1
|
Todorovic S, Simeunovic V, Prvulovic M, Dakic T, Jevdjovic T, Sokanovic S, Kanazir S, Mladenovic A. Dietary restriction alters insulin signaling pathway in the brain. Biofactors 2024; 50:450-466. [PMID: 37975613 DOI: 10.1002/biof.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/07/2023] [Indexed: 11/19/2023]
Abstract
Insulin is known to be a key hormone in the regulation of peripheral glucose homeostasis, but beyond that, its effects on the brain are now undisputed. Impairments in insulin signaling in the brain, including changes in insulin levels, are thought to contribute significantly to declines in cognitive performance, especially during aging. As one of the most widely studied experimental interventions, dietary restriction (DR) is considered to delay the neurodegenerative processes associated with aging. Recently, however, data began to suggest that the onset and duration of a restrictive diet play a critical role in the putative beneficial outcome. Because the effects of DR on insulin signaling in the brain have been poorly studied, we decided to examine the effects of DR that differed in onset and duration: long-term DR (LTDR), medium-term DR (MTDR), and short-term DR (STDR) on the expression of proteins involved in insulin signaling in the hippocampus of 18- and 24-month-old male Wistar rats. We found that DR-induced changes in insulin levels in the brain may be independent of what happens in the periphery after restricted feeding. Significantly changed insulin content in the hippocampus, together with altered insulin signaling were found under the influence of DR, but the outcome was highly dependent on the onset and duration of DR.
Collapse
Affiliation(s)
- Smilja Todorovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Valentina Simeunovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milica Prvulovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Dakic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Tanja Jevdjovic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Srdjan Sokanovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Selma Kanazir
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Mladenovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
2
|
Hamilton K, Morrow K, Markantoni E, Harvey J. Leptin prevents aberrant targeting of tau to hippocampal synapses via PI 3 kinase driven inhibition of GSK3β. J Neurochem 2023; 167:520-537. [PMID: 37822142 DOI: 10.1111/jnc.15980] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/11/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
Amyloid-β (Aβ) and hyper-phosphorylated tau are key hallmarks of Alzheimer's disease (AD), with an accumulation of both proteins linked to hippocampal synaptic dysfunction. Recent evidence indicates that Aβ drives mis-localisation of tau from axons to synapses, resulting in AMPA receptor (AMPAR) internalisation and impaired excitatory synaptic function. These tau-driven synaptic impairments are thought to underlie the cognitive deficits in AD. Consequently, limiting the synapto-toxic effects of tau may prevent AD-related cognitive deficits. Increasing evidence links leptin dysfunction with higher AD risk, and numerous studies have identified neuroprotective properties of leptin in AD models of Aβ-induced toxicity. However, it is unclear if leptin protects against tau-related synaptic dysfunction. Here we show that Aβ1-42 significantly increases dendritic and synaptic levels of tau and p-tau in hippocampal neurons, and these effects were blocked by leptin. In accordance with GSK-3β being involved in tau phosphorylation, the protective effects of leptin involve PI 3-kinase (PI3K) activation and inhibition of GSK-3β. Aβ1-42 -driven synaptic targeting of tau was associated with the removal of GluA1-containing AMPARs from synapses, which was also inhibited by leptin-driven inhibition of GSK-3β. Direct application of oligomeric tau to hippocampal neurons caused internalisation of GluA1-containing AMPARs and this effect was blocked by prior application of leptin. Similarly, leptin prevented the ability of tau to block induction of activity-dependent long-term potentiation (LTP) at hippocampal SC-CA1 synapses. These findings increase our understanding of the neuroprotective actions of leptin in the early pre-clinical stages of AD and further validate the leptin system as a therapeutic target in AD.
Collapse
Affiliation(s)
- Kirsty Hamilton
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Kate Morrow
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Ermione Markantoni
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Jenni Harvey
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| |
Collapse
|
3
|
Carvalho da Silva AM, Lemos C, Silva HB, Ferreira IL, Tomé AR, Rego AC, Cunha RA. Simultaneous Alteration of the Circadian Variation of Memory, Hippocampal Synaptic Plasticity, and Metabolism in a Triple Transgenic Mouse Model of Alzheimer’s Disease. Front Aging Neurosci 2022; 14:835885. [PMID: 35431906 PMCID: PMC9009366 DOI: 10.3389/fnagi.2022.835885] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/02/2022] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by progressive memory deficits accompanied by synaptic and metabolic deficits, namely of mitochondrial function. AD patients also display a disrupted circadian pattern. Thus, we now compared memory performance, synaptic plasticity, and mitochondria function in 24-week-old non-transgenic (non-Tg) and triple transgenic male mice modeling AD (3xTg-AD) at Zeitgeber 04 (ZT-4, inactive phase) and ZT-16 (active phase). Using the Morris water maze test to minimize the influence of circadian-associated locomotor activity, we observed a circadian variation in hippocampus-dependent learning performance in non-Tg mice, which was impaired in 3xTg-AD mice. 3xTg-AD mice also displayed a lack of circadian variation of their performance in the reversal spatial learning task. Additionally, the amplitude of hippocampal long-term potentiation also exhibited a circadian profile in non-Tg mice, which was not observed in 3xTg-AD mice. Moreover, cerebral cortical synaptosomes of non-Tg mice also displayed a circadian variation of FCCP-stimulated oxygen consumption as well as in mitochondrial calcium retention that were blunted in 3xTg-AD mice. In sum, this multidimensional study shows that the ability to maintain a circadian oscillation in brain behavior, synaptic plasticity, and synaptic mitochondria function are simultaneously impaired in 3xTg-AD mice, highlighting the effects of circadian misalignment in AD.
Collapse
Affiliation(s)
- António M. Carvalho da Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- *Correspondence: António M. Carvalho da Silva,
| | - Cristina Lemos
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Henrique B. Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ildete L. Ferreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Angelo R. Tomé
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - A. Cristina Rego
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- A. Cristina Rego,
| | - Rodrigo A. Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Rodrigo A. Cunha,
| |
Collapse
|
4
|
Irving A, Harvey J. Regulation of hippocampal synaptic function by the metabolic hormone leptin: Implications for health and disease. Prog Lipid Res 2021; 82:101098. [PMID: 33895229 DOI: 10.1016/j.plipres.2021.101098] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
Significant advances have been made in our understanding of the hormone, leptin and its CNS actions in recent years. It is now evident that leptin has a multitude of brain functions, that extend beyond its established role in the hypothalamic control of energy balance. Additional brain regions including the hippocampus are important targets for leptin, with a high density of leptin receptors (LepRs) expressed in specific hippocampal regions and localised to CA1 synapses. Extensive evidence indicates that leptin has pro-cognitive actions, as it rapidly modifies synaptic efficacy at excitatory Schaffer collateral (SC)-CA1 and temporoammonic (TA)-CA1 synapses and enhances performance in hippocampal-dependent memory tasks. There is a functional decline in hippocampal responsiveness to leptin with age, with significant reductions in the modulatory effects of leptin at SC-CA1 and TA-CA1 synapses in aged, compared to adult hippocampus. As leptin has pro-cognitive effects, this decline in leptin sensitivity is likely to have negative consequences for cognitive function during the aging process. Here we review how evaluation of the hippocampal actions of leptin has improved our knowledge of the regulatory brain functions of leptin in health and provided significant insight into the impact of leptin in age-related neurodegenerative disorders linked to cognitive decline.
Collapse
Affiliation(s)
- Andrew Irving
- School of Biomolecular and Biomedical Science, The Conway Institute, University College Dublin, Dublin, Ireland
| | - Jenni Harvey
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom.
| |
Collapse
|
5
|
Hamilton K, Harvey J. Leptin regulation of hippocampal synaptic function in health and disease. VITAMINS AND HORMONES 2021; 115:105-127. [PMID: 33706945 DOI: 10.1016/bs.vh.2020.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
It is widely accepted that the metabolic hormone leptin regulates food intake and body weight via activation of hypothalamic leptin receptors. However, as leptin receptors are also highly expressed in other brain regions, such as the hippocampus, alterations in leptin responsiveness also impacts on key functions of the hippocampus, like learning and memory. Within the hippocampus, high levels of leptin receptors are expressed at excitatory synapses, and in accordance with a synaptic localization, leptin potently regulates synaptic transmission at both Schaffer collateral (SC) and temporoammonic (TA) inputs to CA1 pyramidal neurons. Increasing evidence from cellular and behavioral studies examining leptin action at CA1 synapses support the notion that leptin is a potential cognitive enhancer. However, the capacity of leptin to regulate synaptic efficacy at SC-CA1 and TA-CA1 synapses declines in an age-dependent manner. Moreover, clinical evidence that supports a link between circulating leptin levels and the risk of the age-related neurodegenerative disorder, Alzheimer's disease (AD) is accumulating. Consequently, it has been proposed that the leptin system is a potential therapeutic target in AD, and that boosting the hippocampal actions of leptin may be beneficial in the treatment of AD. Here we review recent progress in our understanding of the neuronal and hippocampal synaptic functions that are regulated by leptin and how alterations in the leptin system influence age-related CNS-related disorders like AD.
Collapse
Affiliation(s)
- Kirsty Hamilton
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Jenni Harvey
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom.
| |
Collapse
|
6
|
Hamilton K, Harvey J. The Neuronal Actions of Leptin and the Implications for Treating Alzheimer's Disease. Pharmaceuticals (Basel) 2021; 14:ph14010052. [PMID: 33440796 PMCID: PMC7827292 DOI: 10.3390/ph14010052] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
It is widely accepted that the endocrine hormone leptin controls food intake and energy homeostasis via activation of leptin receptors expressed on hypothalamic arcuate neurons. The hippocampal formation also displays raised levels of leptin receptor expression and accumulating evidence indicates that leptin has a significant impact on hippocampal synaptic function. Thus, cellular and behavioural studies support a cognitive enhancing role for leptin as excitatory synaptic transmission, synaptic plasticity and glutamate receptor trafficking at hippocampal Schaffer collateral (SC)-CA1 synapses are regulated by leptin, and treatment with leptin enhances performance in hippocampus-dependent memory tasks. Recent studies indicate that hippocampal temporoammonic (TA)-CA1 synapses are also a key target for leptin. The ability of leptin to regulate TA-CA1 synapses has important functional consequences as TA-CA1 synapses are implicated in spatial and episodic memory processes. Moreover, degeneration is initiated in the TA pathway at very early stages of Alzheimer's disease, and recent clinical evidence has revealed links between plasma leptin levels and the incidence of Alzheimer's disease (AD). Additionally, accumulating evidence indicates that leptin has neuroprotective actions in various AD models, whereas dysfunctions in the leptin system accelerate AD pathogenesis. Here, we review the data implicating the leptin system as a potential novel target for AD, and the evidence that boosting the hippocampal actions of leptin may be beneficial.
Collapse
|
7
|
Associations of Objectively-Assessed Physical Activity and Sedentary Time with Hippocampal Gray Matter Volume in Children with Overweight/Obesity. J Clin Med 2020; 9:jcm9041080. [PMID: 32290290 PMCID: PMC7231303 DOI: 10.3390/jcm9041080] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 02/08/2023] Open
Abstract
This study investigated physical activity (PA) and sedentary time (SED) in relation to hippocampal gray matter volume (GMV) in pediatric overweight/obesity. Ninety-three children (10 ± 1 year) were classified as overweight, obesity type I, or type II–III. PA was assessed with non-dominant wrist accelerometers. GMV was acquired by magnetic resonance imaging (MRI). Neither PA nor SED associated with GMV in the hippocampus in the whole sample (p > 0.05). However, we found some evidence of moderation by weight status (p < 0.150). Moderate-to-vigorous PA (MVPA) positively associated with GMV in the right hippocampus in obesity type I (B = 5.62, p = 0.017), which remained when considering SED, light PA, and sleep using compositional data (γ = 375.3, p = 0.04). Compositional models also depicted a negative association of SED relative to the remaining behaviors with GMV in the right hippocampus in overweight (γ = −1838.4, p = 0.038). Reallocating 20 min/day of SED to MVPA was associated with 100 mm3 GMV in the right hippocampus in obesity type I. Multivariate pattern analysis showed a negative-to-positive association pattern between PA of increasing intensity and GMV in the right hippocampus in obesity type II–III. Our findings support that reducing SED and increasing MVPA are associated with greater GMV in the right hippocampus in pediatric overweight/obesity. Further studies should corroborate our findings.
Collapse
|
8
|
Barra R, Morgan C, Sáez-Briones P, Reyes-Parada M, Burgos H, Morales B, Hernández A. Facts and hypotheses about the programming of neuroplastic deficits by prenatal malnutrition. Nutr Rev 2020; 77:65-80. [PMID: 30445479 DOI: 10.1093/nutrit/nuy047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Studies in rats have shown that a decrease in either protein content or total dietary calories results in molecular, structural, and functional changes in the cerebral cortex and hippocampus, among other brain regions, which lead to behavioral disturbances, including learning and memory deficits. The neurobiological bases underlying those effects depend at least in part on fetal programming of the developing brain, which in turn relies on epigenetic regulation of specific genes via stable and heritable modifications of chromatin. Prenatal malnutrition also leads to epigenetic programming of obesity, and obesity on its own can lead to poor cognitive performance in humans and experimental animals, complicating understanding of the factors involved in the fetal programming of neuroplasticity deficits. This review focuses on the role of epigenetic mechanisms involved in prenatal malnutrition-induced brain disturbances, which are apparent at a later postnatal age, through either a direct effect of fetal programming on brain plasticity or an indirect effect on the brain mediated by the postnatal development of obesity.
Collapse
Affiliation(s)
- Rafael Barra
- School of Medicine, Faculty of Medical Sciences, University of Santiago de Chile, Santiago, Chile
| | - Carlos Morgan
- Laboratory of Nutrition and Metabolic Regulation, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Patricio Sáez-Briones
- School of Medicine, Faculty of Medical Sciences, University of Santiago de Chile, Santiago, Chile
| | - Miguel Reyes-Parada
- School of Medicine, Faculty of Medical Sciences, University of Santiago de Chile, Santiago, Chile.,Facultad de Ciencias de la Salud Universidad Autónoma de Chile, Talca, Chile
| | - Héctor Burgos
- Núcleo Disciplinar Psicología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Center of Innovation on Information Technologies for Social Applications (CITIAPS), University of Santiago de Chile, Santiago, Chile
| | - Bernardo Morales
- Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Alejandro Hernández
- Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| |
Collapse
|
9
|
Cortés-Álvarez NY, Vuelvas-Olmos CR, Pinto-González MF, Guzmán-Muñiz J, Gonzalez-Perez O, Moy-López NA. A high-fat diet during pregnancy impairs memory acquisition and increases leptin receptor expression in the hippocampus of rat offspring. Nutr Neurosci 2020; 25:146-158. [DOI: 10.1080/1028415x.2020.1728473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Nadia Yanet Cortés-Álvarez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima, Mexico
- Medical Sciences Program, School of Medicine, University of Colima, Colima, Mexico
| | - César Rubén Vuelvas-Olmos
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima, Mexico
- Medical Sciences Program, School of Medicine, University of Colima, Colima, Mexico
| | - María Fernanda Pinto-González
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima, Mexico
- Medical Sciences Program, School of Medicine, University of Colima, Colima, Mexico
| | - Jorge Guzmán-Muñiz
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima, Mexico
| | - Oscar Gonzalez-Perez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima, Mexico
| | | |
Collapse
|
10
|
Hahm JR, Jo MH, Ullah R, Kim MW, Kim MO. Metabolic Stress Alters Antioxidant Systems, Suppresses the Adiponectin Receptor 1 and Induces Alzheimer's Like Pathology in Mice Brain. Cells 2020; 9:cells9010249. [PMID: 31963819 PMCID: PMC7016950 DOI: 10.3390/cells9010249] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/09/2020] [Accepted: 01/15/2020] [Indexed: 01/02/2023] Open
Abstract
Oxidative stress and insulin resistance play major roles in numerous neurodegenerative diseases, including Alzheimer’s disease (AD). A high-fat diet induces obesity-associated oxidative stress, neuronal insulin resistance, microglial activation, and neuroinflammation, which are considered important risk factors for neurodegeneration. Obesity-related metabolic dysfunction is a risk factor for cognitive decline. The present study aimed to elucidate whether chronic consumption of a high-fat diet (HFD; 24 weeks) can induce insulin resistance, neuroinflammation, and amyloid beta (Aβ) deposition in mouse brains. Male C57BL/6N mice were used for a high-fat diet (HFD)-induced pre-clinical model of obesity. The protein expression levels were examined via Western blot, immunofluorescence, and the behavior analysis was performed using the Morris water maze test. To obtain metabolic parameters, insulin sensitivity and glucose tolerance tests were performed. We found that metabolic perturbations from the chronic consumption of HFD elevated neuronal oxidative stress and insulin resistance through adiponectin receptor (AdipoR1) suppression in HFD-fed mice. Similarly, our in vitro results also indicated that knockdown of AdipoR1 in the embryonic mouse hippocampal cell line mHippoE-14 leads to increased oxidative stress in neurons. In addition, HFD markedly increased neuroinflammatory markers’ glial activation in the cortex and hippocampus regions of HFD mouse brains. More importantly, we observed that AdipoR1 suppression increased the amyloidogenic pathway both in vivo and in vitro. Furthermore, deregulated synaptic proteins and behavioral deficits were observed in the HFD mouse brains. Taken together, our findings suggest that excessive consumption of an HFD has a profound impact on brain function, which involves the acceleration of cognitive impairment due to increased obesity-associated oxidative stress, insulin resistance, and neuroinflammation, which ultimately may cause early onset of Alzheimer’s pathology via the suppression of AdipoR1 signaling in the brain.
Collapse
Affiliation(s)
- Jong Ryeal Hahm
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Gyeongsang National University Hospital and Institute of Health Sciences and Department of Internal Medicine, College of Medicine, Gyeongsang National University, Jinju 52828, Korea;
| | - Myeung Hoon Jo
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (M.H.J.); (R.U.); (M.W.K.)
| | - Rahat Ullah
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (M.H.J.); (R.U.); (M.W.K.)
| | - Min Woo Kim
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (M.H.J.); (R.U.); (M.W.K.)
| | - Myeong Ok Kim
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (M.H.J.); (R.U.); (M.W.K.)
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
11
|
Mechanism of the neuroprotective effect of GLP-1 in a rat model of Parkinson's with pre-existing diabetes. Neurochem Int 2019; 131:104583. [DOI: 10.1016/j.neuint.2019.104583] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/15/2019] [Accepted: 10/18/2019] [Indexed: 01/21/2023]
|
12
|
Rivera O, McHan L, Konadu B, Patel S, Sint Jago S, Talbert ME. A high-fat diet impacts memory and gene expression of the head in mated female Drosophila melanogaster. J Comp Physiol B 2019; 189:179-198. [PMID: 30810797 PMCID: PMC6711602 DOI: 10.1007/s00360-019-01209-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/12/2019] [Accepted: 02/17/2019] [Indexed: 12/25/2022]
Abstract
Obesity predisposes humans to a range of life-threatening comorbidities, including type 2 diabetes and cardiovascular disease. Obesity also aggravates neural pathologies, such as Alzheimer's disease, but this class of comorbidity is less understood. When Drosophila melanogaster (flies) are exposed to high-fat diet (HFD) by supplementing a standard medium with coconut oil, they adopt an obese phenotype of decreased lifespan, increased triglyceride storage, and hindered climbing ability. The latter development has been previously regarded as a potential indicator of neurological decline in fly models of neurodegenerative disease. Our objective was to establish the obesity phenotype in Drosophila and identify a potential correlation, if any, between obesity and neurological decline through behavioral assays and gene expression microarray. We found that mated female w1118 flies exposed to HFD maintained an obese phenotype throughout adult life starting at 7 days, evidenced by increased triglyceride stores, diminished life span, and impeded climbing ability. While climbing ability worsened cumulatively between 7 and 14 days of exposure to HFD, there was no corresponding alteration in triglyceride content. Microarray analysis of the mated female w1118 fly head revealed HFD-induced changes in expression of genes with functions in memory, metabolism, olfaction, mitosis, cell signaling, and motor function. Meanwhile, an Aversive Phototaxis Suppression assay in mated female flies indicated reduced ability to recall an entrained memory 6 h after training. Overall, our results support the suitability of mated female flies for examining connections between diet-induced obesity and nervous or neurobehavioral pathology, and provide many directions for further investigation.
Collapse
Affiliation(s)
- Osvaldo Rivera
- Program in Biology, School of Sciences, University of Louisiana at Monroe, 700 University Avenue, Monroe, LA, 71209, USA
| | - Lara McHan
- Program in Biology, School of Sciences, University of Louisiana at Monroe, 700 University Avenue, Monroe, LA, 71209, USA
| | - Bridget Konadu
- Program in Biology, School of Sciences, University of Louisiana at Monroe, 700 University Avenue, Monroe, LA, 71209, USA
| | - Sumitkumar Patel
- Program in Biology, School of Sciences, University of Louisiana at Monroe, 700 University Avenue, Monroe, LA, 71209, USA
| | - Silvienne Sint Jago
- Program in Biology, School of Sciences, University of Louisiana at Monroe, 700 University Avenue, Monroe, LA, 71209, USA
| | - Matthew E Talbert
- Program in Biology, School of Sciences, University of Louisiana at Monroe, 700 University Avenue, Monroe, LA, 71209, USA.
| |
Collapse
|
13
|
Lewis AR, Singh S, Youssef FF. Cafeteria-diet induced obesity results in impaired cognitive functioning in a rodent model. Heliyon 2019; 5:e01412. [PMID: 30976688 PMCID: PMC6441847 DOI: 10.1016/j.heliyon.2019.e01412] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 02/25/2019] [Accepted: 03/20/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES This study seeks to characterize the progressive course of physiological and behavioural outcomes in rodents following excessive caloric intake through the chronic consumption of a highly palatable diet, the cafeteria (CAF) diet. METHODS Male Sprague Dawley rats were maintained on either CAF or chow (CON) diets for 20 weeks. Metabolic and physiological parameters were monitored throughout the feeding period. From week 18, rats were subjected to behavioural testing, which included the Morris water maze (MWM) and novel object recognition (NOR) tasks. RESULTS CAF rats consistently showed higher food intakes and consumed six times the energy of chow-fed rats, being significantly heavier by week 5. CAF rats further exhibited greater abdominal widths, fat pads, and larger fatty livers, as well as compromised glucose tolerance. Hyperinsulinemia and dyslipidaemia with elevated serum cholesterol and triglyceride levels and reduced HDL cholesterol were also evident along with a pro-inflammatory profile in the CAF rats. Cognitive decline in CAF rats manifested as a decline in long-term retention memory in the MWM. Further, CAF rats exhibited deficits in recognition memory as they spent less time exploring the novel object than chow-fed rats in the NOR task. DISCUSSION This model of obesity is a robust paradigm for producing an obese animal phenotype that closely mimics the evolution of human obesity, complete with metabolic dysfunctions that are indicative of pre-diabetes. Additionally, chronic CAF-diet induced obesity promotes cognitive impairments in hippocampal-dependent reference and working memory.
Collapse
Affiliation(s)
- Aneisha R. Lewis
- Department of Pre-Clinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Shamjeet Singh
- School of Pharmacy, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Farid. F. Youssef
- Department of Pre-Clinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| |
Collapse
|
14
|
McGregor G, Harvey J. Regulation of Hippocampal Synaptic Function by the Metabolic Hormone, Leptin: Implications for Health and Neurodegenerative Disease. Front Cell Neurosci 2018; 12:340. [PMID: 30386207 PMCID: PMC6198461 DOI: 10.3389/fncel.2018.00340] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/13/2018] [Indexed: 11/13/2022] Open
Abstract
The role of the endocrine hormone leptin in controlling energy homeostasis in the hypothalamus are well documented. However the CNS targets for leptin are not restricted to the hypothalamus as a high density of leptin receptors are also expressed in several parts of the brain involved in higher cognitive functions including the hippocampus. Numerous studies have identified that in the hippocampus, leptin has cognitive enhancing actions as exogenous application of this hormone facilitates hippocampal-dependent learning and memory, whereas lack or insensitivity to leptin results in significant memory deficits. Leptin also markedly influences some of the main cellular changes that are involved in learning and memory including NMDA-receptor dependent synaptic plasticity and glutamate receptor trafficking. Like other metabolic hormones, there is a significant decline in neuronal sensitivity to leptin during the ageing process. Indeed, the capacity of leptin to modulate the functioning of hippocampal synapses is substantially reduced in aged compared to adult tissue. Clinical studies have also identified an association between circulating leptin levels and the risk of certain neurodegenerative disorders such as Alzheimer’s disease (AD). In view of this, targeting leptin and/or its receptor/signaling mechanisms may be an innovative approach for developing therapies to treat AD. In support of this, accumulating evidence indicates that leptin has cognitive enhancing and neuroprotective actions in various models of AD. Here we assess recent evidence that supports an important regulatory role for leptin at hippocampal CA1 synapses, and we discuss how age-related alterations in this hormonal system influences neurodegenerative disease.
Collapse
Affiliation(s)
- Gemma McGregor
- Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Jenni Harvey
- Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
15
|
Role of gamma-amino-butyric acid in the dorsal anterior cingulate in age-associated changes in cognition. Neuropsychopharmacology 2018; 43:2285-2291. [PMID: 30050047 PMCID: PMC6135795 DOI: 10.1038/s41386-018-0134-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/13/2018] [Accepted: 06/18/2018] [Indexed: 11/08/2022]
Abstract
GABAergic mechanisms have been shown to contribute to cognitive aging in animal models, but there is currently limited in vivo evidence to support this relationship in humans. It is also unclear whether aging is associated with changes in GABA levels measured with proton magnetic resonance spectroscopy (MRS). Spectral-editing MRS at 3 T was used to measure GABA in the dorsal anterior cingulate cortex (dACC) for a large sample of healthy volunteers (N = 229) aged 18-55. In a subset of 171 participants, age effects on several cognitive tasks were studied. We formally tested whether the MRS measures mediated the relationship between age and cognition. Robust associations of age with performance were found for the Wisconsin Card Sorting Test ([WCST], p < 0.0001). Age was also significantly associated with declining levels of GABA in the dACC (p < 0.001), and GABA levels significantly predicted WCST performance (p < 0.0004). Mediation analysis revealed that GABA in the dACC mediated the effect of age on WCST performance (p < 0.01). Other metabolites were similarly associated with age, but only GABA and creatine levels were significantly associated with WCST performance. No association with age or cognitive performance was found in a frontal white matter control region in a subset of participants. The association of GABA with WCST performance was not related to the amount of brain atrophy associated with aging as measured by the proportion of CSF, gray, and white matter in the MRS voxel. These results implicate GABAergic and possibly energetic metabolism in the dACC as mechanisms of age effects in executive function.
Collapse
|
16
|
RNA-Sequencing of Drosophila melanogaster Head Tissue on High-Sugar and High-Fat Diets. G3-GENES GENOMES GENETICS 2018; 8:279-290. [PMID: 29141990 PMCID: PMC5765356 DOI: 10.1534/g3.117.300397] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity has been shown to increase risk for cardiovascular disease and type-2 diabetes. In addition, it has been implicated in aggravation of neurological conditions such as Alzheimer’s. In the model organism Drosophila melanogaster, a physiological state mimicking diet-induced obesity can be induced by subjecting fruit flies to a solid medium disproportionately higher in sugar than protein, or that has been supplemented with a rich source of saturated fat. These flies can exhibit increased circulating glucose levels, increased triglyceride content, insulin-like peptide resistance, and behavior indicative of neurological decline. We subjected flies to variants of the high-sugar diet, high-fat diet, or normal (control) diet, followed by a total RNA extraction from fly heads of each diet group for the purpose of Poly-A selected RNA-Sequencing. Our objective was to identify the effects of obesogenic diets on transcriptome patterns, how they differed between obesogenic diets, and identify genes that may relate to pathogenesis accompanying an obesity-like state. Gene ontology analysis indicated an overrepresentation of affected genes associated with immunity, metabolism, and hemocyanin in the high-fat diet group, and CHK, cell cycle activity, and DNA binding and transcription in the high-sugar diet group. Our results also indicate differences in the effects of the high-fat diet and high-sugar diet on expression profiles in head tissue of flies, despite the reportedly similar phenotypic impacts of the diets. The impacted genes, and how they may relate to pathogenesis in the Drosophila obesity-like state, warrant further experimental investigation.
Collapse
|
17
|
Sarfert KS, Knabe ML, Gunawansa NS, Blythe SN. Western-style diet induces object recognition deficits and alters complexity of dendritic arborization in the hippocampus and entorhinal cortex of male rats. Nutr Neurosci 2017; 22:344-353. [PMID: 29039252 DOI: 10.1080/1028415x.2017.1388557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Kathryn S. Sarfert
- Department of Neuroscience, Washington and Lee University, 204 W. Washington St., Lexington, VA 24450, USA
| | - Melina L. Knabe
- Department of Neuroscience, Washington and Lee University, 204 W. Washington St., Lexington, VA 24450, USA
| | - Nicole S. Gunawansa
- Department of Neuroscience, Washington and Lee University, 204 W. Washington St., Lexington, VA 24450, USA
| | - Sarah N. Blythe
- Department of Neuroscience, Washington and Lee University, 204 W. Washington St., Lexington, VA 24450, USA
- Department of Biology, Washington and Lee University, 204 W. Washington St., Lexington, VA 24450, USA
| |
Collapse
|
18
|
McGregor G, Harvey J. Food for thought: Leptin regulation of hippocampal function and its role in Alzheimer's disease. Neuropharmacology 2017; 136:298-306. [PMID: 28987937 DOI: 10.1016/j.neuropharm.2017.09.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/27/2017] [Accepted: 09/30/2017] [Indexed: 01/08/2023]
Abstract
Accumulating evidence indicates that diet and body weight are important factors associated with Alzheimer's disease (AD), with a significant increase in AD risk linked to mid-life obesity, and weight loss frequently occurring in the early stages of AD. This has fuelled interest in the hormone leptin, as it is an important hypothalamic regulator of food intake and body weight, but leptin also markedly influences the functioning of the hippocampus; a key brain region that degenerates in AD. Increasing evidence indicates that leptin has cognitive enhancing properties as it facilitates the cellular events that underlie hippocampal-dependent learning and memory. However, significant reductions in leptin's capacity to regulate hippocampal synaptic function occurs with age and dysfunctions in the leptin system are associated with an increased risk of AD. Moreover, leptin is a potential novel target in AD as leptin treatment has beneficial effects in various models of AD. Here we summarise recent advances in leptin neurobiology with particular focus on regulation of hippocampal synaptic function by leptin and the implications of this for neurodegenerative disorders like AD. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Gemma McGregor
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom
| | - Jenni Harvey
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom.
| |
Collapse
|
19
|
Jorgensen A, Kalliokoski O, Forsberg K, Breitenstein K, Weimann A, Henriksen T, Hau J, Wörtwein G, Poulsen HE, Jorgensen MB. A chronic increase of corticosterone age-dependently reduces systemic DNA damage from oxidation in rats. Free Radic Biol Med 2017; 104:64-74. [PMID: 28069523 DOI: 10.1016/j.freeradbiomed.2017.01.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/04/2017] [Accepted: 01/06/2017] [Indexed: 11/26/2022]
Abstract
Stress and depression are associated with an acceleration of brain and bodily aging; effects which have been attributed to chronic elevations of glucocorticoids. We tested the hypothesis that a three week administration of stress-associated levels of corticosterone (CORT, the principal rodent glucocorticoid) would increase systemic and CNS DNA and RNA damage from oxidation; a phenomenon known to be centrally involved in the aging process. We also hypothesized that older individuals would be more sensitive to this effect and that the chronic CORT administration would exacerbate age-related memory decline. Young and old male Sprague-Dawley rats were non-invasively administered CORT by voluntary ingestion of nut paste containing either CORT (25mg/kg) or vehicle for a total of 22 days. CORT increased the 24h urinary excretion of the hormone to the levels previously observed after experimental psychological stress and caused a downregulation of the glucocorticoid receptor in the CA1 area of the hippocampus. Contrary to our hypothesis, 24h excretion of 8-oxodG/8-oxoGuo (markers of DNA/RNA damage from oxidation) was reduced in CORT-treated young animals, whereas old animals showed no significant differences. In old animals, CORT caused a borderline significant reduction of RNA oxidation in CNS, which was paralleled by a normalization of performance in an object location memory test. To our knowledge, this is the first demonstration that chronic stress-associated levels of CORT can reduce nucleic acid damage from oxidation. These findings contradict the notion of elevated CORT as a mediator of the accelerated aging observed in stress and depression.
Collapse
Affiliation(s)
- Anders Jorgensen
- Psychiatric Center Copenhagen (Rigshospitalet), Mental Health Services of the Capital Region of Denmark, Denmark; Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen and Institute of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Denmark.
| | - Otto Kalliokoski
- Department of Experimental Medicine, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Kristin Forsberg
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen and Institute of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Katrine Breitenstein
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen and Institute of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Allan Weimann
- Laboratory of Clinical Pharmacology, Copenhagen University Hospital Rigshospitalet, Denmark; Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Denmark
| | - Trine Henriksen
- Laboratory of Clinical Pharmacology, Copenhagen University Hospital Rigshospitalet, Denmark; Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Denmark
| | - Jann Hau
- Department of Experimental Medicine, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Gitta Wörtwein
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen and Institute of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Henrik Enghusen Poulsen
- Laboratory of Clinical Pharmacology, Copenhagen University Hospital Rigshospitalet, Denmark; Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Denmark
| | - Martin Balslev Jorgensen
- Psychiatric Center Copenhagen (Rigshospitalet), Mental Health Services of the Capital Region of Denmark, Denmark; Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen and Institute of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Denmark
| |
Collapse
|
20
|
Klaren RE, Hubbard EA, Wetter NC, Sutton BP, Motl RW. Objectively measured sedentary behavior and brain volumetric measurements in multiple sclerosis. Neurodegener Dis Manag 2017; 7:31-37. [DOI: 10.2217/nmt-2016-0036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aim: This study examined the association between sedentary behavior patterns and whole brain gray matter (GM), white matter (WM) and subcortical GM structures in persons with multiple sclerosis (MS). Methods: 36 persons with MS wore an accelerometer and underwent a brain MRI. Whole brain GM and WM and deep GM structures were calculated from 3D T1-weighted structural brain images. Results: There were statistically significant (p < 0.01) and moderate or large associations between number of sedentary bouts/day and brain volume measures. The primary result was a consistent negative association between number of sedentary bouts/day and whole brain GM and WM, and deep GM structures. Conclusion: We provide novel evidence for decreased brain volume as a correlate of a sedentary behavior pattern in persons with MS.
Collapse
Affiliation(s)
- Rachel E Klaren
- Department of Kinesiology & Community Health, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Elizabeth A Hubbard
- Department of Kinesiology & Community Health, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Nathan C Wetter
- Jump Trading Stimulation & Education Center, Peoria, IL 61603, USA
| | - Bradley P Sutton
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Robert W Motl
- Department of Physical Therapy, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
21
|
de la Monte SM. Insulin Resistance and Neurodegeneration: Progress Towards the Development of New Therapeutics for Alzheimer's Disease. Drugs 2017; 77:47-65. [PMID: 27988872 PMCID: PMC5575843 DOI: 10.1007/s40265-016-0674-0] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) should be regarded as a degenerative metabolic disease caused by brain insulin resistance and deficiency, and overlapping with the molecular, biochemical, pathophysiological, and metabolic dysfunctions in diabetes mellitus, non-alcoholic fatty liver disease, and metabolic syndrome. Although most of the diagnostic and therapeutic approaches over the past several decades have focused on amyloid-beta (Aβ42) and aberrantly phosphorylated tau, which could be caused by consequences of brain insulin resistance, the broader array of pathologies including white matter atrophy with loss of myelinated fibrils and leukoaraiosis, non-Aβ42 microvascular disease, dysregulated lipid metabolism, mitochondrial dysfunction, astrocytic gliosis, neuro-inflammation, and loss of synapses vis-à-vis growth of dystrophic neurites, is not readily accounted for by Aβ42 accumulations, but could be explained by dysregulated insulin/IGF-1 signaling with attendant impairments in signal transduction and gene expression. This review covers the diverse range of brain abnormalities in AD and discusses how insulins, incretins, and insulin sensitizers could be utilized to treat at different stages of neurodegeneration.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Neurology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Pierre Galletti Research Building, 55 Claverick Street, Room 419, Providence, RI, 02903, USA.
- Department of Neurosurgery, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Neuropathology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Pathology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
22
|
Obesity Weighs down Memory through a Mechanism Involving the Neuroepigenetic Dysregulation of Sirt1. J Neurosci 2016; 36:1324-35. [PMID: 26818519 DOI: 10.1523/jneurosci.1934-15.2016] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED Aberrant gene expression within the hippocampus has recently been implicated in the pathogenesis of obesity-induced memory impairment. Whether a dysregulation of epigenetic modifications mediates this disruption in gene transcription has yet to be established. Here we report evidence of obesity-induced alterations in DNA methylation of memory-associated genes, including Sirtuin 1 (Sirt1), within the hippocampus, and thus offer a novel mechanism by which SIRT1 expression within the hippocampus is suppressed during obesity. Forebrain neuron-specific Sirt1 knock-out closely recapitulated the memory deficits exhibited by obese mice, consistent with the hypothesis that the high-fat diet-mediated reduction of hippocampal SIRT1 could be responsible for obesity-linked memory impairment. Obese mice fed a diet supplemented with the SIRT1-activating molecule resveratrol exhibited increased hippocampal SIRT1 activity and preserved hippocampus-dependent memory, further strengthening this conclusion. Thus, our findings suggest that the memory-impairing effects of diet-induced obesity may potentially be mediated by neuroepigenetic dysregulation of SIRT1 within the hippocampus. SIGNIFICANCE STATEMENT Previous studies have implicated transcriptional dysregulation within the hippocampus as being a relevant pathological concomitant of obesity-induced memory impairment, yet a deeper understanding of the basis for, and etiological significance of, transcriptional dysregulation in this context is lacking. Here we present the first evidence of epigenetic dysregulation (i.e., altered DNA methylation and hydroxymethylation) of memory-related genes, including Sirt1, within the hippocampus of obese mice. Furthermore, experiments using transgenic and pharmacological approaches strongly implicate reduced hippocampal SIRT1 as being a principal pathogenic mediator of obesity-induced memory impairment. This paper offers a novel working model that may serve as a conceptual basis for the development of therapeutic interventions for obesity-induced memory impairment.
Collapse
|
23
|
McGregor G, Malekizadeh Y, Harvey J. Minireview: Food for thought: regulation of synaptic function by metabolic hormones. Mol Endocrinol 2016; 29:3-13. [PMID: 25470238 DOI: 10.1210/me.2014-1328] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The peripheral actions of the metabolic hormones, leptin and insulin, are well documented. However, the functions of these hormones are not restricted to the periphery because evidence is growing that both leptin and insulin can readily cross the blood-brain barrier and have widespread central actions. The hippocampus in particular expresses high levels of both insulin and leptin receptors as well as key components of their associated signaling cascades. Moreover, recent studies indicate that both hormones are potential cognitive enhancers. Indeed, it has been demonstrated that both leptin and insulin markedly influence key cellular events that underlie hippocampal learning and memory including activity-dependent synaptic plasticity and the trafficking of glutamate receptors to and away from hippocampal synapses. The hippocampal formation is also a prime site for the neurodegenerative processes that occur during Alzheimer's disease, and impairments in either leptin or insulin function have been linked to central nervous system-driven diseases like Alzheimer's disease. Thus, the capacity of the metabolic hormones, leptin and insulin, to regulate hippocampal synaptic function has significant implications for normal brain function and also central nervous system-driven disease.
Collapse
Affiliation(s)
- Gemma McGregor
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom
| | | | | |
Collapse
|
24
|
Amylin-mediated control of glycemia, energy balance, and cognition. Physiol Behav 2016; 162:130-40. [PMID: 26922873 DOI: 10.1016/j.physbeh.2016.02.034] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/20/2016] [Accepted: 02/22/2016] [Indexed: 12/26/2022]
Abstract
Amylin, a peptide hormone produced in the pancreas and in the brain, has well-established physiological roles in glycemic regulation and energy balance control. It improves postprandial blood glucose levels by suppressing gastric emptying and glucagon secretion; these beneficial effects have led to the FDA-approved use of the amylin analog pramlintide in the treatment of diabetes mellitus. Amylin also acts centrally as a satiation signal, reducing food intake and body weight. The ability of amylin to promote negative energy balance, along with its unique capacity to cooperatively facilitate or enhance the intake- and body weight-suppressive effects of other neuroendocrine signals like leptin, have made amylin a leading target for the development of novel pharmacotherapies for the treatment of obesity. In addition to these more widely studied effects, a growing body of literature suggests that amylin may play a role in processes related to cognition, including the neurodegeneration and cognitive deficits associated with Alzheimer's disease (AD). Although the function of amylin in AD is still unclear, intriguing recent reports indicate that amylin may improve cognitive ability and reduce hallmarks of neurodegeneration in the brain. The frequent comorbidity of diabetes mellitus and obesity, as well as the increased risk for and occurrence of AD associated with these metabolic diseases, suggests that amylin-based pharmaceutical strategies may provide multiple therapeutic benefits. This review will discuss the known effects of amylin on glycemic regulation, energy balance control, and cognitive/motivational processes. Particular focus will be devoted to the current and/or potential future clinical use of amylin pharmacotherapies for the treatment of diseases in each of these realms.
Collapse
|
25
|
Abstract
The hippocampus has a pivotal role in learning and in the formation and consolidation of memory and is critically involved in the regulation of emotion, fear, anxiety, and stress. Studies of the hippocampus have been central to the study of memory in humans and in recent years, the regional specialization and organization of hippocampal functions have been elucidated in experimental models and in human neurological and psychiatric diseases. The hippocampus has long been considered a classic model for the study of neuroplasticity as many examples of synaptic plasticity such as long-term potentiation and -depression have been identified and demonstrated in hippocampal circuits. Neuroplasticity is the ability to adapt and reorganize the structure or function to internal or external stimuli and occurs at the cellular, population, network or behavioral level and is reflected in the cytological and network architecture as well as in intrinsic properties of hippocampal neurons and circuits. The high degree of hippocampal neuroplasticity might, however, be also negatively reflected in the pronounced vulnerability of the hippocampus to deleterious conditions such as ischemia, epilepsy, chronic stress, neurodegeneration and aging targeting hippocampal structure and function and leading to cognitive deficits. Considering this framework of plasticity and vulnerability, we here review basic principles of hippocampal anatomy and neuroplasticity on various levels as well as recent findings regarding the functional organization of the hippocampus in light of the regional vulnerability in Alzheimer's disease, ischemia, epilepsy, neuroinflammation and aging.
Collapse
Affiliation(s)
- T Bartsch
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital Schleswig-Holstein, Kiel, Germany.
| | - P Wulff
- Institute of Physiology, Neurophysiology, University of Kiel, Olshausenstrasse 40, 24098 Kiel, Germany.
| |
Collapse
|
26
|
Models and mechanisms for hippocampal dysfunction in obesity and diabetes. Neuroscience 2015; 309:125-39. [PMID: 25934036 DOI: 10.1016/j.neuroscience.2015.04.045] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/15/2015] [Accepted: 04/21/2015] [Indexed: 01/05/2023]
Abstract
Clinical studies suggest that obesity and Type 2 (insulin-resistant) diabetes impair the structural integrity of medial temporal lobe regions involved in memory and confer greater vulnerability to neurological insults. While eliminating obesity and its endocrine comorbidities would be the most straightforward way to minimize cognitive risk, structural barriers to physical activity and the widespread availability of calorically dense, highly palatable foods will likely necessitate additional strategies to maintain brain health over the lifespan. Research in rodents has identified numerous correlates of hippocampal functional impairment in obesity and diabetes, with several studies demonstrating causality in subsequent mechanistic studies. This review highlights recent work on pathways and cell-cell interactions underlying the synaptic consequences of obesity, diabetes, or in models with both pathological conditions. Although the mechanisms vary across different animal models, immune activation has emerged as a shared feature of obesity and diabetes, with synergistic exacerbation of neuroinflammation in model systems with both conditions. This review discusses these findings with reference to the benefits of incorporating existing models from the fields of obesity and metabolic disease. Many transgenic lines with basal metabolic alterations or differential susceptibility to diet-induced obesity have yet to be characterized with respect to their cognitive and synaptic phenotype. Adopting these models, and building on the extensive knowledge base used to generate them, is a promising avenue for understanding interactions between peripheral disease states and neurodegenerative disorders.
Collapse
|
27
|
Hashimoto T, Takeuchi H, Taki Y, Yokota S, Hashizume H, Asano K, Asano M, Sassa Y, Nouchi R, Kawashima R. Increased posterior hippocampal volumes in children with lower increase in body mass index: a 3-year longitudinal MRI study. Dev Neurosci 2015; 37:153-60. [PMID: 25721327 DOI: 10.1159/000370064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 11/24/2014] [Indexed: 11/19/2022] Open
Abstract
People are generally lean during childhood and show more variability in body sizes and shapes later in life. Cortical development generally correlates with body growth. However, in children cortical growth may be impaired with oversized body growth. Inverse correlations between body mass index (BMI) and brain volumes suggest that lean bodies may be associated with increased cortical volume. To clarify the positive effects of a lean body on a child's cortical development, we used MRI to measure brain structures longitudinally in 107 children and adolescents aged 5-16 years. The relationships between changes in BMI and cortical volumes during 3 years of development were investigated, while controlling for age, gender and intracranial volume changes. Voxel-based morphometry analyses revealed that an increase in the volume of the right posterior medial temporal lobe – including the hippocampus and parahippocampal gyrus – was associated with lower BMI increases. No correlations were observed between higher BMI increases and cortical volumes. Our results suggest that keeping a lean body – or not getting fat – during childhood can induce an increase in regional cortical volume rather than impair growth. This is the first longitudinal study showing positive effects of a lean body on cortical development in children.
Collapse
Affiliation(s)
- Teruo Hashimoto
- Division of Developmental Cognitive Neuroscience, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Torma F, Bori Z, Koltai E, Felszeghy K, Vacz G, Koch L, Britton S, Boldogh I, Radak Z. Eating habits modulate short term memory and epigenetical regulation of brain derived neurotrophic factor in hippocampus of low- and high running capacity rats. Brain Res Bull 2014; 107:54-60. [PMID: 25043449 DOI: 10.1016/j.brainresbull.2014.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/07/2014] [Accepted: 07/09/2014] [Indexed: 12/20/2022]
Abstract
Exercise capacity and dietary restriction (DR) are linked to improved quality of life, including enhanced brain function and neuro-protection. Brain derived neurotrophic factor (BDNF) is one of the key proteins involved in the beneficial effects of exercise on brain. Low capacity runner (LCR) and high capacity runner (HCR) rats were subjected to DR in order to investigate the regulation of BDNF. HCR-DR rats out-performed other groups in a passive avoidance test. BDNF content increased significantly in the hippocampus of HCR-DR groups compared to control groups (p<0.05). The acetylation of H3 increased significantly only in the LCR-DR group. However, chip-assay revealed that the specific binding between acetylated histone H3 and BNDF promoter was increased in both LCR-DR and HCR-DR groups. In spite of these increases in binding, at the transcriptional level only, the LCR-DR group showed an increase in BDNF mRNA content. Additionally, DR also induced the activity of cAMP response element-binding protein (CREB), while the content of SIRT1 was not altered. Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) was elevated in HCR-DR groups. But, based on the levels of nuclear respiratory factor-1 and cytocrome c oxidase, it appears that DR did not cause mitochondrial biogenesis. The data suggest that DR-mediated induction of BDNF levels includes chromatin remodeling. Moreover, DR does not induce mitochondrial biogenesis in the hippocampus of LCR/HCR rats. DR results in different responses to a passive avoidance test, and BDNF regulation in LCR and HCR rats.
Collapse
Affiliation(s)
- Ferenc Torma
- Research Institute of Sport Science, Semmelweis University, Budapest, Hungary
| | - Zoltan Bori
- Research Institute of Sport Science, Semmelweis University, Budapest, Hungary
| | - Erika Koltai
- Research Institute of Sport Science, Semmelweis University, Budapest, Hungary
| | - Klara Felszeghy
- Research Institute of Sport Science, Semmelweis University, Budapest, Hungary
| | - Gabriella Vacz
- Research Institute of Sport Science, Semmelweis University, Budapest, Hungary
| | - Lauren Koch
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Steven Britton
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Zsolt Radak
- Research Institute of Sport Science, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
29
|
Murphy T, Dias GP, Thuret S. Effects of diet on brain plasticity in animal and human studies: mind the gap. Neural Plast 2014; 2014:563160. [PMID: 24900924 PMCID: PMC4037119 DOI: 10.1155/2014/563160] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/17/2014] [Indexed: 02/07/2023] Open
Abstract
Dietary interventions have emerged as effective environmental inducers of brain plasticity. Among these dietary interventions, we here highlight the impact of caloric restriction (CR: a consistent reduction of total daily food intake), intermittent fasting (IF, every-other-day feeding), and diet supplementation with polyphenols and polyunsaturated fatty acids (PUFAs) on markers of brain plasticity in animal studies. Moreover, we also discuss epidemiological and intervention studies reporting the effects of CR, IF and dietary polyphenols and PUFAs on learning, memory, and mood. In particular, we evaluate the gap in mechanistic understanding between recent findings from animal studies and those human studies reporting that these dietary factors can benefit cognition, mood, and anxiety, aging, and Alzheimer's disease-with focus on the enhancement of structural and functional plasticity markers in the hippocampus, such as increased expression of neurotrophic factors, synaptic function and adult neurogenesis. Lastly, we discuss some of the obstacles to harnessing the promising effects of diet on brain plasticity in animal studies into effective recommendations and interventions to promote healthy brain function in humans. Together, these data reinforce the important translational concept that diet, a modifiable lifestyle factor, holds the ability to modulate brain health and function.
Collapse
Affiliation(s)
- Tytus Murphy
- Institute of Psychiatry, King's College London, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Gisele Pereira Dias
- Institute of Psychiatry, King's College London, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Sandrine Thuret
- Institute of Psychiatry, King's College London, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
30
|
Ménard C, Quirion R, Bouchard S, Ferland G, Gaudreau P. Glutamatergic signaling and low prodynorphin expression are associated with intact memory and reduced anxiety in rat models of healthy aging. Front Aging Neurosci 2014; 6:81. [PMID: 24847259 PMCID: PMC4019859 DOI: 10.3389/fnagi.2014.00081] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 04/16/2014] [Indexed: 11/13/2022] Open
Abstract
The LOU/C/Jall (LOU) rat strain is considered a model of healthy aging due to its increased longevity, maintenance of stable body weight (BW) throughout life and low incidence of age-related diseases. However, aging LOU rat cognitive and anxiety status has yet to be investigated. In the present study, male and female LOU rat cognitive performances (6-42 months) were assessed using novel object recognition and Morris Water Maze tasks. Recognition memory remained intact in all LOU rats up to 42 months of age. As for spatial memory, old LOU rat performed similarly as young animals for learning acquisition, reversal learning, and retention. While LOU rat BW remained stable despite aging, 20-month-old ad-libitum-fed (OAL) male Sprague Dawley rats become obese. We determined if long-term caloric restriction (LTCR) prevents age-related BW increase and cognitive deficits in this rat strain, as observed in the obesity-resistant LOU rats. Compared to young animals, recognition memory was impaired in OAL but intact in 20-month-old calorie-restricted (OCR) rats. Similarly, OAL spatial learning acquisition was impaired but LTCR prevented the deficits. Exacerbated stress responses may favor age-related cognitive decline. In the elevated plus maze and open field tasks, LOU and OCR rats exhibited high levels of exploratory activity whereas OAL rats displayed anxious behaviors. Expression of prodynorphin (Pdyn), an endogenous peptide involved in stress-related memory impairments, was increased in the hippocampus of OAL rats. Group 1 metabotropic glutamate receptor 5 and immediate early genes Homer 1a and Arc expression, both associated with successful cognitive aging, were unaltered in aging LOU rats but lower in OAL than OCR rats. Altogether, our results, supported by principal component analysis and correlation matrix, suggest that intact memory and low anxiety are associated with glutamatergic signaling and low Pdyn expression in the hippocampus of non-obese aging rats.
Collapse
Affiliation(s)
- Caroline Ménard
- Neuroscience Division, Douglas Mental Health University Institute Research Center Montreal, QC, Canada ; Department of Psychiatry, McGill University Montreal, QC, Canada ; Laboratory of Neuroendocrinology of Aging, Centre Hospitalier de l'Université de Montréal Research Center Montreal, QC, Canada ; Department of Medicine, University of Montreal Montreal, QC, Canada
| | - Rémi Quirion
- Neuroscience Division, Douglas Mental Health University Institute Research Center Montreal, QC, Canada ; Department of Psychiatry, McGill University Montreal, QC, Canada
| | - Sylvain Bouchard
- Faculty of Medicine, University of Montreal Montreal, QC, Canada
| | - Guylaine Ferland
- Hôpital du Sacré-Coeur de Montréal Research Center Montreal, QC, Canada ; Department of Nutrition, University of Montreal Montreal, QC, Canada
| | - Pierrette Gaudreau
- Laboratory of Neuroendocrinology of Aging, Centre Hospitalier de l'Université de Montréal Research Center Montreal, QC, Canada ; Department of Medicine, University of Montreal Montreal, QC, Canada
| |
Collapse
|
31
|
Selkrig J, Wong P, Zhang X, Pettersson S. Metabolic tinkering by the gut microbiome: Implications for brain development and function. Gut Microbes 2014; 5:369-80. [PMID: 24685620 PMCID: PMC4153776 DOI: 10.4161/gmic.28681] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Brain development is an energy demanding process that relies heavily upon diet derived nutrients. Gut microbiota enhance the host's ability to extract otherwise inaccessible energy from the diet via fermentation of complex oligosaccharides in the colon. This nutrient yield is estimated to contribute up to 10% of the host's daily caloric requirement in humans and fluctuates in response to environmental variations. Research over the past decade has demonstrated a surprising role for the gut microbiome in normal brain development and function. In this review we postulate that perturbations in the gut microbial-derived nutrient supply, driven by environmental variation, profoundly impacts upon normal brain development and function.
Collapse
Affiliation(s)
- Joel Selkrig
- School of Biological Sciences; Nanyang Technological University; Singapore, Singapore,Lee Kong Chain School of Medicine; Nanyang Technological University; Singapore, Singapore
| | - Peiyan Wong
- Program in Neuroscience and Behavioral Disorders; Duke-NUS Graduate Medical School Singapore; Singapore, Singapore,Behavioural Phenotyping Core Facility; Duke-NUS; Duke University Medical Center; Durham, NC USA
| | - Xiaodong Zhang
- Program in Neuroscience and Behavioral Disorders; Duke-NUS Graduate Medical School Singapore; Singapore, Singapore,Department of Physiology; National University of Singapore; Singapore, Singapore,Departments of Psychiatry and Behavioral Sciences; Duke University Medical Center; Durham, NC USA
| | - Sven Pettersson
- Lee Kong Chain School of Medicine; Nanyang Technological University; Singapore, Singapore,Department of Microbiology, Tumor, and Cell Biology (MTC); Karolinska Institute; Stockholm, Sweden,Correspondence to: Sven Pettersson,
| |
Collapse
|
32
|
Verge VMK, Andreassen CS, Arnason TG, Andersen H. Mechanisms of disease: role of neurotrophins in diabetes and diabetic neuropathy. HANDBOOK OF CLINICAL NEUROLOGY 2014; 126:443-60. [PMID: 25410238 DOI: 10.1016/b978-0-444-53480-4.00032-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neuropathy is an insidious and devastating consequence of diabetes. Early studies provided a strong rationale for deficient neurotrophin support in the pathogenesis of diabetic neuropathy in a number of critical tissues and organs. It has now been over a decade since the first failed human neurotrophin supplementation clinical trials, but mounting evidence still implicates these trophic factors in diabetic neuropathy. Since then, tremendous advances have been made in our understanding of the complexities of neurotrophin signaling and processing and how the diabetic milieu might impact this. This in turn changes both our perception of how the altered trophic environment contributes to the etiology of diabetic neuropathy and the design of future neurotrophin therapeutic interventions. This chapter summarizes some of these findings and attempts to integrate neurotrophin actions on the nervous system with an increasing appreciation of their role in the regulation of metabolic processes in diabetes that impact the diabetic neuropathic state.
Collapse
Affiliation(s)
- Valerie M K Verge
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Canada; Cameco MS Neuroscience Research Center, University of Saskatchewan, Saskatoon City Hospital, Saskatoon, Canada.
| | - Christer S Andreassen
- Department of Otorhinolaryngology and Head and Neck Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Terra G Arnason
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Canada; Department of Medicine, Division of Endocrinology and Metabolism, University of Saskatchewan, Saskatoon, Canada
| | - Henning Andersen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
33
|
Compensatory actions of orexinergic neurons in the lateral hypothalamus during metabolic or cortical challenges may enable the coupling of metabolic dysfunction and cortical dysfunction. Med Hypotheses 2013; 80:520-6. [DOI: 10.1016/j.mehy.2013.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 01/03/2013] [Accepted: 02/07/2013] [Indexed: 11/20/2022]
|
34
|
Marques-Aleixo I, Oliveira PJ, Moreira PI, Magalhães J, Ascensão A. Physical exercise as a possible strategy for brain protection: Evidence from mitochondrial-mediated mechanisms. Prog Neurobiol 2012; 99:149-62. [DOI: 10.1016/j.pneurobio.2012.08.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 07/14/2012] [Accepted: 08/17/2012] [Indexed: 01/01/2023]
|
35
|
Heyward FD, Walton RG, Carle MS, Coleman MA, Garvey WT, Sweatt JD. Adult mice maintained on a high-fat diet exhibit object location memory deficits and reduced hippocampal SIRT1 gene expression. Neurobiol Learn Mem 2012; 98:25-32. [PMID: 22542746 DOI: 10.1016/j.nlm.2012.04.005] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Revised: 04/09/2012] [Accepted: 04/12/2012] [Indexed: 10/28/2022]
Abstract
Mounting evidence has established that diet-induced obesity (DIO) is associated with deficits in hippocampus-dependent memory. The bulk of research studies dealing with this topic have utilized rats fed a high-fat diet as an experimental model. To date, there has been a paucity of research studies that have established whether the memory deficits exhibited in DIO rats can be recapitulated in mice. Moreover, the majority of experiments that have evaluated memory performance in rodent models of DIO have utilized memory tests that are essentially aversive in nature (i.e., Morris water maze). The current study sought to fill an empirical void by determining if mice maintained on a high-fat diet exhibit deficits in two non-aversive memory paradigms: novel object recognition (NOR) and object location memory (OLM). Here we report that mice fed a high-fat diet over 23 weeks exhibit intact NOR, albeit a marked impairment in hippocampus-dependent OLM. We also determined the existence of corresponding aberrations in gene expression within the hippocampus of DIO mice. DIO mice exhibited significant reductions in both SIRT1 and PP1 mRNA within the hippocampus. Our data suggest that mice maintained on a high-fat diet present with impaired hippocampus-dependent spatial memory and a corresponding alteration in the expression of genes that have been implicated in memory consolidation.
Collapse
Affiliation(s)
- Frankie D Heyward
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, 1074 Shelby Building, 1825 University Boulevard, Birmingham, USA
| | | | | | | | | | | |
Collapse
|
36
|
Wikgren J, Mertikas GG, Raussi P, Tirkkonen R, Äyräväinen L, Pelto-Huikko M, Koch LG, Britton SL, Kainulainen H. Selective breeding for endurance running capacity affects cognitive but not motor learning in rats. Physiol Behav 2012; 106:95-100. [PMID: 22285210 DOI: 10.1016/j.physbeh.2012.01.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 12/09/2011] [Accepted: 01/16/2012] [Indexed: 01/10/2023]
Abstract
The ability to utilize oxygen has been shown to affect a wide variety of physiological factors often considered beneficial for survival. As the ability to learn can be seen as one of the core factors of survival in mammals, we studied whether selective breeding for endurance running, an indication of aerobic capacity, also has an effect on learning. Rats selectively bred over 23 generations for their ability to perform forced treadmill running were trained in an appetitively motivated discrimination-reversal classical conditioning task, an alternating T-maze task followed by a rule change (from a shift-win to stay-win rule) and motor learning task. In the discrimination-reversal and T-maze tasks, the high-capacity runner (HCR) rats outperformed the low-capacity runner (LCR) rats, most notably in the phases requiring flexible cognition. In the Rotarod (motor-learning) task, the HCR animals were overall more agile but learned at a similar rate with the LCR group as a function of training. We conclude that the intrinsic ability to utilize oxygen is associated especially with tasks requiring plasticity of the brain structures implicated in flexible cognition.
Collapse
Affiliation(s)
- Jan Wikgren
- Department of Psychology, University of Jyväskylä, Finland.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Leptin: a novel therapeutic target in Alzheimer's disease? Int J Alzheimers Dis 2012; 2012:594137. [PMID: 22254146 PMCID: PMC3255100 DOI: 10.1155/2012/594137] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 10/15/2011] [Indexed: 12/12/2022] Open
Abstract
It is well established that the hormone leptin circulates in the plasma in amounts proportional to body fat content and it regulates food intake and body weight via its actions in the hypothalamus. However, numerous studies have shown that leptin receptors are widely expressed throughout the CNS and evidence is growing that leptin plays a role in modulating a variety of neuronal processes. In particular, recent studies have highlighted a potential cognitive enhancing role for leptin as it regulates diverse aspects of hippocampal synaptic function that are thought to underlie learning and memory processes including glutamate receptor trafficking, dendritic morphology, and activity-dependent synaptic plasticity. Characterisation of the novel actions of leptin in limbic brain regions is providing valuable insights into leptin's role in higher cognitive functions in health and disease.
Collapse
|
38
|
Zilberter T. Carbohydrate-biased control of energy metabolism: the darker side of the selfish brain. FRONTIERS IN NEUROENERGETICS 2011; 3:8. [PMID: 22194720 PMCID: PMC3243104 DOI: 10.3389/fnene.2011.00008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 12/02/2011] [Indexed: 01/24/2023]
|
39
|
McNay EC, Recknagel AK. Reprint of: 'Brain insulin signaling: A key component of cognitive processes and a potential basis for cognitive impairment in type 2 diabetes'. Neurobiol Learn Mem 2011; 96:517-28. [PMID: 22085799 DOI: 10.1016/j.nlm.2011.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Understanding of the role of insulin in the brain has gradually expanded, from initial conceptions of the brain as insulin-insensitive through identification of a role in regulation of feeding, to recent demonstration of insulin as a key component of hippocampal memory processes. Conversely, systemic insulin resistance such as that seen in type 2 diabetes is associated with a range of cognitive and neural deficits. Here we review the evidence for insulin as a cognitive and neural modulator, including potential effector mechanisms, and examine the impact that type 2 diabetes has on these mechanisms in order to identify likely bases for the cognitive impairments seen in type 2 diabetic patients.
Collapse
Affiliation(s)
- Ewan C McNay
- Behavioral Neuroscience and Center for Neuroscience Research, University at Albany (SUNY), SS399, 1400 Washington Avenue, Albany, NY 12222, USA.
| | | |
Collapse
|
40
|
McNay EC, Recknagel AK. Brain insulin signaling: a key component of cognitive processes and a potential basis for cognitive impairment in type 2 diabetes. Neurobiol Learn Mem 2011; 96:432-42. [PMID: 21907815 DOI: 10.1016/j.nlm.2011.08.005] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Revised: 07/09/2011] [Accepted: 08/12/2011] [Indexed: 12/16/2022]
Abstract
Understanding of the role of insulin in the brain has gradually expanded, from initial conceptions of the brain as insulin-insensitive through identification of a role in regulation of feeding, to recent demonstration of insulin as a key component of hippocampal memory processes. Conversely, systemic insulin resistance such as that seen in type 2 diabetes is associated with a range of cognitive and neural deficits. Here we review the evidence for insulin as a cognitive and neural modulator, including potential effector mechanisms, and examine the impact that type 2 diabetes has on these mechanisms in order to identify likely bases for the cognitive impairments seen in type 2 diabetic patients.
Collapse
Affiliation(s)
- Ewan C McNay
- Behavioral Neuroscience and Center for Neuroscience Research, University at Albany (SUNY), SS399, 1400 Washington Avenue, Albany, NY 12222, USA.
| | | |
Collapse
|
41
|
|
42
|
Stranahan AM, Cutler RG, Button C, Telljohann R, Mattson MP. Diet-induced elevations in serum cholesterol are associated with alterations in hippocampal lipid metabolism and increased oxidative stress. J Neurochem 2011; 118:611-5. [PMID: 21682722 DOI: 10.1111/j.1471-4159.2011.07351.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The structure and function of the hippocampus, a brain region critical for learning and memory, is impaired by obesity and hyperlipidemia. Peripheral cholesterol and sphingolipids increase progressively with aging and are associated with a range of age-related diseases. However, the mechanisms linking peripheral cholesterol metabolism to hippocampal neuroplasticity remain poorly understood. To determine whether diets that elevate serum cholesterol influence lipid metabolism in the hippocampus, we maintained rats on a diet with high amounts of saturated fat and simple sugars for 3 months and then analyzed hippocampal lipid species using tandem mass spectrometry. The high fat diet was associated with increased serum and liver cholesterol and triglyceride levels, and also promoted cholesterol accumulation in the hippocampus. Increases in hippocampal cholesterol were associated with elevated galactosyl ceramide and sphingomyelin. To determine whether changes in lipid composition exerted biological effects, we measured levels of the lipid peroxidation products 4-hydroxynonenal-lysine and 4-hydroxynonenal-histidine; both were increased locally in the hippocampus, indicative of cell membrane-associated oxidative stress. Taken together, these observations support the existence of a potentially pathogenic relationship between dietary fat intake, peripheral cholesterol and triglyceride levels, brain cell sphingolipid metabolism, and oxidative stress.
Collapse
Affiliation(s)
- Alexis M Stranahan
- Cellular and Molecular Neuroscience Section, Laboratory of Neurosciences, National Institute on Aging, Biomedical Research Center, Baltimore, Maryland, USA.
| | | | | | | | | |
Collapse
|