San Biagio PL, Di Giacinto ML, Cristaldi L, Nuzzo D, Dispenza C, Giacomazza D, Di Carlo M. Ionizing radiation-engineered nanogels as insulin nanocarriers for the development of a new strategy for the treatment of Alzheimer's disease. Biomaterials 2015; 80:179-194. [PMID: 26708643 DOI: 10.1016/j.biomaterials.2015.11.057] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/16/2015] [Accepted: 11/29/2015] [Indexed: 01/07/2023]
Abstract
A growing body of evidence shows the protective role of insulin in Alzheimer's disease (AD). A nanogel system (NG) to deliver insulin to the brain, as a tool for the development of a new therapy for Alzheimer's Disease (AD), is designed and synthetized. A carboxyl-functionalized poly(N-vinyl pyrrolidone) nanogel system produced by ionizing radiation is chosen as substrate for the covalent attachment of insulin or fluorescent molecules relevant for its characterization. Biocompatibility and hemocompatibility of the naked carrier is demonstrated. The insulin conjugated to the NG (NG-In) is protected by protease degradation and able to bind to insulin receptor (IR), as demonstrated by immunofluorescence measurements showing colocalization of NG-In(FITC) with IR. Moreover, after binding to the receptor, NG-In is able to trigger insulin signaling via AKT activation. Neuroprotection of NG-In against dysfunction induced by amyloid β (Aβ), a peptide mainly involved in AD, is verified. Finally, the potential of NG-In to be efficiently transported across the Blood Brain Barrier (BBB) is demonstrated. All together these results indicate that the synthesized NG-In is a suitable vehicle system for insulin deliver in biomedicine and a very promising tool to develop new therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Pasquale Picone
- Istituto di Biomedicina e Immunologia Molecolare "A. Monroy" (IBIM), Consiglio Nazionale delle Ricerche, Via U. La Malfa, 153, 90146 Palermo (PA), Italy
| | - Lorena Anna Ditta
- Dipartimento di Ingegneria Chimica, Gestionale, Informatica, Meccanica, Università di Palermo, Viale delle Scienze, Building 6, 90128 Palermo (PA), Italy
| | - Maria Antonietta Sabatino
- Dipartimento di Ingegneria Chimica, Gestionale, Informatica, Meccanica, Università di Palermo, Viale delle Scienze, Building 6, 90128 Palermo (PA), Italy
| | - Valeria Militello
- Dipartimento di Fisica e Chimica, Università di Palermo, Viale delle Scienze, Building 18, 90128 Palermo (PA), Italy
| | - Pier Luigi San Biagio
- Istituto di BioFisica (IBF), Consiglio Nazionale delle Ricerche, Via U. La Malfa, 153, 90146 Palermo (PA), Italy
| | - Maria Laura Di Giacinto
- Istituto di Biomedicina e Immunologia Molecolare "A. Monroy" (IBIM), Consiglio Nazionale delle Ricerche, Via U. La Malfa, 153, 90146 Palermo (PA), Italy
| | - Laura Cristaldi
- Istituto di Biomedicina e Immunologia Molecolare "A. Monroy" (IBIM), Consiglio Nazionale delle Ricerche, Via U. La Malfa, 153, 90146 Palermo (PA), Italy
| | - Domenico Nuzzo
- Istituto di Biomedicina e Immunologia Molecolare "A. Monroy" (IBIM), Consiglio Nazionale delle Ricerche, Via U. La Malfa, 153, 90146 Palermo (PA), Italy
| | - Clelia Dispenza
- Dipartimento di Ingegneria Chimica, Gestionale, Informatica, Meccanica, Università di Palermo, Viale delle Scienze, Building 6, 90128 Palermo (PA), Italy; School of Chemical Science and Engineering, Department of Fiber and Polymer Technology, Royal Institute of Technology (KTH), SE-100 44, Stockholm, Sweden.
| | - Daniela Giacomazza
- Istituto di BioFisica (IBF), Consiglio Nazionale delle Ricerche, Via U. La Malfa, 153, 90146 Palermo (PA), Italy.
| | - Marta Di Carlo
- Istituto di Biomedicina e Immunologia Molecolare "A. Monroy" (IBIM), Consiglio Nazionale delle Ricerche, Via U. La Malfa, 153, 90146 Palermo (PA), Italy.
| |
Collapse
|
357
|
Rosenblat JD, Brietzke E, Mansur RB, Maruschak NA, Lee Y, McIntyre RS. Inflammation as a neurobiological substrate of cognitive impairment in bipolar disorder: Evidence, pathophysiology and treatment implications. J Affect Disord 2015; 188:149-59. [PMID: 26363613 DOI: 10.1016/j.jad.2015.08.058] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/05/2015] [Accepted: 08/26/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Bipolar disorder (BD) has been associated with cognitive impairment during depressed, manic and euthymic periods. Inflammation has been shown to be involved in the pathophysiology of BD and cognitive impairment. METHODS For this systematic review, the MEDLINE/PubMed, Embase, Google Scholar and ClinicalTrials.gov databases were searched for relevant articles assessing the association between cognitive function and inflammatory markers in BD subjects. A discussion of potential mechanisms and therapeutic implications is also included to provide further context to the subject matter. RESULTS Eight studies, including a total of 555 BD subjects, assessing the association between cognitive function and inflammatory markers were identified. Cognitive dysfunction was associated with elevated levels of pro-inflammatory markers YKL40, IL-6, sCD40L, IL-1Ra, hsCRP and TNF-α. Mechanistically, elevation in inflammatory cytokines alters monoamine levels leading to cognitive and affective dysfunction. Neuro-inflammation, manifesting as microglial activation, leads to increased oxidative stress, pathologic synaptic pruning and impaired neuroplasticity in key brain regions sub-serving mood and cognition. Immune dysfunction also activates the hypothalamic-pituitary-adrenal (HPA) axis leading to hypercortisolemia and metabolic dysfunction, further promoting neuronal dysfunction. Anti-inflammatory agents are therefore currently being investigated in the treatment of BD and appear to exert an antidepressant effect; however, cognitive outcomes have yet to be reported. CONCLUSION Several studies suggest that immune dysfunction is associated with cognitive impairment in BD. Several neurobiological pathways have been identified whereby immune dysfunction may promote cognitive impairment in BD. Future investigations of anti-inflammatory agents targeting cognitive function as a treatment outcome are merited.
Collapse
Affiliation(s)
- Joshua D Rosenblat
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Elisa Brietzke
- Interdisciplinary Laboratory of Clinical Neurosciences (LINC), Federal University of Sao Paulo, Sao Paulo, Brazil; Program of Recognition and Intervention in Individuals in AT-Risk Mental States (PRISMA), Department of Psychiatry, Universidade FeInterdisciplinary Laboratory of Clinical Neurosciences (LINC), Federal University of Sao Pauloderal de São Paulo, São Paulo, Brazil
| | - Rodrigo B Mansur
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Interdisciplinary Laboratory of Clinical Neurosciences (LINC), Federal University of Sao Paulo, Sao Paulo, Brazil; Program of Recognition and Intervention in Individuals in AT-Risk Mental States (PRISMA), Department of Psychiatry, Universidade FeInterdisciplinary Laboratory of Clinical Neurosciences (LINC), Federal University of Sao Pauloderal de São Paulo, São Paulo, Brazil
| | - Nadia A Maruschak
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Yena Lee
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Roger S McIntyre
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
358
|
Fadel JR, Reagan LP. Stop signs in hippocampal insulin signaling: the role of insulin resistance in structural, functional and behavioral deficits. Curr Opin Behav Sci 2015; 9:47-54. [PMID: 26955646 DOI: 10.1016/j.cobeha.2015.12.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In peripheral tissues insulin activates signaling cascades to facilitate glucose uptake from the blood into tissues like liver, muscle and fat. While insulin appears to play a minor role in the regulation of glucose uptake in the central nervous system (CNS), insulin is known to play a major role in regulating synaptic plasticity in brain regions like the hippocampus. The concept that insulin regulates hippocampal neuroplasticity is further supported from animal models of type 2 diabetes (T2DM) and Alzheimer's disease (AD). The goal of this review is to provide an overview of these studies, as well as the studies that have examined whether deficits in hippocampal insulin signaling are amenable to intervention strategies.
Collapse
Affiliation(s)
- Jim R Fadel
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Lawrence P Reagan
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA; WJB Dorn Veterans Affairs Medical Center, Columbia, SC, USA
| |
Collapse
|
359
|
Kuo SC, Lai SW, Hung HC, Muo CH, Hung SC, Liu LL, Chang CW, Hwu YJ, Chen SL, Sung FC. Association between comorbidities and dementia in diabetes mellitus patients: population-based retrospective cohort study. J Diabetes Complications 2015; 29:1071-6. [PMID: 26233574 DOI: 10.1016/j.jdiacomp.2015.06.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 06/17/2015] [Accepted: 06/25/2015] [Indexed: 01/02/2023]
Abstract
AIMS Most diabetes mellitus (DM) patients have several comorbidities; the correlation of these comorbidities with dementia in DM requires clarification. METHODS Using claims data from Taiwan National Health Insurance, we identified 33,709 DM adults before the year 2000 and randomly selected 67,066 non-DM patients matched by sex and age. Subjects were followed until diagnosis with dementia, excluded due to death/withdrawal from the insurance program, or followed until 2011. We compared the incidence and hazard ratio (HR) for dementia in both cohorts. RESULTS Comorbidities were more prevalent in DM patients, including hypertension, hyperlipidemia, stroke, coronary artery and/or kidney disease. The HR was higher for the DM cohort with comorbidities than those without: 1.88 vs. 1.46 with hypertension; 1.56 vs. 1.39 with hyperlipidemia; 1.73 vs. 1.37 with coronary artery disease; 2.36 vs. 2.29 with stroke and 1.88 vs. 1.50 with kidney disease. The HR for dementia in diabetics rose from 1.41 in those without comorbidities to 2.49 in those with ≥4 comorbidities. In the DM cohort, HR was 1.22 for non-insulin-users and 1.41 for insulin-users, and 1.49 for type 1 DM and 1.23 for type 2 DM. CONCLUSION Diabetic patients have an elevated risk of dementia, and comorbidity increases this risk.
Collapse
Affiliation(s)
- Shu-Chen Kuo
- Department of Nursing, Lo-Sheng Sanatorium and Hospital, Ministry of Health and Welfare, New Taipei, 242, Taiwan; Department of Business Administration, Asia University, Taichung, 413, Taiwan
| | - Shih-Wei Lai
- College of Medicine, China Medical University, Taichung, 404, Taiwan; Department of Family Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Hung-Chang Hung
- Department of Internal Medicine, Nantou Hospital, Ministry of Health and Welfare, Nantou, 540, Taiwan
| | - Chih-Hsin Muo
- Management Office for Health Data, China Medical University Hospital, Taichung, 404, Taiwan
| | - Shih-Chang Hung
- Department of Emergency Medicine, Nantou Hospital, Ministry of Health and Welfare, Nantou, 540, Taiwan.
| | - Ling-Ling Liu
- Department of Emergency Medicine, Nantou Hospital, Ministry of Health and Welfare, Nantou, 540, Taiwan
| | - Chia-Wei Chang
- Department of Neurology, Nantou Hospital, Ministry of Health and Welfare, Nantou, 540, Taiwan
| | - Yueh-Juen Hwu
- College of Nursing, Central Taiwan University of Science and Technology, Taichung, 406, Taiwan
| | - Shieh-Liang Chen
- Department of Business Administration, Asia University, Taichung, 413, Taiwan
| | - Fung-Chung Sung
- Management Office for Health Data, China Medical University Hospital, Taichung, 404, Taiwan; Institute of Clinical Medical Science, China Medical University, Taichung, 404, Taiwan
| |
Collapse
|
360
|
HAMPL R, BIČÍKOVÁ M, SOSVOROVÁ L. How Hormones Influence Composition and Physiological Function of the Brain-Blood Barrier. Physiol Res 2015; 64:S259-64. [DOI: 10.33549/physiolres.933110] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Hormones exert many actions in the brain. Their access and effects in the brain are regulated by the blood-brain barrier (BBB). Hormones as other substances may enter the brain and vice versa either by paracellular way requiring breaching tight junctions stitching the endothelial cells composing the BBB, or by passage through the cells (transcellular way). Hormones influence both ways through their receptors, both membrane and intracellular, present on/in the BBB. In the review the main examples are outlined how hormones influence the expression and function of proteins forming the tight junctions, as well as how they regulate expression and function of major protein transporters mediating transport of various substances including hormone themselves.
Collapse
Affiliation(s)
- R. HAMPL
- Institute of Endocrinology, Prague, Czech Republic
| | | | | |
Collapse
|
361
|
Abstract
Clinical studies suggest a link between type 2 diabetes mellitus (T2DM) and insulin resistance (IR) and cognitive dysfunction, but there are significant gaps in our knowledge of the mechanisms underlying this relationship. Animal models of IR help to bridge these gaps and point to hippocampal IR as a potential mediator of cognitive dysfunction in T2DM, as well as in Alzheimer disease (AD). This Review highlights these observations and discusses intervention studies which suggest that the restoration of insulin activity in the hippocampus may be an effective strategy to alleviate the cognitive decline associated with T2DM and AD.
Collapse
|
362
|
Rosiglitazone improves learning and memory ability in rats with type 2 diabetes through the insulin signaling pathway. Am J Med Sci 2015; 350:121-8. [PMID: 25973687 DOI: 10.1097/maj.0000000000000499] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Diabetes mellitus (DM) is associated with moderate cognitive deficits and neurophysiologic and structural changes in the brain, a condition that is referred to as diabetic encephalopathy. This study was performed to investigate the effect of rosiglitazone (RSG) on learning and memory in rats with DM and elucidate possible mechanisms underlying this condition. Thirty-two male Sprague-Dawley rats were randomly divided into 4 groups: control (C, n = 8), DM (n = 8), RSG-administered control (C + RSG, n = 8) and RSG-administered DM groups (DM + RSG, n = 8). At 8 weeks after drug administration, Morris water maze was used to perform a training and probe trial to detect spatial learning and memory abilities. Western blot and immunohistochemistry were also used to detect changes in proteins involved in the insulin signal transduction pathway, such as the insulin receptor, insulin receptor substrate-1, protein kinase B, phosphorylated cAMP response element-binding protein and B-cell lymphoma 2, in the hippocampus of the rats. This study found that RSG could normalize the impaired insulin signal transduction in type 2 DM. The authors showed that RSG modulated the central insulin signaling axis.
Collapse
|
363
|
Zeng Y, Zhang L, Hu Z. Cerebral insulin, insulin signaling pathway, and brain angiogenesis. Neurol Sci 2015; 37:9-16. [PMID: 26442674 DOI: 10.1007/s10072-015-2386-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 09/23/2015] [Indexed: 01/06/2023]
Abstract
Insulin performs unique non-metabolic functions within the brain. Broadly speaking, two major areas of these functions are those related to brain endothelial cells and the blood-brain barrier (BBB) function, and those related to behavioral effects, like cognition in disease states (Alzheimer's disease, AD) and in health. Recent studies showed that both these functions are associated with brain angiogenesis. These findings raise interesting questions such as how they are linked to each other and whether modifying brain angiogenesis by targeting certain insulin signaling pathways could be an effective strategy to treat dementia as in AD, or even to help secure healthy longevity. The two canonical downstream pathways involved in mediating the insulin signaling pathway, the phosphoinositide-3 kinase (PI3K), and mitogen-activated protein kinase (MAPK) cascades, in the brain are supposed to be similar to those in the periphery. PI3K and MAPK pathways play important roles in angiogenesis. Both are involved in stimulating hypoxia inducible factor (HIF) in angiogenesis and could be activated by the insulin signaling pathway. This suggests that PI3K and MAPK pathways might act as cross-talk between the insulin signaling pathway and the angiogenesis pathway in brain. But the cerebral insulin, insulin signaling pathway, and the detailed mechanism in the connection of insulin signaling pathway, brain angiogenesis pathway, and healthy aging or dementias are still mostly not clear and need further studies.
Collapse
Affiliation(s)
- Yi Zeng
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Le Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Zhiping Hu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
364
|
Daniele G, Iozzo P, Molina-Carrion M, Lancaster J, Ciociaro D, Cersosimo E, Tripathy D, Triplitt C, Fox P, Musi N, DeFronzo R, Gastaldelli A. Exenatide Regulates Cerebral Glucose Metabolism in Brain Areas Associated With Glucose Homeostasis and Reward System. Diabetes 2015; 64:3406-12. [PMID: 26116695 PMCID: PMC6908417 DOI: 10.2337/db14-1718] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 06/21/2015] [Indexed: 01/01/2023]
Abstract
Glucagon-like peptide 1 receptors (GLP-1Rs) have been found in the brain, but whether GLP-1R agonists (GLP-1RAs) influence brain glucose metabolism is currently unknown. The study aim was to evaluate the effects of a single injection of the GLP-1RA exenatide on cerebral and peripheral glucose metabolism in response to a glucose load. In 15 male subjects with HbA1c of 5.7 ± 0.1%, fasting glucose of 114 ± 3 mg/dL, and 2-h glucose of 177 ± 11 mg/dL, exenatide (5 μg) or placebo was injected in double-blind, randomized fashion subcutaneously 30 min before an oral glucose tolerance test (OGTT). The cerebral glucose metabolic rate (CMRglu) was measured by positron emission tomography after an injection of [(18)F]2-fluoro-2-deoxy-d-glucose before the OGTT, and the rate of glucose absorption (RaO) and disposal was assessed using stable isotope tracers. Exenatide reduced RaO0-60 min (4.6 ± 1.4 vs. 13.1 ± 1.7 μmol/min ⋅ kg) and decreased the rise in mean glucose0-60 min (107 ± 6 vs. 138 ± 8 mg/dL) and insulin0-60 min (17.3 ± 3.1 vs. 24.7 ± 3.8 mU/L). Exenatide increased CMRglu in areas of the brain related to glucose homeostasis, appetite, and food reward, despite lower plasma insulin concentrations, but reduced glucose uptake in the hypothalamus. Decreased RaO0-60 min after exenatide was inversely correlated to CMRglu. In conclusion, these results demonstrate, for the first time in man, a major effect of a GLP-1RA on regulation of brain glucose metabolism in the absorptive state.
Collapse
Affiliation(s)
| | | | | | - Jack Lancaster
- University of Texas Health Science Center, San Antonio, TX
| | | | | | | | | | - Peter Fox
- University of Texas Health Science Center, San Antonio, TX
| | - Nicolas Musi
- University of Texas Health Science Center, San Antonio, TX
| | - Ralph DeFronzo
- University of Texas Health Science Center, San Antonio, TX
| | - Amalia Gastaldelli
- University of Texas Health Science Center, San Antonio, TX Institute of Clinical Physiology, Pisa, Italy
| |
Collapse
|
365
|
Rahmani-Nia F, Mirzaei B, Rahimi R. Effects of resistance exercise volume on appetite regulation and lipid profile in overweight young men. Sci Sports 2015. [DOI: 10.1016/j.scispo.2015.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
366
|
Soylu-Kucharz R, Adlesic N, Baldo B, Kirik D, Petersén Å. Hypothalamic overexpression of mutant huntingtin causes dysregulation of brown adipose tissue. Sci Rep 2015; 5:14598. [PMID: 26419281 PMCID: PMC4588570 DOI: 10.1038/srep14598] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 09/01/2015] [Indexed: 12/27/2022] Open
Abstract
Expression of mutant huntingtin (htt) protein has been shown to cause metabolic imbalance in animal models of Huntington disease (HD). The pathways involved are not fully understood but dysfunction of both the hypothalamus and brown adipose tissue (BAT) has been implicated. Here we show that targeted expression of mutant HTT in the hypothalamus leads to loss of the A13 dopaminergic cell group located in the zona incerta and reduced mRNA expression of neuropeptide Y1 receptor in the hypothalamus. Furthermore, this is accompanied by downregulation of uncoupling protein 1 expression and PPARγ coactivator-1 alpha in BAT and a rapid body weight gain. Taken together, our data might provide a mechanistic link between expression of mutant HTT, reduced activity of a hypothalamic dopaminergic pathway and dysfunction of BAT and in part explain the development of an obese phenotype in HD mouse models.
Collapse
Affiliation(s)
- Rana Soylu-Kucharz
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Sciences, Lund University, Sweden
| | - Natalie Adlesic
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Sciences, Lund University, Sweden
| | - Barbara Baldo
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Sciences, Lund University, Sweden
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (B.R.A.I.N.S.) Unit, Department of Experimental Medical Sciences Lund University, Sweden
| | - Åsa Petersén
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Sciences, Lund University, Sweden
| |
Collapse
|
367
|
Frazier DT, Bettcher BM, Dutt S, Patel N, Mungas D, Miller J, Green R, Kramer JH. Relationship between Insulin-Resistance Processing Speed and Specific Executive Function Profiles in Neurologically Intact Older Adults. J Int Neuropsychol Soc 2015; 21:622-8. [PMID: 26272269 PMCID: PMC4764989 DOI: 10.1017/s1355617715000624] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
This study investigated the relationship between insulin-resistance and constituent components of executive function in a sample of neurologically intact older adult subjects using the homeostasis model assessment (HOMA-IR) and latent factors of working memory, cognitive control and processing speed derived from confirmatory factor analysis. Low-density lipoprotein (LDL), mean arterial pressure (MAP), along with body mass index (BMI) and white matter hypointensity (WMH) were used to control for vascular risk factors, adiposity and cerebrovascular injury. The study included 119 elderly subjects recruited from the University of California, San Francisco Memory and Aging Center. Subjects underwent neuropsychological assessment, fasting blood draw and brain magnetic resonance imaging (MRI). Partial correlations and linear regression models were used to examine the HOMA-IR-executive function relationship. Pearson correlation adjusting for age showed a significant relationship between HOMA-IR and working memory (rp = -.18; p = .047), a trend with cognitive control (rp = -.17; p = .068), and no relationship with processing speed (rp = .013; p = .892). Linear regression models adjusting for demographic factors (age, education, and gender), LDL, MAP, BMI, and WMH indicated that HOMA-IR was negatively associated with cognitive control (r = -.256; p = .026) and working memory (r = -.234; p = .054). These results suggest a greater level of peripheral insulin-resistance is associated with decreased cognitive control and working memory. After controlling for demographic factors, vascular risk, adiposity and cerebrovascular injury, HOMA-IR remained significantly associated with cognitive control, with working memory showing a trend. These findings substantiate the insulin-resistance-executive function hypothesis and suggest a complex interaction, demonstrated by the differential impact of insulin-resistance on processing speed and specific aspects of executive function.
Collapse
Affiliation(s)
- Darvis T. Frazier
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Brianne M. Bettcher
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Shubir Dutt
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Nihar Patel
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Dan Mungas
- Department of Neurology, School of Medicine, University of California, Davis, Davis, CA 95817
| | - Joshua Miller
- Department of Pathology and Laboratory Medicine, University of California, Davis, Davis, CA 95817
| | - Ralph Green
- Department of Pathology and Laboratory Medicine, University of California, Davis, Davis, CA 95817
| | - Joel H. Kramer
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
368
|
Ma X, Fang F, Song M, Ma S. The effect of isoliquiritigenin on learning and memory impairments induced by high-fat diet via inhibiting TNF-α/JNK/IRS signaling. Biochem Biophys Res Commun 2015; 464:1090-1095. [DOI: 10.1016/j.bbrc.2015.07.081] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 07/16/2015] [Indexed: 12/29/2022]
|
369
|
Ross KA, Brenza TM, Binnebose AM, Phanse Y, Kanthasamy AG, Gendelman HE, Salem AK, Bartholomay LC, Bellaire BH, Narasimhan B. Nano-enabled delivery of diverse payloads across complex biological barriers. J Control Release 2015; 219:548-559. [PMID: 26315817 DOI: 10.1016/j.jconrel.2015.08.039] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/18/2015] [Accepted: 08/20/2015] [Indexed: 01/12/2023]
Abstract
Complex biological barriers are major obstacles for preventing and treating disease. Nanocarriers are designed to overcome such obstacles by enhancing drug delivery through physiochemical barriers and improving therapeutic indices. This review critically examines both biological barriers and nanocarrier payloads for a variety of drug delivery applications. A spectrum of nanocarriers is discussed that have been successfully developed for improving tissue penetration for preventing or treating a range of infectious, inflammatory, and degenerative diseases.
Collapse
Affiliation(s)
- Kathleen A Ross
- Chemical and Biological Engineering, Iowa State University, 2114 Sweeney Hall, Ames 50011, USA
| | - Timothy M Brenza
- Chemical and Biological Engineering, Iowa State University, 2114 Sweeney Hall, Ames 50011, USA
| | - Andrea M Binnebose
- Veterinary Microbiology and Preventive Medicine, Iowa State University, 2180 Vet Med, Ames 50011, USA
| | - Yashdeep Phanse
- Pathobiological Sciences, University of Wisconsin-Madison, 1656 Linden Dr., Madison 53706, USA
| | | | - Howard E Gendelman
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, Omaha 68198, USA
| | - Aliasger K Salem
- Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, 115 S. Grand Avenue, Iowa City 52242, USA
| | - Lyric C Bartholomay
- Pathobiological Sciences, University of Wisconsin-Madison, 1656 Linden Dr., Madison 53706, USA
| | - Bryan H Bellaire
- Veterinary Microbiology and Preventive Medicine, Iowa State University, 2180 Vet Med, Ames 50011, USA
| | - Balaji Narasimhan
- Chemical and Biological Engineering, Iowa State University, 2114 Sweeney Hall, Ames 50011, USA.
| |
Collapse
|
370
|
Krishnan N, Krishnan K, Connors CR, Choy MS, Page R, Peti W, Van Aelst L, Shea SD, Tonks NK. PTP1B inhibition suggests a therapeutic strategy for Rett syndrome. J Clin Invest 2015. [PMID: 26214522 DOI: 10.1172/jci80323] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The X-linked neurological disorder Rett syndrome (RTT) presents with autistic features and is caused primarily by mutations in a transcriptional regulator, methyl CpG-binding protein 2 (MECP2). Current treatment options for RTT are limited to alleviating some neurological symptoms; hence, more effective therapeutic strategies are needed. We identified the protein tyrosine phosphatase PTP1B as a therapeutic candidate for treatment of RTT. We demonstrated that the PTPN1 gene, which encodes PTP1B, was a target of MECP2 and that disruption of MECP2 function was associated with increased levels of PTP1B in RTT models. Pharmacological inhibition of PTP1B ameliorated the effects of MECP2 disruption in mouse models of RTT, including improved survival in young male (Mecp2-/y) mice and improved behavior in female heterozygous (Mecp2-/+) mice. We demonstrated that PTP1B was a negative regulator of tyrosine phosphorylation of the tyrosine kinase TRKB, the receptor for brain-derived neurotrophic factor (BDNF). Therefore, the elevated PTP1B that accompanies disruption of MECP2 function in RTT represents a barrier to BDNF signaling. Inhibition of PTP1B led to increased tyrosine phosphorylation of TRKB in the brain, which would augment BDNF signaling. This study presents PTP1B as a mechanism-based therapeutic target for RTT, validating a unique strategy for treating the disease by modifying signal transduction pathways with small-molecule drugs.
Collapse
|
371
|
Bilotta F, Lauretta MP, Tewari A, Haque M, Hara N, Uchino H, Rosa G. Insulin and the Brain: A Sweet Relationship With Intensive Care. J Intensive Care Med 2015; 32:48-58. [PMID: 26168800 DOI: 10.1177/0885066615594341] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/28/2015] [Accepted: 05/15/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Insulin receptors (IRs) in the brain have unique molecular features and a characteristic pattern of distribution. Their possible functions extend beyond glucose utilization. In this systematic review, we explore the interactions between insulin and the brain and its implications for anesthesiologists, critical care physicians, and other medical disciplines. METHODS A literature search of published preclinical and clinical studies between 1978 and 2014 was conducted, yielding 5996 articles. After applying inclusion and exclusion criteria, 92 studies were selected for this systematic review. RESULTS The IRs have unique molecular features, pattern of distribution, and mechanism of action. It has effects on neuronal function, metabolism, and neurotransmission. The IRs are involved in neuronal apoptosis and neurodegenerative processes. CONCLUSION In this systematic review, we present a close relationship between insulin and the brain, with discernible effects on memory, learning abilities, and motor functions. The potential therapeutic effects extend from acute brain insults such as traumatic brain injury, brain ischemia, and hemorrhage, to chronic neurodegenerative diseases such as Alzheimer and Parkinson disease. An understanding of the wider effects of insulin conveyed in this review will prompt anaesthesiologists and critical care physicians to consider its therapeutic potential and guide future studies.
Collapse
Affiliation(s)
- F Bilotta
- Department of Anesthesiology, Critical Care and Pain Medicine, "Sapienza" University of Rome, Rome, Italy
| | - M P Lauretta
- Anesthesia and Intensive Care Department, "La Sapienza" University of Rome, Rome, Italy .,Critical Care Department, University College London Hospital, London, United Kingdom
| | - A Tewari
- Department of Pediatric Neuroanesthesia and IONM, Cincinnati Children Hospital & Medical Center, Cincinnati, OH, USA
| | - M Haque
- Anesthesia and Critical Care Department, University College London Hospital, London, United Kingdom
| | - N Hara
- Department of Anesthesiology, Tokyo Medical University, Tokyo, Japan
| | - H Uchino
- Department of Anesthesiology, Tokyo Medical University, Tokyo, Japan
| | - G Rosa
- Department of Anesthesiology, Critical Care and Pain Medicine, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
372
|
Bedse G, Di Domenico F, Serviddio G, Cassano T. Aberrant insulin signaling in Alzheimer's disease: current knowledge. Front Neurosci 2015; 9:204. [PMID: 26136647 PMCID: PMC4468388 DOI: 10.3389/fnins.2015.00204] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 05/22/2015] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia affecting elderly people. AD is a multifaceted pathology characterized by accumulation of extracellular neuritic plaques, intracellular neurofibrillary tangles (NFTs) and neuronal loss mainly in the cortex and hippocampus. AD etiology appears to be linked to a multitude of mechanisms that have not been yet completely elucidated. For long time, it was considered that insulin signaling has only peripheral actions but now it is widely accepted that insulin has neuromodulatory actions in the brain. Insulin signaling is involved in numerous brain functions including cognition and memory that are impaired in AD. Recent studies suggest that AD may be linked to brain insulin resistance and patients with diabetes have an increased risk of developing AD compared to healthy individuals. Indeed insulin resistance, increased inflammation and impaired metabolism are key pathological features of both AD and diabetes. However, the precise mechanisms involved in the development of AD in patients with diabetes are not yet fully understood. In this review we will discuss the role played by aberrant brain insulin signaling in AD. In detail, we will focus on the role of insulin signaling in the deposition of neuritic plaques and intracellular NFTs. Considering that insulin mitigates beta-amyloid deposition and phosphorylation of tau, pharmacological strategies restoring brain insulin signaling, such as intranasal delivery of insulin, could have significant therapeutic potential in AD treatment.
Collapse
Affiliation(s)
- Gaurav Bedse
- Department of Physiology and Pharmacology "V. Erspamer," Sapienza University of Rome Rome, Italy ; Department of Biochemical Sciences, Sapienza University of Rome Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome Rome, Italy
| | - Gaetano Serviddio
- Department of Medical and Surgical Sciences, University of Foggia Foggia, Italy
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| |
Collapse
|
373
|
Vandal M, Bourassa P, Calon F. Can insulin signaling pathways be targeted to transport Aβ out of the brain? Front Aging Neurosci 2015; 7:114. [PMID: 26136681 PMCID: PMC4468380 DOI: 10.3389/fnagi.2015.00114] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 05/29/2015] [Indexed: 12/11/2022] Open
Abstract
Although the causal role of Amyloid-β (Aβ) in Alzheimer’s disease (AD) is unclear, it is still reasonable to expect that lowering concentrations of Aβ in the brain may decrease the risk of developing the neurocognitive symptoms of the disease. Brain capillary endothelial cells forming the blood-brain barrier (BBB) express transporters regulating the efflux of Aβ out of the cerebral tissue. Age-related BBB dysfunctions, that have been identified in AD patients, might impair Aβ clearance from the brain. Thus, targeting BBB outward transport systems has been suggested as a way to stimulate the clearance of Aβ from the brain. Recent data indicate that the increase in soluble brain Aβ and behavioral impairments in 3×Tg-AD mice generated by months of intake of a high-fat diet can be acutely reversed by the administration of a single dose of insulin. A concomitant increase in plasma Aβ suggests that clearance from the brain through the BBB is a likely mechanism for this rapid effect of insulin. Here, we review how BBB insulin response pathways could be stimulated to decrease brain Aβ concentrations and improve cognitive performance, at least on the short term.
Collapse
Affiliation(s)
- Milene Vandal
- Faculté de Pharmacie, Université Laval Quebec, QC, Canada ; Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUL) Québec, QC, Canada ; Institut des Nutraceutiques et des Aliments Fonctionnels, Université Laval Québec, QC, Canada
| | - Philippe Bourassa
- Faculté de Pharmacie, Université Laval Quebec, QC, Canada ; Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUL) Québec, QC, Canada ; Institut des Nutraceutiques et des Aliments Fonctionnels, Université Laval Québec, QC, Canada
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval Quebec, QC, Canada ; Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUL) Québec, QC, Canada ; Institut des Nutraceutiques et des Aliments Fonctionnels, Université Laval Québec, QC, Canada
| |
Collapse
|
374
|
Lioutas VA, Alfaro-Martinez F, Bedoya F, Chung CC, Pimentel DA, Novak V. Intranasal Insulin and Insulin-Like Growth Factor 1 as Neuroprotectants in Acute Ischemic Stroke. Transl Stroke Res 2015; 6:264-75. [PMID: 26040423 DOI: 10.1007/s12975-015-0409-7] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 04/16/2015] [Accepted: 05/13/2015] [Indexed: 12/22/2022]
Abstract
Treatment options for stroke remain limited. Neuroprotective therapies, in particular, have invariably failed to yield the expected benefit in stroke patients, despite robust theoretical and mechanistic background and promising animal data. Insulin and insulin-like growth factor 1 (IGF-1) play a pivotal role in critical brain functions, such as energy homeostasis, neuronal growth, and differentiation. They may exhibit neuroprotective properties in acute ischemic stroke based upon their vasodilatory, anti-inflammatory and antithrombotic effects, as well as improvements of functional connectivity, neuronal metabolism, neurotransmitter regulation, and remyelination. Intranasally administered insulin has demonstrated a benefit for prevention of cognitive decline in older people, and IGF-1 has shown potential benefit to improve functional outcomes in animal models of acute ischemic stroke. The intranasal route presents a feasible, tolerable, safe, and particularly effective administration route, bypassing the blood-brain barrier and maximizing distribution to the central nervous system (CNS), without the disadvantages of systemic side effects and first-pass metabolism. This review summarizes the neuroprotective potential of intranasally administered insulin and IGF-1 in stroke patients. We present the theoretical background and pathophysiologic mechanisms, animal and human studies of intranasal insulin and IGF-1, and the safety and feasibility of intranasal route for medication administration to the CNS.
Collapse
Affiliation(s)
- Vasileios-Arsenios Lioutas
- Department of Neurology, Division of Cerebrovascular Diseases, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Palmer 127, Boston, MA, 02215, USA,
| | | | | | | | | | | |
Collapse
|
375
|
Petrov D, Pedrós I, Artiach G, Sureda FX, Barroso E, Pallàs M, Casadesús G, Beas-Zarate C, Carro E, Ferrer I, Vazquez-Carrera M, Folch J, Camins A. High-fat diet-induced deregulation of hippocampal insulin signaling and mitochondrial homeostasis deficiences contribute to Alzheimer disease pathology in rodents. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1687-99. [PMID: 26003667 DOI: 10.1016/j.bbadis.2015.05.004] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 04/09/2015] [Accepted: 05/06/2015] [Indexed: 01/04/2023]
Abstract
Global obesity is a pandemic status, estimated to affect over 2 billion people, that has resulted in an enormous strain on healthcare systems worldwide. The situation is compounded by the fact that apart from the direct costs associated with overweight pathology, obesity presents itself with a number of comorbidities, including an increased risk for the development of neurodegenerative disorders. Alzheimer disease (AD), the main cause of senile dementia, is no exception. Spectacular failure of the pharmaceutical industry to come up with effective AD treatment strategies is forcing the broader scientific community to rethink the underlying molecular mechanisms leading to cognitive decline. To this end, the emphasis is once again placed on the experimental animal models of the disease. In the current study, we have focused on the effects of a high-fat diet (HFD) on hippocampal-dependent memory in C57/Bl6 Wild-type (WT) and APPswe/PS1dE9 (APP/PS1) mice, a well-established mouse model of familial AD. Our results indicate that the continuous HFD administration starting at the time of weaning is sufficient to produce β-amyloid-independent, hippocampal-dependent memory deficits measured by a 2-object novel-object recognition test (NOR) in mice as early as 6months of age. Furthermore, the resulting metabolic syndrome appears to have direct effects on brain insulin regulation and mitochondrial function. We have observed pathological changes related to both the proximal and distal insulin signaling pathway in the brains of HFD-fed WT and APP/PS1 mice. These changes are accompanied by a significantly reduced OXPHOS metabolism, suggesting that mitochondria play an important role in hippocampus-dependent memory formation and retention in both the HFD-treated and AD-like rodents at a relatively young age.
Collapse
Affiliation(s)
- Dmitry Petrov
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina de la UB (IBUB), Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ignacio Pedrós
- Unitats de Bioquímica i Farmacologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus (Tarragona), Spain; Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Gonzalo Artiach
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina de la UB (IBUB), Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Francesc X Sureda
- Unitats de Bioquímica i Farmacologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus (Tarragona), Spain; Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Emma Barroso
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina de la UB (IBUB), Universitat de Barcelona, Barcelona, Spain; Centros de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Mercè Pallàs
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina de la UB (IBUB), Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Gemma Casadesús
- Department of Biological Sciences Kent State University, Kent, OH, USA
| | - Carlos Beas-Zarate
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, CIBO, IMSS, México; Laboratorio de Regeneración y Desarrollo Neural, Instituto de Neurobiología, Departamento de Biología Celular y Molecular, CUCBA, México
| | - Eva Carro
- Neuroscience Group, Instituto de Investigacion Hospital 12 de Octubre, Madrid, Spain
| | - Isidro Ferrer
- Institute of Neuropathology, Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - Manuel Vazquez-Carrera
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina de la UB (IBUB), Universitat de Barcelona, Barcelona, Spain; Centros de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Jaume Folch
- Unitats de Bioquímica i Farmacologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus (Tarragona), Spain; Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Antoni Camins
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina de la UB (IBUB), Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Universidad Nacional de Loja, Department of Biotechnology, Ecuador.
| |
Collapse
|
376
|
Niyomchan A, Watcharasit P, Visitnonthachai D, Homkajorn B, Thiantanawat A, Satayavivad J. Insulin attenuates arsenic-induced neurite outgrowth impairments by activating the PI3K/Akt/SIRT1 signaling pathway. Toxicol Lett 2015; 236:138-44. [PMID: 25982963 DOI: 10.1016/j.toxlet.2015.05.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/07/2015] [Accepted: 05/11/2015] [Indexed: 10/23/2022]
Abstract
Arsenic neurotoxicity has a broad range of adverse effects on human health, which are induced in part by inhibition of neurite outgrowth. Insulin has been reported to promote neurite extension. The present study investigated whether insulin can protect neurons from impaired neurite outgrowth induced by arsenic, and examined the signaling pathway involved in this action. The study demonstrated that NaAsO2 caused inhibition of neurite outgrowth in differentiated SH-SY5Y cells indicating its neurotoxicity. This inhibitory effect of NaAsO2 was attenuated by insulin. It was found that blocking PI3K or Akt by selective inhibitors canceled the protective effect of insulin against NaAsO2-induced neurite outgrowth impairment suggesting the essential role of active PI3K and Akt in insulin's protective action. Inhibition of GSK3, which mimics an effect of insulin stimulation, had no effect on the impairment of neurite outgrowth by NaAsO2 implying that the insulin protective action is probably not due to its mediation of GSK3 inhibition ability. Moreover, NaAsO2 decreased the Akt activity, as it caused reduction in Akt phosphorylation, and downregulated expression of SIRT1. Additionally, the reduction of these signals by NaAsO2 was attenuated by insulin. Taken together, these results show that insulin attenuates arsenic-induced neurite outgrowth impairment possibly via activation of PI3K/Akt/SIRT1 signaling, and arsenic may exert neurite outgrowth inhibition through a mechanism involving reduction of signaling molecules downstream from insulin, PI3K/Akt/SIRT1. Our findings raise the possibility of using insulin to combat arsenic neurotoxicity.
Collapse
Affiliation(s)
- Apichaya Niyomchan
- Laboratory of Pharmacology, Chulabhorn Research Institute, Thailand; Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6 Rd, Bangkok 10210, Thailand
| | - Piyajit Watcharasit
- Laboratory of Pharmacology, Chulabhorn Research Institute, Thailand; Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6 Rd, Bangkok 10210, Thailand; Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Higher Education Commission, Thailand.
| | | | | | - Apinya Thiantanawat
- Laboratory of Pharmacology, Chulabhorn Research Institute, Thailand; Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6 Rd, Bangkok 10210, Thailand; Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Higher Education Commission, Thailand
| | - Jutamaad Satayavivad
- Laboratory of Pharmacology, Chulabhorn Research Institute, Thailand; Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6 Rd, Bangkok 10210, Thailand; Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Higher Education Commission, Thailand
| |
Collapse
|
377
|
Meredith ME, Salameh TS, Banks WA. Intranasal Delivery of Proteins and Peptides in the Treatment of Neurodegenerative Diseases. AAPS JOURNAL 2015; 17:780-7. [PMID: 25801717 DOI: 10.1208/s12248-015-9719-7] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/12/2015] [Indexed: 12/28/2022]
Abstract
The blood-brain barrier (BBB) is a major impediment to the therapeutic delivery of peptides and proteins to the brain. Intranasal delivery often provides a non-invasive means to bypass the BBB. Advantages of using intranasal delivery include minimizing exposure to peripheral organs and tissues, thus reducing systemic side effects. It also allows substances that typically have rapid degradation in the blood time to exert their effect. Intranasal delivery provides the ability to target proteins and peptides to specific regions of the brain when administered with substrates like cyclodextrins. In this review, we examined the use of intranasal delivery of various proteins and peptides that have implications in the treatment of neurodegenerative diseases, focusing especially on albumin, exendin/GLP-1, GALP, insulin, leptin, and PACAP. We have described their rationale for use, distribution in the brain after intranasal injection, how intranasal administration differed from other modes of delivery, and their use in clinical trials, if applicable. Intranasal delivery of drugs, peptides, and other proteins could be very useful in the future for the prevention or treatment of brain related diseases.
Collapse
|
378
|
Cline BH, Costa-Nunes JP, Cespuglio R, Markova N, Santos AI, Bukhman YV, Kubatiev A, Steinbusch HWM, Lesch KP, Strekalova T. Dicholine succinate, the neuronal insulin sensitizer, normalizes behavior, REM sleep, hippocampal pGSK3 beta and mRNAs of NMDA receptor subunits in mouse models of depression. Front Behav Neurosci 2015; 9:37. [PMID: 25767439 PMCID: PMC4341562 DOI: 10.3389/fnbeh.2015.00037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/01/2015] [Indexed: 11/13/2022] Open
Abstract
Central insulin receptor-mediated signaling is attracting the growing attention of researchers because of rapidly accumulating evidence implicating it in the mechanisms of plasticity, stress response, and neuropsychiatric disorders including depression. Dicholine succinate (DS), a mitochondrial complex II substrate, was shown to enhance insulin-receptor mediated signaling in neurons and is regarded as a sensitizer of the neuronal insulin receptor. Compounds enhancing neuronal insulin receptor-mediated transmission exert an antidepressant-like effect in several pre-clinical paradigms of depression; similarly, such properties for DS were found with a stress-induced anhedonia model. Here, we additionally studied the effects of DS on several variables which were ameliorated by other insulin receptor sensitizers in mice. Pre-treatment with DS of chronically stressed C57BL6 mice rescued normal contextual fear conditioning, hippocampal gene expression of NMDA receptor subunit NR2A, the NR2A/NR2B ratio and increased REM sleep rebound after acute predation. In 18-month-old C57BL6 mice, a model of elderly depression, DS restored normal sucrose preference and activated the expression of neural plasticity factors in the hippocampus as shown by Illumina microarray. Finally, young naïve DS-treated C57BL6 mice had reduced depressive- and anxiety-like behaviors and, similarly to imipramine-treated mice, preserved hippocampal levels of the phosphorylated (inactive) form of GSK3 beta that was lowered by forced swimming in pharmacologically naïve animals. Thus, DS can ameliorate behavioral and molecular outcomes under a variety of stress- and depression-related conditions. This further highlights neuronal insulin signaling as a new factor of pathogenesis and a potential pharmacotherapy of affective pathologies.
Collapse
Affiliation(s)
- Brandon H Cline
- Faculté de Médecine, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg Strasbourg, France
| | - Joao P Costa-Nunes
- Department of Neuroscience, Maastricht University Maastricht, Netherlands ; Group of Behavioural Neuroscience and Pharmacology, Institute for Hygiene and Tropical Medicine, New University of Lisbon Lisbon, Portugal
| | - Raymond Cespuglio
- Faculty of Medicine, Neuroscience Research Center of Lyon, INSERM U1028, C. Bernard University Lyon, France
| | - Natalyia Markova
- Laboratory of Biomolecular Screening, Institute of Physiologically Active Compounds, Russian Academy of Sciences Moscow, Russia ; Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences Moscow, Russia
| | - Ana I Santos
- Faculdade de Ciências Médicas, NOVA Medical School, Universidade Nova de Lisboa Lisboa, Portugal
| | - Yury V Bukhman
- Great Lakes Bioenergy Research Center, Computational Biology, Wisconsin Energy Institute, University of Wisconsin Madison, WI, USA
| | - Aslan Kubatiev
- Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences Moscow, Russia
| | | | - Klaus-Peter Lesch
- Department of Neuroscience, Maastricht University Maastricht, Netherlands ; Laboratory of Translational Neuroscience, Division of Molecular Psychiatry, Centre of Mental Health, University of Wuerzburg Wuerzburg, Germany
| | - Tatyana Strekalova
- Department of Neuroscience, Maastricht University Maastricht, Netherlands ; Group of Behavioural Neuroscience and Pharmacology, Institute for Hygiene and Tropical Medicine, New University of Lisbon Lisbon, Portugal ; Laboratory of Biomolecular Screening, Institute of Physiologically Active Compounds, Russian Academy of Sciences Moscow, Russia
| |
Collapse
|
379
|
Abstract
The pancreatic hormone insulin plays a well-described role in the periphery, based principally on its ability to lower circulating glucose levels via activation of glucose transporters. However, insulin also acts within the central nervous system (CNS) to alter a number of physiological outcomes ranging from energy balance and glucose homeostasis to cognitive performance. Insulin is transported into the CNS by a saturable receptor-mediated process that is proposed to be dependent on the insulin receptor. Transport of insulin into the brain is dependent on numerous factors including diet, glycemia, a diabetic state and notably, obesity. Obesity leads to a marked decrease in insulin transport from the periphery into the CNS and the biological basis of this reduction of transport remains unresolved. Despite decades of research into the effects of central insulin on a wide range of physiological functions and its transport from the periphery to the CNS, numerous questions remain unanswered including which receptor is responsible for transport and the precise mechanisms of action of insulin within the brain.
Collapse
Affiliation(s)
- Denovan P Begg
- School of Psychology, University of New South Wales (UNSW, Australia), Sydney, New South Wales, Australia.
| |
Collapse
|
380
|
Enabling nanomaterial, nanofabrication and cellular technologies for nanoneuromedicines. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:715-29. [PMID: 25652894 DOI: 10.1016/j.nano.2014.12.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 12/15/2014] [Accepted: 12/18/2014] [Indexed: 12/11/2022]
Abstract
Nanoparticulate delivery systems represent an area of particular promise for nanoneuromedicines. They possess significant potential for desperately needed therapies designed to combat a range of disorders associated with aging. As such, the field was selected as the focus for the 2014 meeting of the American Society for Nanomedicine. Regenerative, protective, immune modulatory, anti-microbial and anti-inflammatory products, or imaging agents are readily encapsulated in or conjugated to nanoparticles and as such facilitate the delivery of drug payloads to specific action sites across the blood-brain barrier. Diagnostic imaging serves to precisely monitor disease onset and progression while neural stem cell replacement can regenerate damaged tissue through control of stem cell fates. These, taken together, can improve disease burden and limit systemic toxicities. Such enabling technologies serve to protect the nervous system against a broad range of degenerative, traumatic, metabolic, infectious and immune disorders. From the clinical editor: Nanoneuromedicine is a branch of nanomedicine that specifically looks at the nervous system. In the clinical setting, a fundamental hurdle in nervous system disorders is due to an inherent inability of nerve cells to regenerate after damage. Nanotechnology can offer new approaches to overcome these challenges. This review describes recent developments in nanomedicine delivery systems that would affect stem cell repair and regeneration in the nervous system.
Collapse
|
381
|
Burette AC, Park H, Weinberg RJ. Postsynaptic distribution of IRSp53 in spiny excitatory and inhibitory neurons. J Comp Neurol 2015; 522:2164-78. [PMID: 24639075 DOI: 10.1002/cne.23526] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 12/12/2013] [Accepted: 12/12/2013] [Indexed: 11/07/2022]
Abstract
The 53 kDa insulin receptor substrate protein (IRSp53) is highly enriched in the brain. Despite evidence that links mutations of IRSp53 with autism and other neuropsychiatric problems, the functional significance of this protein remains unclear. We used light and electron microscopic immunohistochemistry to demonstrate that IRSp53 is expressed throughout the adult rat brain. Labeling concentrated selectively in dendritic spines, where it was associated with the postsynaptic density (PSD). Surprisingly, its organization within the PSD of spiny excitatory neurons of neocortex and hippocampus differed from that within spiny inhibitory neurons of neostriatum and cerebellar cortex. The present data support previous suggestions that IRSp53 is involved in postsynaptic signaling, while hinting that its signaling role may differ in different types of neurons.
Collapse
Affiliation(s)
- Alain C Burette
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | | | | |
Collapse
|
382
|
Hypothalamic differences in expression of genes involved in monoamine synthesis and signaling pathways after insulin injection in chickens from lines selected for high and low body weight. Neurogenetics 2015; 16:133-44. [PMID: 25582322 DOI: 10.1007/s10048-014-0435-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/17/2014] [Indexed: 10/24/2022]
Abstract
Long-term selection for juvenile body weight from a common founder population resulted in two divergent chicken lines (low-weight selected line (LWS), high-weight selected line (HWS)) that display distinct food intake and blood glucose responses to exogenous neuropeptides and insulin. The objective of this study was to elucidate putative targets affecting food intake and energy homeostasis by sequencing hypothalamic RNA from LWS and HWS chickens after insulin injection. Ninety-day-old female LWS and HWS chickens were injected with either vehicle or insulin and hypothalamus collected at 1 h postinjection. Through RNA sequencing, a total of 361 differentially expressed genes (DEGs) were identified. There was greater expression of genes, mainly tyrosine hydroxylase (TH), L-aromatic amino acid decarboxylase (DDC), and vesicular monoamine transporter (VMAT), involved in serotonin and dopamine biosynthesis and signaling in LWS than in HWS vehicle-injected chickens. In contrast, after insulin injection, these genes were more highly expressed in HWS than in LWS. We identified 90 single nucleotide polymorphisms (SNPs) existing only in the HWS and 121 SNPs specific to LWS and 5119 SNPs close to fixation (with absolute frequency difference ≥0.9). Four were located in genes encoding enzymes associated with serotonergic and dopaminergic pathways, such as DDC, TH, and solute carrier family 18, member 2 (VMAT). These data implicate differences in biogenic amines such as serotonin and dopamine in hypothalamic physiology between the chicken lines, and these differences might be associated with polymorphisms during long-term selection. Changes in serotonergic and dopaminergic signaling pathways in response to insulin injection suggest a role in whole-body energy homeostasis.
Collapse
|
383
|
Mauro C, De Rosa V, Marelli-Berg F, Solito E. Metabolic syndrome and the immunological affair with the blood-brain barrier. Front Immunol 2015; 5:677. [PMID: 25601869 PMCID: PMC4283608 DOI: 10.3389/fimmu.2014.00677] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/16/2014] [Indexed: 12/29/2022] Open
Abstract
Epidemiological studies reveal an increased incidence of obesity worldwide, which is associated with increased prevalence and severity of cognitive disorders. The blood–brain barrier (BBB) represents the interface between the peripheral circulation and the brain, and plays a fundamental role in the cross-talk between these two compartments. The homeostatic function of the BBB is the protection of the brain from peripheral insult/inflammation. Alterations in the function of the BBB lead to pathologies of the central nervous system. Recently, metabolic imbalance has been shown to be an important risk factor associated with the decline of BBB integrity and function. This has direct etiological consequences on a variety of cerebrovascular and neurodegenerative pathologies with great impact to society. Priority areas for future preclinical research include strategies to improve clinicians’ ability to diagnose, prevent, and manage BBB abnormalities. In sharp contrast with epidemiological studies and clinical needs, little is known about the mechanisms that link metabolic syndrome to BBB functionality and cognitive disorders. Our view is that immune responses caused by metabolic stress might play a major role in this conundrum.
Collapse
Affiliation(s)
- Claudio Mauro
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London , London , UK
| | - Veronica De Rosa
- Istituto Per L'Endocrinologia e L'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (IEOS-CNR) , Naples , Italy
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London , London , UK
| | - Egle Solito
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London , London , UK
| |
Collapse
|
384
|
Calvo-Ochoa E, Arias C. Cellular and metabolic alterations in the hippocampus caused by insulin signalling dysfunction and its association with cognitive impairment during aging and Alzheimer's disease: studies in animal models. Diabetes Metab Res Rev 2015; 31:1-13. [PMID: 24464982 DOI: 10.1002/dmrr.2531] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 12/03/2013] [Accepted: 01/15/2014] [Indexed: 01/09/2023]
Abstract
A growing body of animal and epidemiological studies suggest that metabolic diseases such as obesity, insulin resistance, metabolic syndrome and type 2 diabetes mellitus are associated with the development of cognitive impairment, dementia and Alzheimer's disease, particularly in aging. Several lines of evidence suggest that insulin signalling dysfunction produces these metabolic alterations and underlie the development of these neurodegenerative diseases. In this article, we address normal insulin function in the synapse; we review and discuss the physiopathological hallmarks of synaptic insulin signalling dysfunction associated with metabolic alterations. Additionally, we describe and review the major animal models of obesity, insulin resistance, metabolic syndrome and type 2 diabetes mellitus. The comprehensive knowledge of the molecular mechanisms behind the association of metabolic alterations and cognitive impairment could facilitate the early detection of neurodegenerative diseases in patients with metabolic alterations, with treatment that focus on neuroprotection. It could also help in the development of metabolic-based therapies and drugs for using in dementia and Alzheimer's disease patients to alleviate their symptoms in a more efficient and comprehensive way.
Collapse
Affiliation(s)
- Erika Calvo-Ochoa
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, DF, Mexico
| | | |
Collapse
|
385
|
Brouwer-Brolsma EM, de Groot LCPGM. Vitamin D and cognition in older adults: an update of recent findings. Curr Opin Clin Nutr Metab Care 2015; 18:11-6. [PMID: 25225898 DOI: 10.1097/mco.0000000000000114] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Ageing is a generally known risk factor for cognitive decline and dementia. Underlying mechanisms are expected to be multifactorial, but the exact causes are still elusive. This article reviews the potential role of vitamin D in brain function by presenting an overview of recently published mechanistic, rodent as well as human studies. RECENT FINDINGS There is emerging evidence that suggests a beneficial role for vitamin D in brain physiology, for instance by the promotion of neurotransmission, neurogenesis, synaptogenesis, amyloid clearance and the prevention of neuronal death. In addition, several observational studies have shown associations between higher serum vitamin D concentrations and better cognitive performance. To date, imaging studies and randomized controlled trials are scarce, but these studies are expected to fulfil a crucial role towards a better understanding on vitamin D-mediated brain processes in the future. SUMMARY Despite accumulating evidence supporting a role of vitamin D in brain function, only a handful of human trials have been performed. Consequently, the question whether the association between vitamin D, cognitive decline and dementia is causal cannot be sufficiently answered yet.
Collapse
|
386
|
Kealy J, Bennett R, Lowry JP. Real-time effects of insulin-induced hypoglycaemia on hippocampal glucose and oxygen. Brain Res 2014; 1598:76-87. [PMID: 25511995 DOI: 10.1016/j.brainres.2014.12.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 12/01/2014] [Accepted: 12/05/2014] [Indexed: 01/07/2023]
Abstract
The hippocampus plays a vital role in learning and memory and is susceptible to damage following hypoglycaemic shock. The effect of an acute administration of insulin on hippocampal function has been described in terms of behavioural deficits but its effect on hippocampal oxygen and glucose is unclear. Glucose oxidase biosensors (detecting glucose) and carbon paste electrodes (detecting oxygen) were implanted into the hippocampus of Sprague Dawley rats. Animals were allowed to recover and real-time recordings were made in order to determine the effects of fasting, insulin administration (15 U/kg; i.p.) and reintroduction of food on hippocampal oxygen and glucose. Fasting caused a significant decrease in hippocampal glucose over the course of 24h. Insulin administration produced a significant decrease in hippocampal glucose along with a significant increase in hippocampal oxygen. Finally, the reintroduction of food resulted in glucose levels significantly increasing along with a transient but significant increase in oxygen levels. The findings presented here suggest that even a single acute period of hypoglycaemia may substantially disrupt hippocampal oxygen and glucose and therefore affect hippocampal function.
Collapse
Affiliation(s)
- John Kealy
- Maynooth University Department of Chemistry, Maynooth University, Maynooth, Co. Kildare, Ireland.
| | - Rachel Bennett
- Maynooth University Department of Chemistry, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - John P Lowry
- Maynooth University Department of Chemistry, Maynooth University, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
387
|
Werner H, LeRoith D. Insulin and insulin-like growth factor receptors in the brain: physiological and pathological aspects. Eur Neuropsychopharmacol 2014; 24:1947-53. [PMID: 24529663 DOI: 10.1016/j.euroneuro.2014.01.020] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 01/23/2014] [Indexed: 12/31/2022]
Abstract
The involvement of insulin, the insulin-like growth factors (IGF1, IGF2) and their receptors in central nervous system development and function has been the focus of scientific interest for more than 30 years. The insulin-like peptides, both locally-produced proteins as well as those transported from the circulation into the brain via the blood-brain barrier, are involved in a myriad of biological activities. These actions include, among others, neuronal survival, neurogenes, angiogenesis, excitatory and inhibitory neurotransmission, regulation of food intake, and cognition. In recent years, a linkage between brain insulin/IGF1 and certain neuropathologies has been identified. Epidemiological studies have demonstrated a correlation between diabetes (mainly type 2) and Alzheimer׳s disease. In addition, an aberrant decline in IGF1 values was suggested to play a role in the development of Alzheimer׳s disease. The present review focuses on the expression and function of insulin, IGFs and their receptors in the brain in physiological and pathological conditions.
Collapse
Affiliation(s)
- Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Derek LeRoith
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Research Institute at Rambam (LHCRIR), Rambam-Health Care Campus, P.O. Box 9602, Haifa 31096, Israel.
| |
Collapse
|
388
|
Vandal M, White PJ, Tremblay C, St-Amour I, Chevrier G, Emond V, Lefrançois D, Virgili J, Planel E, Giguere Y, Marette A, Calon F. Insulin reverses the high-fat diet-induced increase in brain Aβ and improves memory in an animal model of Alzheimer disease. Diabetes 2014; 63:4291-301. [PMID: 25008180 DOI: 10.2337/db14-0375] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Defects in insulin production and signaling are suspected to share a key role in diabetes and Alzheimer disease (AD), two age-related pathologies. In this study, we investigated the interrelation between AD and diabetes using a high-fat diet (HFD) in a mouse model of genetically induced AD-like neuropathology (3xTg-AD). We first observed that cerebral expression of human AD transgenes led to peripheral glucose intolerance, associated with pancreatic human Aβ accumulation. High-fat diet enhanced glucose intolerance, brain soluble Aβ, and memory impairment in 3xTg-AD mice. Strikingly, a single insulin injection reversed the deleterious effects of HFD on memory and soluble Aβ levels, partly through changes in Aβ production and/or clearance. Our results are consistent with the development of a vicious cycle between AD and diabetes, potentiating both peripheral metabolic disorders and AD neuropathology. The capacity of insulin to rapidly break the deleterious effects of this cycle on soluble Aβ concentrations and memory has important therapeutic implications.
Collapse
Affiliation(s)
- Milene Vandal
- Faculté de Pharmacie, Université Laval, Québec, Canada Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l'Université Laval, Québec, Canada Institut des Nutraceutiques et des Aliments Fonctionnels, Université Laval, Québec, Canada
| | - Phillip J White
- Institut des Nutraceutiques et des Aliments Fonctionnels, Université Laval, Québec, Canada Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Durham, NC Département de Medicine, Axe de Cardiologie, Faculté de Médicine de l'Université Laval, Québec, Canada Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Canada
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l'Université Laval, Québec, Canada
| | - Isabelle St-Amour
- Faculté de Pharmacie, Université Laval, Québec, Canada Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l'Université Laval, Québec, Canada Département de Recherche et Développement, Héma-Québec, Québec, Canada
| | - Geneviève Chevrier
- Institut des Nutraceutiques et des Aliments Fonctionnels, Université Laval, Québec, Canada Département de Medicine, Axe de Cardiologie, Faculté de Médicine de l'Université Laval, Québec, Canada
| | - Vincent Emond
- Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l'Université Laval, Québec, Canada
| | | | | | - Emmanuel Planel
- Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l'Université Laval, Québec, Canada
| | - Yves Giguere
- Faculté de Médicine, Université Laval, Québec, Canada Centre de Recherche du Centre Hospitalier de l'Université de Québec, Hôpital Saint-François d'Assise, Québec, Canada
| | - Andre Marette
- Institut des Nutraceutiques et des Aliments Fonctionnels, Université Laval, Québec, Canada Département de Medicine, Axe de Cardiologie, Faculté de Médicine de l'Université Laval, Québec, Canada Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Canada
| | - Frederic Calon
- Faculté de Pharmacie, Université Laval, Québec, Canada Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l'Université Laval, Québec, Canada Institut des Nutraceutiques et des Aliments Fonctionnels, Université Laval, Québec, Canada
| |
Collapse
|
389
|
Smith GC, McEwen H, Steinberg JD, Shepherd PR. The activation of the Akt/PKB signalling pathway in the brains of clozapine-exposed rats is linked to hyperinsulinemia and not a direct drug effect. Psychopharmacology (Berl) 2014; 231:4553-60. [PMID: 24800899 DOI: 10.1007/s00213-014-3608-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 04/21/2014] [Indexed: 01/01/2023]
Abstract
The second generation antipsychotic drug clozapine is a much more effective therapy for schizophrenia than first generation compounds, but the reasons for this are poorly understood. We have previously shown that one distinguishing feature of clozapine is its ability to raise glucagon levels in animal models and thus causes prolonged hyperinsulinemia without inducing hypoglycaemia. Previous studies have provided evidence that defects in Akt/PKB and GSK3 signalling can contribute to development of psychiatric diseases. Clozapine is known to activate Akt/PKB in the brain, and some studies have indicated that this is due to a direct effect of the drug on the neurons. However, we provide strong evidence that elevated insulin levels induced by clozapine are in fact the real cause of the drug's effects on Akt/PKB and GSK3 in the brain. This suggests that the elevated levels of insulin induced by clozapine may contribute to this drug's therapeutic efficacy.
Collapse
Affiliation(s)
- G C Smith
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand,
| | | | | | | |
Collapse
|
390
|
Wayhs CAY, Mescka CP, Guerreiro G, Moraes TB, Jacques CED, Rosa AP, Ferri MK, Nin MS, Dutra-Filho CS, Barros HMT, Vargas CR. Diabetic encephalopathy-related depression: experimental evidence that insulin and clonazepam restore antioxidant status in rat brain. Cell Biochem Funct 2014; 32:711-9. [PMID: 25431174 DOI: 10.1002/cbf.3076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 10/18/2014] [Indexed: 01/22/2023]
Abstract
There is increasing evidence suggesting that oxidative stress plays an important role in the development of many chronic and degenerative conditions such as diabetic encephalopathy and depression. Considering that diabetic rats and mice present higher depressive-like behaviour when submitted to the forced swimming test and that treatment with insulin and/or clonazepam is able to reverse the behavioural changes of the diabetic rats, the present work investigated the antioxidant status, specifically total antioxidant reactivity and antioxidant potential of insulin and clonazepam, as well as the effect of this drugs upon protein oxidative damage and reactive species formation in cortex, hippocampus and striatum from diabetic rats submitted to forced swimming test. It was verified that longer immobility time in diabetic rats and insulin plus clonazepam treatment reversed this depressive-like behaviour. Moreover, data obtained in this study allowed to demonstrate through different parameters such as protein carbonyl content, 2'7'-dichlorofluorescein oxidation, catalase, superoxide dismutase, glutathione peroxidase assay, total radical-trapping antioxidant potential and total antioxidant reactivity that there is oxidative stress in cortex, hippocampus and striatum from diabetic rats under depressive-like behaviour and highlight the insulin and/or clonazepam effect in these different brain areas, restoring antioxidant status and protein damage.
Collapse
Affiliation(s)
- Carlos Alberto Yasin Wayhs
- Programa de Pós-Graduação em Ciências Farmacêuticas, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil; Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
391
|
Talbot K. Brain insulin resistance in Alzheimer's disease and its potential treatment with GLP-1 analogs. Neurodegener Dis Manag 2014; 4:31-40. [PMID: 24640977 DOI: 10.2217/nmt.13.73] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The prevalence of Alzheimer's disease is increasing rapidly in the absence of truly effective therapies. A promising strategy for developing such therapies is the treatment of brain insulin resistance, a common and early feature of Alzheimer's disease, closely tied to cognitive decline and capable of promoting many biological abnormalities in the disorder. The proximal cause of brain insulin resistance appears to be neuronal elevation in the serine phosphorylation of IRS-1, most likely due to amyloid-β-triggered microglial release of proinflammatory cytokines. Preclinically, the first line of defense is behavior-lowering peripheral insulin resistance (e.g., physical exercise and a Mediterranean diet supplemented with foods rich in flavonoids, curcumin and ω-3 fatty acids). More potent remediation is required, however, at clinical stages. Fortunately, the US FDA-approved antidiabetics exenatide (Byetta; Amylin Pharmaceuticals, Inc., CA, USA) and liraglutide (Victoza; Novo Nordisk A/S, Bagsvaerd, Denmark) are showing much promise in reducing Alzheimer's disease pathology and in restoring normal brain insulin responsiveness and cognitive function.
Collapse
Affiliation(s)
- Konrad Talbot
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
392
|
Therapeutic effect of vagus nerve stimulation on depressive-like behavior, hyperglycemia and insulin receptor expression in Zucker fatty rats. PLoS One 2014; 9:e112066. [PMID: 25365428 PMCID: PMC4218831 DOI: 10.1371/journal.pone.0112066] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 10/11/2014] [Indexed: 01/24/2023] Open
Abstract
Depression and type 2 diabetes (T2D) are common comorbid diseases and highly prevalent in the clinical setting with an unclarified mechanism. Zucker diabetic fatty (ZDF, fa/fa) rats natively develop T2D with hyperglycemia and hyperinsulinemia. Here we studied whether ZDF rats also innately develop depression, what a correlation is between depression and T2D, whether insulin receptor (IR) expression is involved in, and whether transcutaneous auricular vagus nerve stimulation (taVNS) would be beneficial in amelioration of the comorbidity. Six week old male ZDF and Zucker lean (ZL, fa/+) littermates were randomly divided into naïve (ZDF, n = 6; ZL, n = 7) and taVNS (ZDF-taVNS, n = 8; ZL-taVNS, n = 6) groups. Once daily 30 min-taVNS sessions were administrated under anesthesia for 34 consecutive days in taVNS groups. Blood glucose levels were tested weekly, and plasma glycosylated hemoglobin (HbAlc) level and immobility time in forced swimming test were determined on day 35 in all groups. The expression of insulin receptor (IR) in various tissues was also detected by immunostaining and Western blot. We found that naïve ZDF rats developed hyperglycemia steadily. These ZDF rats showed a strong positive correlation between longer immobility time and higher plasma HbAlC level. Long term taVNS treatment simultaneously prevented the development of depression-like behavior and progression of hyperglycemia in ZDF rats. The expression of IR in various tissues of naïve ZDF rats is lower than in naïve ZL and long-term taVNS treated ZDF rats. Collectively, our results indicate that in ZDF rats, i) depression and T2D develop simultaneously, ii) immobility time and HbAlc concentrations are highly and positively correlated, iii) a low expression of IR may be involved in the comorbidity of depression and T2D, and iv) taVNS is antidiabetic and antidepressive possibly through IR expression upregulation.
Collapse
|
393
|
Campos-Bedolla P, Walter FR, Veszelka S, Deli MA. Role of the Blood–Brain Barrier in the Nutrition of the Central Nervous System. Arch Med Res 2014; 45:610-38. [DOI: 10.1016/j.arcmed.2014.11.018] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 11/24/2014] [Indexed: 12/22/2022]
|
394
|
Changes in Brain 14-3-3 Proteins in Response to Insulin Resistance Induced by a High Palatable Diet. Mol Neurobiol 2014; 52:710-8. [PMID: 25280668 DOI: 10.1007/s12035-014-8905-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 09/24/2014] [Indexed: 01/16/2023]
Abstract
The 14-3-3 protein family takes part in a wide range of cellular processes and is expressed in all eukaryotic organisms. In mammals, seven isoforms (β, ε, η, γ, τ, ζ, and σ) have been identified. 14-3-3 proteins are suggested to modulate the insulin-signaling cascade in the brain. The aim of this study was to investigate whether insulin resistance state induced by high palatable diet modulates expression of the 14-3-3 proteins in brain. Wistar male rats (n = 8) were divided into two experimental groups: insulin resistant (IR), induced by high palatable diet, and control (CO) group. Biochemical parameters (glucose tolerance test and plasma lipid profile) were evaluated after 130 days. Brain structures (cortex and hippocampus) were dissected for evaluation of messenger RNA (mRNA) and protein levels of different 14-3-3 proteins. Statistical analyses included Student t test and Pearson correlation. Significant decrease was observed in Ywhah and in Ywahq mRNA levels in the cortex of IR group, while no changes were observed in the hippocampus. Significant increase of θ isoform was observed in hippocampus IR group by immunodetection, while no differences were detected in the remaining isoforms. Inverse correlation was observed between blood glucose levels in cortex IR group and both Ywhah and Ywhaq mRNA levels. Protein levels of Creb and phosphatidylinositide 3-kinases (PI3K) showed to be increased in the hippocampus. These alterations may be due to a compensatory effect of impaired insulin signaling. We demonstrated differential expression of 14-3-3 isoforms throughout brain regions of rats with IR. As a whole, our results indicate that brain 14-3-3 levels are influenced by different diets.
Collapse
|
395
|
Williams DB. Inhibitory effects of insulin on GABAAcurrents modulated by the GABAAalpha subunit. J Recept Signal Transduct Res 2014; 35:516-22. [DOI: 10.3109/10799893.2014.960935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
396
|
Murray S, Tulloch A, Gold MS, Avena NM. Hormonal and neural mechanisms of food reward, eating behaviour and obesity. Nat Rev Endocrinol 2014; 10:540-52. [PMID: 24958311 DOI: 10.1038/nrendo.2014.91] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With rising rates of obesity, research continues to explore the contributions of homeostatic and hedonic mechanisms related to eating behaviour. In this Review, we synthesize the existing information on select biological mechanisms associated with reward-related food intake, dealing primarily with consumption of highly palatable foods. In addition to their established functions in normal feeding, three primary peripheral hormones (leptin, ghrelin and insulin) play important parts in food reward. Studies in laboratory animals and humans also show relationships between hyperphagia or obesity and neural pathways involved in reward. These findings have prompted questions regarding the possibility of addictive-like aspects in food consumption. Further exploration of this topic may help to explain aberrant eating patterns, such as binge eating, and provide insight into the current rates of overweight and obesity.
Collapse
Affiliation(s)
- Susan Murray
- New York Obesity Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, P&S Box 30 DOM/NYORC, 630 West 168th Street, New York, NY 10032-3702, USA
| | - Alastair Tulloch
- New York Obesity Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, P&S Box 30 DOM/NYORC, 630 West 168th Street, New York, NY 10032-3702, USA
| | - Mark S Gold
- Department of Psychiatry, College of Medicine, University of Florida, McKnight Brain Institute, 1149 SW Newell Drive, L4-100, Gainesville, FL 32610, USA
| | - Nicole M Avena
- New York Obesity Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, P&S Box 30 DOM/NYORC, 630 West 168th Street, New York, NY 10032-3702, USA
| |
Collapse
|
397
|
Lindén J, Lensu S, Pohjanvirta R. Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on hormones of energy balance in a TCDD-sensitive and a TCDD-resistant rat strain. Int J Mol Sci 2014; 15:13938-66. [PMID: 25119860 PMCID: PMC4159833 DOI: 10.3390/ijms150813938] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 07/24/2014] [Accepted: 07/29/2014] [Indexed: 01/16/2023] Open
Abstract
One of the hallmarks of the acute toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a drastically reduced feed intake by an unknown mechanism. To further elucidate this wasting syndrome, we followed the effects of a single large dose (100 μg/kg) of TCDD on the serum levels of several energy balance-influencing hormones, clinical chemistry variables, and hepatic aryl hydrocarbon receptor (AHR) expression in two rat strains that differ widely in their TCDD sensitivities, for up to 10 days. TCDD affected most of the analytes in sensitive Long-Evans rats, while there were few alterations in the resistant Han/Wistar strain. However, analyses of feed-restricted unexposed Long-Evans rats indicated several of the perturbations to be secondary to energy deficiency. Notable increases in ghrelin and glucagon occurred in TCDD-treated Long-Evans rats alone, which links these hormones to the wasting syndrome. The newly found energy balance regulators, insulin-like growth factor 1 and fibroblast growth factor 21 (FGF-21), appeared to function in concert in body weight loss-induced metabolic state, and FGF-21 was putatively linked to increased lipolysis induced by TCDD. Finally, we demonstrate a reverse set of changes in the AHR protein and mRNA response to TCDD and feed restriction, suggesting that AHR might function also as a physiological regulator, possibly involved in the maintenance of energy balance.
Collapse
Affiliation(s)
- Jere Lindén
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 66, FI-00014 Helsinki, Finland.
| | - Sanna Lensu
- Department of Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35, FI-40014 Jyväskylä, Finland.
| | - Raimo Pohjanvirta
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 66, FI-00014 Helsinki, Finland.
| |
Collapse
|
398
|
Austin AW, Gordon JL, Lavoie KL, Arsenault A, Dasgupta K, Bacon SL. Differential association of insulin resistance with cognitive and somatic symptoms of depression. Diabet Med 2014; 31:994-1000. [PMID: 24754892 DOI: 10.1111/dme.12465] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 01/09/2014] [Accepted: 03/28/2014] [Indexed: 11/29/2022]
Abstract
AIM To examine the associations of depressive symptoms with insulin resistance, evaluating somatic and cognitive depressive symptoms separately. METHODS A total of 328 individuals (mean age 60 years) referred for exercise stress testing, taking part in the Mechanisms and Outcomes of Silent Myocardial Ischemia study, completed the Beck Depression Inventory II. A fasting venous blood sample was collected for assessments of insulin and glucose level; the HOMA-IR (homeostatic model assessment of insulin resistance) was calculated. In principal component analysis, Beck Depression Inventory II items were forced to load onto two components (somatic and cognitive depressive symptoms). Adjusting for age, sex, BMI, medication use, smoking, physical activity, diabetes and cardiovascular disease, general linear model analyses were conducted to examine the associations between the components and log HOMA-IR . RESULTS Principal component analysis showed that nine items loaded onto a cognitive depressive symptoms component and 10 items loaded onto a somatic depressive symptoms component. When examined separately, both components were significantly associated with log HOMA-IR however, when including both components simultaneously in the model, only somatic depressive symptoms remained significantly associated with log HOMA-IR. Back-transformed, a one-unit change in somatic depressive symptoms was associated with a 1.07 (95% CI 1.002, 1.14) change in HOMA-IR and a one-unit change in cognitive depressive symptoms was associated with a 1.03 (95% CI 0.97, 1.14) change in HOMA-IR. CONCLUSION Somatic depressive symptoms seem to be more strongly associated with insulin resistance than do cognitive depressive symptoms. Monitoring somatic depressive symptoms may be more appropriate than monitoring cognitive depressive symptoms among depressed individuals with high insulin resistance.
Collapse
Affiliation(s)
- A W Austin
- Montreal Behavioural Medicine Centre, Hopital du Sacre-Coeur de Montreal, Montreal, Quebec, Canada; Department of Exercise Science, Concordia University, Montreal, Quebec, Canada; Research Centre, Hopital du Sacre-Coeur de Montreal, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
399
|
Cukierman-Yaffe T, Bosch J, Diaz R, Dyal L, Hancu N, Hildebrandt P, Lanas F, Lewis BS, Marre M, Yale JF, Yusuf S, Gerstein HC. Effects of basal insulin glargine and omega-3 fatty acid on cognitive decline and probable cognitive impairment in people with dysglycaemia: a substudy of the ORIGIN trial. Lancet Diabetes Endocrinol 2014; 2:562-72. [PMID: 24898834 DOI: 10.1016/s2213-8587(14)70062-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Diabetes and non-diabetic dysglycaemia are risk factors for accelerated cognitive decline. In this planned substudy of the Outcome Reduction with Initial Glargine Intervention (ORIGIN) trial, we assessed whether normalising glucose with insulin glargine or administering omega-3 fatty acids in this population may slow this process or affect the development of cognitive impairment. METHODS The ORIGIN trial recruited participants older than 50 years with dysglycaemia who were taking either no or one oral glucose-lowering drug, who had additional risk factors for cardiovascular events, whose HbA1c was less than 9%, and who were not taking insulin. Participants were recruited from 573 sites in 40 countries. Participants were randomly assigned to either titrated basal insulin glargine targeting a fasting plasma glucose concentration of 5.3 mmol/L or lower or standard care and to either omega-3 fatty acid (1 g) or placebo by a factorial design. Outcome adjudicators and data analysts were masked to treatment allocation. Cognitive function was assessed by the Mini-Mental State Examination (MMS) and Digit Symbol Substitution (DSS). The effect of insulin glargine or omega-3 fatty * acid on cognitive function over time, the annualised change in test scores, and the development of probable cognitive impairment were measured. All analyses were restricted to those participants who had a cognitive measurement at both baseline and at least one follow-up visit. The ORIGIN trial is registered with ClinicalTrials.gov, NCT00069784. FINDINGS Participants were randomly assigned between Sept 1, 2003, and Dec 15, 2005. MMSE and DSS were assessed in 11,685 and 3392 ORIGIN participants (mean age 63.4 years [SD 7.7]), who were followed up for a median of 6.2 years (IQR 5.8-6.7). There was no difference in the rate of change of cognitive test scores between the insulin glargine and standard care groups (for the MMSE 0.0046, 95% CI -0.0132 to 0.0224, p=0.39; and for the DSS -0.0362, -0.2180 to 0.1455, p=0.34) or between the omega-3 fatty acid and placebo groups (for the MMSE 0.0013, 95% CI -0.0165 to 0.0191, p=0.21; and for the DSS -0.0605, -0.2422 to 0.1212, p=0.72). Similarly, the incidence of probable cognitive impairment did not differ between the insulin glargine and standard care groups (p=0.065) or the omega-3 fatty acid and placebo groups (p=0.070). In a subgroup analysis, allocation to insulin glargine versus standard care seemed to reduce the decline in the MMSE (but not the DSS) in participants with dysglycaemia but without evidence of diabetes (pinteraction=0.024). INTERPRETATION In this relatively young cohort of people with dysglycaemia, insulin mediated normoglycaemia and omega-3 fatty acid for over 6 years had a neutral effect on the rate of cognitive decline and on incident cognitive impairment. Future studies should assess the effect of these interventions in an older cohort or the effect of other glucometabolic interventions on cognitive decline. FUNDING Sanofi.
Collapse
Affiliation(s)
- Tali Cukierman-Yaffe
- Endocrinology Institute, Gertner Institute Sheba Medical Center, Ramat Gan, Israel; Epidemiology Department, Tel-Aviv University, Tel-Aviv, Israel; Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada.
| | - Jackie Bosch
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada; School of Rehabilitation Science, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
| | - Rafael Diaz
- Estudios Clínicos Latino América, Rosario, Argentina
| | - Leanne Dyal
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
| | - Nicolae Hancu
- Diabetes, Nutrition and Metabolic Diseases Department, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | | - Fernando Lanas
- Department of Internal Medicine, Universidad de La Frontera, Temuco, Chile
| | - Basil S Lewis
- Cardiovascular Clinical Research Institute, Lady Davis Carmel Medical Center and Technion-IIT, Haifa, Israel
| | - Michel Marre
- Service d'Endocrinologie Diabétologie Nutrition, Groupe Hospitalier Bichat, Claude Bernard, Paris, France; Unité INSERM U1138 équipe 2, "Pathophysiology and therapeutics of vascular and renal diseases related to diabetes and nutrition", Centre de Recherches des Cordeliers, Paris, France
| | - Jean-Francois Yale
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Salim Yusuf
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
| | - Hertzel C Gerstein
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
| | | |
Collapse
|
400
|
Mansur RB, Cha DS, Woldeyohannes HO, Soczynska JK, Zugman A, Brietzke E, McIntyre RS. Diabetes mellitus and disturbances in brain connectivity: a bidirectional relationship? Neuromolecular Med 2014; 16:658-68. [PMID: 24974228 DOI: 10.1007/s12017-014-8316-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 06/19/2014] [Indexed: 12/26/2022]
Abstract
Diabetes mellitus (DM) is associated with deficits across multiple cognitive domains. The observed impairments in cognitive function are hypothesized to be subserved by alterations in brain structure and function. Several lines of evidence indicate that alterations in glial integrity and function, as well as abnormal synchrony within brain circuits and associated networks, are observed in adults with DM. Microangiopathy and alterations in insulin homeostasis appear to be principal effector systems, although a unitary explanation subsuming the complex etiopathology of white matter in DM is unavailable. A contemporary model of disease pathophysiology for several mental disorders, including but not limited to mood disorders, posits abnormalities in the synchronization of cellular systems in circuits. The observation that similar abnormalities occur in diabetic populations provides the basis for hypothesizing the convergence of pathoetiological factors. Herein, we propose that abnormal structure, function and chemical composition as well as synchrony within and between circuits is an accompaniment of DM and is shared in common with several mental disorders.
Collapse
Affiliation(s)
- Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit (MDPU), University Health Network, University of Toronto, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada,
| | | | | | | | | | | | | |
Collapse
|