1
|
Netsyk O, Korol SV, Li JP, Birnir B, Jin Z. GABA-activated slow spontaneous inhibitory postsynaptic currents are decreased in dorsal hippocampal dentate gyrus granule cells in an aged mouse model of Alzheimer's disease. J Alzheimers Dis 2025; 104:420-428. [PMID: 39956943 DOI: 10.1177/13872877251317608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
BackgroundSynaptic transmission dysfunction is associated with a range of neurological disorders, including Alzheimer's disease (AD). However, the role of γ-aminobutyric acid (GABA)-mediated synaptic inhibition in AD has not been fully explored.ObjectiveWe studied basal, GABA-activated slow spontaneous synaptic currents (sIPSCs) in dentate gyrus (DG) granule cells in the dorsal hippocampus of an AD mouse model (tg-APPSwe) and investigated insulin's modulatory effects.MethodsGABA-activated slow sIPSCs were recorded in the DG granule cells by whole-cell patch--clamp recordings in dorsal hippocampal brain slices from 5-6 (adult) and 10-12 (aged) months old wild-type (WT) and AD mice, in the presence or absence of insulin (1 nM).ResultsThe median 10-90% rise time of slow sIPSCs significantly decreased with age (10-12 months vs. 5-6 months) only in AD mice. The median amplitude of the slow sIPSCs was decreased in adult and aged AD mice as compared to WT mice whereas the slow sIPSCs frequency was only reduced in the aged WT mice. The median 63% decay time and total current density of the slow IPSCs was significantly decreased in the aged AD mice as compared to both WT mice and to the adult AD mice. Insulin application exerted no effect on slow sIPSCs properties in any of the animal groups.ConclusionsThe characteristics of the slow sIPSCs recorded in DG granule cells of dorsal hippocampus from WT and AD mice are altered by age- and disease-state, whereas insulin has negligible effects.
Collapse
Affiliation(s)
- Olga Netsyk
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Sergiy V Korol
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Bryndis Birnir
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Zhe Jin
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
2
|
Lounici A, Iacob A, Hongler K, Mölling MA, Drechsler M, Hersberger L, Sethi S, Lang UE, Liwinski T. Ketogenic Diet as a Nutritional Metabolic Intervention for Obsessive-Compulsive Disorder: A Narrative Review. Nutrients 2024; 17:31. [PMID: 39796465 PMCID: PMC11723184 DOI: 10.3390/nu17010031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
The substantial evidence supporting the ketogenic diet (KD) in epilepsy management has spurred research into its effects on other neurological and psychiatric conditions. Despite differences in characteristics, symptoms, and underlying mechanisms, these conditions share common pathways that the KD may influence. The KD reverses metabolic dysfunction. Moreover, it has been shown to support neuroprotection through mechanisms such as neuronal energy support, inflammation reduction, amelioration of oxidative stress, and reversing mitochondrial dysfunction. The adequate intake of dietary nutrients is essential for maintaining normal brain functions, and strong evidence supports the role of nutrition in the treatment and prevention of many psychiatric and neurological disorders. Obsessive-compulsive disorder (OCD) is a neuropsychiatric condition marked by persistent, distressing thoughts or impulses (obsessions) and repetitive behaviors performed in response to these obsessions (compulsions). Recent studies have increasingly examined the role of nutrition and metabolic disorders in OCD. This narrative review examines current evidence on the potential role of the KD in the treatment of OCD. We explore research on the KD's effects on psychiatric disorders to assess its potential relevance for OCD treatment. Additionally, we identify key gaps in the preclinical and clinical research that warrant further study in applying the KD as a metabolic therapy for OCD.
Collapse
Affiliation(s)
- Astrid Lounici
- Clinic for Adults, University Psychiatric Clinics Basel, University of Basel, 4031 Basel, Switzerland; (A.L.); (K.H.); (U.E.L.)
| | - Ana Iacob
- Pôle de Psychiatrie et Psychothérapie (PPP), Unité de Psychiatrie de Liaison, Hôpital du Valais, 1950 Sion, Switzerland;
| | - Katarzyna Hongler
- Clinic for Adults, University Psychiatric Clinics Basel, University of Basel, 4031 Basel, Switzerland; (A.L.); (K.H.); (U.E.L.)
| | | | - Maria Drechsler
- Stiftung für Ganzheitliche Medizin (SGM), Klinik SGM Langenthal, 4900 Langenthal, Switzerland; (M.D.); (L.H.)
| | - Luca Hersberger
- Stiftung für Ganzheitliche Medizin (SGM), Klinik SGM Langenthal, 4900 Langenthal, Switzerland; (M.D.); (L.H.)
| | - Shebani Sethi
- Metabolic Psychiatry, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA;
| | - Undine E. Lang
- Clinic for Adults, University Psychiatric Clinics Basel, University of Basel, 4031 Basel, Switzerland; (A.L.); (K.H.); (U.E.L.)
| | - Timur Liwinski
- Clinic for Adults, University Psychiatric Clinics Basel, University of Basel, 4031 Basel, Switzerland; (A.L.); (K.H.); (U.E.L.)
| |
Collapse
|
3
|
Karimani F, Asgari Taei A, Kaveh N, Rabiei Ghahfarokhi M, Abolghasemi Dehaqani MR, Dargahi L. Hippocampal sharp-wave ripples and hippocampal-prefrontal synchrony regulate memory-enhancing effects of intranasal insulin in an STZ-induced Alzheimer's model. Life Sci 2024; 357:123094. [PMID: 39362588 DOI: 10.1016/j.lfs.2024.123094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/26/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
AIMS Alzheimer's disease is characterized by memory loss and pathological changes in the brain, such as amyloid beta and tau pathology, disruptions in neural circuits and neuronal oscillations are also significant indicators of this disease and potential therapeutic targets. We studied how intranasal insulin impacts memory and neural oscillations in an Alzheimer's disease rat model induced by STZ. MAIN METHODS Male Wistar rats were intracerebroventricularly injected with STZ, followed by intranasal insulin therapy. Electrophysiological recordings were conducted in the hippocampus and medial prefrontal cortex to assess local field potentials. Memory was assessed using novel object recognition and Y-maze tests. Amyloid and tau pathology and neuronal loss were also evaluated in the hippocampus. KEY FINDING Alterations in theta-gamma oscillations following insulin treatment were not significant. However, insulin administration ameliorated hippocampal sharp-wave ripples deficit and augmented hippocampal-prefrontal theta coherence. Concurrently, insulin therapy enhanced spatial memory and object recognition memory performance in behavioral tests. Insulin mitigated tau and amyloid pathology and hippocampal neuronal loss. SIGNIFICANCE Our findings underscore the potential of intranasal insulin to enhance memory function by modulating hippocampal-prefrontal cortical synchronization and alleviating impairments in hippocampal sharp-wave ripples.
Collapse
Affiliation(s)
- Farnaz Karimani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsaneh Asgari Taei
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Kaveh
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Lee DW, Park HC, Kim DH. Transgenic zebrafish as a model for investigating diabetic peripheral neuropathy: investigation of the role of insulin signaling. Front Cell Neurosci 2024; 18:1441827. [PMID: 39381501 PMCID: PMC11458509 DOI: 10.3389/fncel.2024.1441827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/03/2024] [Indexed: 10/10/2024] Open
Abstract
Diabetic peripheral neuropathy (DPN), a complication of diabetes mellitus (DM), is a neurodegenerative disorder that results from hyperglycemic damage and deficient insulin receptor (IR) signaling in peripheral nerves, triggered by failure of insulin production and insulin resistance. IR signaling plays an important role in nutrient metabolism and synaptic formation and maintenance in peripheral neurons. Although several animal models of DPN have been developed to identify new drug candidates using cytotoxic reagents, nutrient-rich diets, and genetic manipulations, a model showing beneficial effects remains to be established. In this study, we aimed to develop a DPN animal model using zebrafish to validate the effects of drug candidates on sensory neuropathy through in vivo imaging during the early larval stage. To achieve this, we generated Tg (ins:gal4p16);Tg (5uas:epNTR-p2a-mcherry) zebrafish using an enhanced potency nitroreductase (epNTR)-mediated chemogenetic ablation system, which showed highly efficient ablation of pancreatic β-cells following treatment with low-dose metronidazole (MTZ). Using in vivo live imaging, we observed that sensory nerve endings and postsynaptic formation in the peripheral lateral line (PLL) were defective, followed by a disturbance in rheotaxis behavior without any locomotory behavioral changes. Despite defects in sensory nerves and elevated glucose levels, both reactive oxygen species (ROS) levels, a primary cause of DPN, and the number of ganglion cells, remained normal. Furthermore, we found that the activity of mTOR, a downstream target of IR signaling, was decreased in the PLL ganglion cells of the transgenic zebrafish. Our data indicates that peripheral neuropathy results from the loss of IR signaling due to insulin deficiency rather than hyperglycemia alone.
Collapse
Affiliation(s)
- Dong-Won Lee
- Core Research and Development Center, Korea University Ansan Hospital, Ansan, Republic of Korea
- Zebrafish Translational Medical Research Center, College of Medicine, Korea University, Ansan, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, Republic of Korea
| | - Dong Hwee Kim
- Department of Physical Medicine and Rehabilitation, College of Medicine, Korea University, Ansan, Republic of Korea
| |
Collapse
|
5
|
Arda DB, Tunç KC, Bozkurt MF, Bora ES, Çiğel A, Erbaş O. Intranasal Insulin Eases Autism in Rats via GDF-15 and Anti-Inflammatory Pathways. Curr Issues Mol Biol 2024; 46:10530-10544. [PMID: 39329976 PMCID: PMC11431515 DOI: 10.3390/cimb46090624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
In rat models, it is well-documented that chronic administration of propionic acid (PPA) leads to autism-like behaviors. Although the intranasal (IN) insulin approach is predominantly recognized for its effects on food restriction, it has also been shown to enhance cognitive memory by influencing various proteins, modulating anti-inflammatory pathways in the brain, and reducing signaling molecules such as interleukins. This study seeks to explore the potential therapeutic benefits of IN insulin in a rat model of autism induced by PPA. Thirty male Wistar albino rats were categorized into three cohorts: the control group, the PPA-induced autism (250 mg/kg/day intraperitoneal PPA dosage for five days) group, treated with saline via IN, and the PPA-induced autism group, treated with 25 U/kg/day (250 µL/kg/day) insulin via IN. All treatments were administered for 15 days. After behavioral testing, all animals were euthanized, and brain tissue and blood samples were collected for histopathological and biochemical assessments. Following insulin administration, a substantial reduction in autism symptoms was observed in all three social behavior tests conducted on the rats. Moreover, insulin exhibited noteworthy capabilities in decreasing brain MDA, IL-2, IL-17, and TNF-α levels within autism models. Additionally, there is a notable elevation in the brain nerve growth factor level (p < 0.05) and GDF-15 (p < 0.05). The assessment of cell counts within the hippocampal region and cerebellum revealed that insulin displayed effects in decreasing glial cells and inducing a significant augmentation in cell types such as the Purkinje and Pyramidal cells. The administration of insulin via IN exhibits alleviating effects on autism-like behavioral, biochemical, and histopathological alterations induced by PPA in rats. Insulin-dependent protective effects show anti-inflammatory, anti-oxidative, and neuroprotective roles of insulin admitted nasally.
Collapse
Affiliation(s)
- Duygu Burcu Arda
- Department of Pediatrics, Taksim Research and Training Hospital, Istanbul 34365, Türkiye;
| | - Kerem Can Tunç
- Department of Biology, Faculty of Science, Adnan Menderes University, Aydın 09010, Türkiye;
| | - Mehmet Fatih Bozkurt
- Department of Pathology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyon 03100, Türkiye;
| | - Ejder Saylav Bora
- Department of Emergency Medicine, Faculty of Medicine, Izmir Katip Çelebi University, Izmir 35150, Türkiye
| | - Ayşe Çiğel
- Department of Physiology, Faculty of Medicine, Izmir Democracy University, Izmir 35150, Türkiye;
| | - Oytun Erbaş
- Department of Physiology, Faculty of Medicine, Demiroğlu Bilim University, Istanbul 34381, Türkiye;
| |
Collapse
|
6
|
Xu ZQ, Liu TT, Qin QR, Yuan H, Li XM, Qiu CY, Hu WP. Insulin enhances acid-sensing ion channel currents in rat primary sensory neurons. Sci Rep 2024; 14:18077. [PMID: 39103432 PMCID: PMC11300854 DOI: 10.1038/s41598-024-69139-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/01/2024] [Indexed: 08/07/2024] Open
Abstract
Insulin has been shown to modulate neuronal processes through insulin receptors. The ion channels located on neurons may be important targets for insulin/insulin receptor signaling. Both insulin receptors and acid-sensing ion channels (ASICs) are expressed in dorsal root ganglia (DRG) neurons. However, it is still unclear whether there is an interaction between them. Therefore, the purpose of this investigation was to determine the effects of insulin on the functional activity of ASICs. A 5 min application of insulin rapidly enhanced acid-evoked ASIC currents in rat DRG neurons in a concentration-dependent manner. Insulin shifted the concentration-response plot for ASIC currents upward, with an increase of 46.2 ± 7.6% in the maximal current response. The insulin-induced increase in ASIC currents was eliminated by the insulin receptor antagonist GSK1838705, the tyrosine kinase inhibitor lavendustin A, and the phosphatidylinositol-3 kinase antagonist wortmannin. Moreover, insulin increased the number of acid-triggered action potentials by activating insulin receptors. Finally, local administration of insulin exacerbated the spontaneous nociceptive behaviors induced by intraplantar acid injection and the mechanical hyperalgesia induced by intramuscular acid injections through peripheral insulin receptors. These results suggested that insulin/insulin receptor signaling enhanced the functional activity of ASICs via tyrosine kinase and phosphatidylinositol-3 kinase pathways. Our findings revealed that ASICs were targets in primary sensory neurons for insulin receptor signaling, which may underlie insulin modulation of pain.
Collapse
Affiliation(s)
- Zhong-Qing Xu
- School of Pharmacy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Ting-Ting Liu
- School of Pharmacy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Qing-Rui Qin
- School of Pharmacy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Huan Yuan
- School of Pharmacy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Xue-Mei Li
- School of Pharmacy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Chun-Yu Qiu
- School of Pharmacy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Wang-Ping Hu
- School of Pharmacy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China.
- Department of Physiology, Hubei College of Chinese Medicine, 87 Xueyuan Road, Jingzhou, 434020, Hubei, People's Republic of China.
| |
Collapse
|
7
|
Jin Z, Hammoud H, Bhandage AK, Korol SV, Trujeque-Ramos O, Koreli S, Gong Z, Chowdhury AI, Sandbaumhüter FA, Jansson ET, Lindsay RS, Christoffersson G, Andrén PE, Carlsson PO, Bergsten P, Kamali-Moghaddam M, Birnir B. GABA-mediated inhibition of human CD4 + T cell functions is enhanced by insulin but impaired by high glucose levels. EBioMedicine 2024; 105:105217. [PMID: 38943728 PMCID: PMC11260598 DOI: 10.1016/j.ebiom.2024.105217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND γ-aminobutyric acid (GABA), known as the main inhibitory neurotransmitter in the brain, exerts immunomodulatory functions by interaction with immune cells, including T cells. Metabolic programs of T cells are closely linked to their effector functions including proliferation, differentiation, and cytokine production. The physiological molecules glucose and insulin may provide environmental cues and guidance, but whether they coordinate to regulate GABA-mediated T cell immunomodulation is still being examined. METHODS CD4+ T cells that were isolated from blood samples from healthy individuals and from patients with type 1 diabetes (T1D) were activated in vitro. We carried out metabolic assays, multiple proximity extension assay (PEA), ELISA, qPCR, immunoblotting, immunofluorescence staining, flow cytometry analysis, MS-based proteomics, as well as electrophysiology and live-cell Ca2+ imaging. FINDINGS We demonstrate that GABA-mediated reduction of metabolic activity and the release of inflammatory proteins, including IFNγ and IL-10, were abolished in human CD4+ T cells from healthy individuals and patients with T1D when the glucose concentration was elevated above levels typically observed in healthy people. Insulin increased GABAA receptor-subunit ρ2 expression, enhanced the GABAA receptors-mediated currents and Ca2+ influx. GABA decreased, whereas insulin sustained, hexokinase activity and glycolysis in a glucose concentration-dependent manner. INTERPRETATION These findings support that metabolic factors, such as glucose and insulin, influence the GABA-mediated immunomodulation of human primary T cells effector functions. FUNDING The Swedish Children's Diabetes Foundation, The Swedish Diabetes Foundation, The Swedish Research Council 2018-02952, EXODIAB, The Ernfors Foundation, The Thurings Foundation and the Science for Life Laboratory.
Collapse
Affiliation(s)
- Zhe Jin
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Hayma Hammoud
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | | | | | - Stasini Koreli
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Zhitao Gong
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | | | - Erik Tomas Jansson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | | | - Per Erik Andrén
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Peter Bergsten
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Masood Kamali-Moghaddam
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Bryndis Birnir
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
8
|
Ribarič S. The Contribution of Type 2 Diabetes to Parkinson's Disease Aetiology. Int J Mol Sci 2024; 25:4358. [PMID: 38673943 PMCID: PMC11050090 DOI: 10.3390/ijms25084358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Type 2 diabetes (T2D) and Parkinson's disease (PD) are chronic disorders that have a significant health impact on a global scale. Epidemiological, preclinical, and clinical research underpins the assumption that insulin resistance and chronic inflammation contribute to the overlapping aetiologies of T2D and PD. This narrative review summarises the recent evidence on the contribution of T2D to the initiation and progression of PD brain pathology. It also briefly discusses the rationale and potential of alternative pharmacological interventions for PD treatment.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| |
Collapse
|
9
|
García AP, Gaydou L, Pérez E, Barrantes FJ. Insulin resistance induced by long-term hyperinsulinemia abolishes the effects of acute insulin exposure on cell-surface nicotinic acetylcholine receptor levels and actin cytoskeleton morphology. Biochem Biophys Res Commun 2023; 685:149165. [PMID: 37922786 DOI: 10.1016/j.bbrc.2023.149165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023]
Abstract
Using CHO-K1/A5 cells, a clonal cell line that robustly expresses adult muscle-type nicotinic acetylcholine receptor (nAChR), we explored whether insulin resistance in these mammalian cells affects cell-surface expression of the nAChR, its endocytic internalization, and actin cytoskeleton integrity. Acute nanomolar insulin stimulation resulted in a slow increase in nAChR cell-surface levels, reaching maximum levels at ∼1 h. Long periods of insulin incubation caused CHO-K1/A5 cells to become insulin resistant, as previously observed with several other cell types. Furthermore, long-term insulin treatment abolished the effects of short-term insulin exposure on cell-surface nAChR levels, suggestive of a desensitization phenomenon. It also affected the kinetics of ligand-induced nAChR internalization. Since the integrity of the cortical actin cytoskeleton affects nAChR endocytosis, we also studied the effects of long-term insulin treatment on this meshwork. We found that it significantly affected the cortical actin morphology of CHO-K1/A5 cells and the response of the actin cytoskeleton to a subsequent short-term insulin stimulus. Overall, the present results show for the first time the effects of insulin signaling on cell-surface nAChR expression and actin cytoskeleton-associated internalization.
Collapse
Affiliation(s)
- Ana Paula García
- Laboratorio de Neurobiología Molecular, Instituto de Investigaciones Biomédicas (BIOMED) UCA-CONICET, Facultad de Ciencias Médicas, Universidad Católica de Argentina, Buenos Aires, Argentina; Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina
| | - Luisa Gaydou
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina; Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Eugenia Pérez
- Laboratorio de Neurobiología Molecular, Instituto de Investigaciones Biomédicas (BIOMED) UCA-CONICET, Facultad de Ciencias Médicas, Universidad Católica de Argentina, Buenos Aires, Argentina
| | - Francisco J Barrantes
- Laboratorio de Neurobiología Molecular, Instituto de Investigaciones Biomédicas (BIOMED) UCA-CONICET, Facultad de Ciencias Médicas, Universidad Católica de Argentina, Buenos Aires, Argentina.
| |
Collapse
|
10
|
Zhang L, Zhi K, Su Y, Peng W, Meng X. Effect of eIF2α in Neuronal Injury Induced by High Glucose and the Protective Mechanism of Resveratrol. Mol Neurobiol 2023; 60:6043-6059. [PMID: 37410333 DOI: 10.1007/s12035-023-03457-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/21/2023] [Indexed: 07/07/2023]
Abstract
Diabetes mellitus (DM) is a type of metabolic disease characterized by chronic hyperglycemia, which can lead to different degrees of cognitive decline. Therefore, it is crucial to explore the molecular biological mechanisms of neuronal injury. In this study, we investigated the effect of high glucose on eIF2α expression and the mechanism of neuronal injury, and on this basis, the protective mechanism of resveratrol is explored. Treatment with 50 mM high glucose in cortical neurons increased the levels of eIF2α phosphorylation; the expressions of ATF4 and CHOP increased. ISRIB alleviated high glucose-induced neuronal injury by reducing eIF2α phosphorylation when neurons were pretreated with ISRIB before high glucose treatment. Compared with the high glucose-treated group, resveratrol pretreatment reduced eIF2α phosphorylation, the levels of its downstream molecules ATF4 and CHOP, and LDH release. Resveratrol reduced the level of cortical eIF2α phosphorylation and the expression of its downstream molecules in DM mice and improved the ability of spatial memory and learning in DM mice without affecting anxiety and motor performance. Meanwhile, resveratrol modulated the expression of Bcl-2 protein and also effectively decreased the DM-induced up-regulation of Bax, caspase-3, p53, p21, and p16. Taken together, these results suggested that high glucose caused neuronal injury through the eIF2α/ATF4/CHOP pathway which was inhibited by ISRIB and resveratrol. The present study indicates that eIF2α is the new target for the treatment of high glucose-induced neuronal injury, and resveratrol is a potential new medicine to treat diabetes encephalopathy.
Collapse
Affiliation(s)
- Lijing Zhang
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kaining Zhi
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yanfang Su
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenpeng Peng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xianfang Meng
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
11
|
Kumar S, Senapati S, Bhattacharya N, Bhattacharya A, Maurya SK, Husain H, Bhatti JS, Pandey AK. Mechanism and recent updates on insulin-related disorders. World J Clin Cases 2023; 11:5840-5856. [PMID: 37727490 PMCID: PMC10506040 DOI: 10.12998/wjcc.v11.i25.5840] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/06/2023] [Accepted: 08/07/2023] [Indexed: 09/01/2023] Open
Abstract
Insulin, a small protein with 51 amino acids synthesized by pancreatic β-cells, is crucial to sustain glucose homeostasis at biochemical and molecular levels. Numerous metabolic dysfunctions are related to insulin-mediated altered glucose homeostasis. One of the significant pathophysiological conditions linked to the insulin associated disorder is diabetes mellitus (DM) (type 1, type 2, and gestational). Insulin resistance (IR) is one of the major underlying causes of metabolic disorders despite its association with several physiological conditions. Metabolic syndrome (MS) is another pathophysiological condition that is associated with IR, hypertension, and obesity. Further, several other pathophysiological disorders/diseases are associated with the insulin malfunctioning, which include polycystic ovary syndrome, neuronal disorders, and cancer. Insulinomas are an uncommon type of pancreatic β-cell-derived neuroendocrine tumor that makes up 2% of all pancreatic neoplasms. Literature revealed that different biochemical events, molecular signaling pathways, microRNAs, and microbiota act as connecting links between insulin disorder and associated pathophysiology such as DM, insuloma, neurological disorder, MS, and cancer. In this review, we focus on the insulin-related disorders and the underlying mechanisms associated with the pathophysiology.
Collapse
Affiliation(s)
- Shashank Kumar
- Department of Biochemistry, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Sabyasachi Senapati
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Neetu Bhattacharya
- Department of Zoology, Dyal Singh College, University of Delhi, New Delhi 110003, India
| | - Amit Bhattacharya
- Department of Zoology, Ramjas College, University of Delhi, New Delhi 110007, India
| | | | - Hadiya Husain
- Department of Zoology, University of Lucknow, Lucknow 226007, India
| | - Jasvinder Singh Bhatti
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Abhay Kumar Pandey
- Department of Biochemistry, University of Allahabad, Allahabad (Prayagraj) 211002, India
| |
Collapse
|
12
|
Daniel JM, Lindsey SH, Mostany R, Schrader LA, Zsombok A. Cardiometabolic health, menopausal estrogen therapy and the brain: How effects of estrogens diverge in healthy and unhealthy preclinical models of aging. Front Neuroendocrinol 2023; 70:101068. [PMID: 37061205 PMCID: PMC10725785 DOI: 10.1016/j.yfrne.2023.101068] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/23/2023] [Accepted: 04/10/2023] [Indexed: 04/17/2023]
Abstract
Research in preclinical models indicates that estrogens are neuroprotective and positively impact cognitive aging. However, clinical data are equivocal as to the benefits of menopausal estrogen therapy to the brain and cognition. Pre-existing cardiometabolic disease may modulate mechanisms by which estrogens act, potentially reducing or reversing protections they provide against cognitive decline. In the current review we propose mechanisms by which cardiometabolic disease may alter estrogen effects, including both alterations in actions directly on brain memory systems and actions on cardiometabolic systems, which in turn impact brain memory systems. Consideration of mechanisms by which estrogen administration can exert differential effects dependent upon health phenotype is consistent with the move towards precision or personalized medicine, which aims to determine which treatment interventions will work for which individuals. Understanding effects of estrogens in both healthy and unhealthy models of aging is critical to optimizing the translational link between preclinical and clinical research.
Collapse
Affiliation(s)
- Jill M Daniel
- Department of Psychology and Brain Institute, Tulane University, New Orleans, LA, United States.
| | - Sarah H Lindsey
- Department of Pharmacology and Brain Institute, Tulane University, New Orleans, LA, United States
| | - Ricardo Mostany
- Department of Pharmacology and Brain Institute, Tulane University, New Orleans, LA, United States
| | - Laura A Schrader
- Department of Cell & Molecular Biology and Brain Institute, Tulane University, New Orleans, LA, United States
| | - Andrea Zsombok
- Department of Physiology and Brain Institute, Tulane University, New Orleans, LA, United States
| |
Collapse
|
13
|
de Bartolomeis A, De Simone G, De Prisco M, Barone A, Napoli R, Beguinot F, Billeci M, Fornaro M. Insulin effects on core neurotransmitter pathways involved in schizophrenia neurobiology: a meta-analysis of preclinical studies. Implications for the treatment. Mol Psychiatry 2023; 28:2811-2825. [PMID: 37085712 PMCID: PMC10615753 DOI: 10.1038/s41380-023-02065-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/23/2023]
Abstract
Impairment of insulin action and metabolic dysregulation have traditionally been associated with schizophrenia, although the molecular basis of such association remains still elusive. The present meta-analysis aims to assess the impact of insulin action manipulations (i.e., hyperinsulinemia, hypoinsulinemia, systemic or brain insulin resistance) on glutamatergic, dopaminergic, γ-aminobutyric acid (GABA)ergic, and serotonergic pathways in the central nervous system. More than one hundred outcomes, including transcript or protein levels, kinetic parameters, and other components of the neurotransmitter pathways, were collected from cultured cells, animals, or humans, and meta-analyzed by applying a random-effects model and adopting Hedges'g to compare means. Two hundred fifteen studies met the inclusion criteria, of which 180 entered the quantitative synthesis. Significant impairments in key regulators of synaptic plasticity processes were detected as the result of insulin handlings. Specifically, protein levels of N-methyl-D-aspartate receptor (NMDAR) subunits including type 2A (NR2A) (Hedges' g = -0.95, 95%C.I. = -1.50, -0.39; p = 0.001; I2 = 47.46%) and 2B (NR2B) (Hedges'g = -0.69, 95%C.I. = -1.35, -0.02; p = 0.043; I2 = 62.09%), and Postsynaptic density protein 95 (PSD-95) (Hedges'g = -0.91, 95%C.I. = -1.51, -0.32; p = 0.003; I2 = 77.81%) were found reduced in insulin-resistant animal models. Moreover, insulin-resistant animals showed significantly impaired dopamine transporter activity, whereas the dopamine D2 receptor mRNA expression (Hedges'g = 3.259; 95%C.I. = 0.497, 6.020; p = 0.021; I2 = 90.61%) increased under insulin deficiency conditions. Insulin action modulated glutamate and GABA release, as well as several enzymes involved in GABA and serotonin synthesis. These results suggest that brain neurotransmitter systems are susceptible to insulin signaling abnormalities, resembling the discrete psychotic disorders' neurobiology and possibly contributing to the development of neurobiological hallmarks of treatment-resistant schizophrenia.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Odontostomatology University of Naples "Federico II", School of Medicine, Via Pansini 5, 80131, Naples, Italy.
| | - Giuseppe De Simone
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Odontostomatology University of Naples "Federico II", School of Medicine, Via Pansini 5, 80131, Naples, Italy
| | - Michele De Prisco
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Odontostomatology University of Naples "Federico II", School of Medicine, Via Pansini 5, 80131, Naples, Italy
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, 170 Villarroel st, 12-0, 08036, Barcelona, Catalonia, Spain
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Odontostomatology University of Naples "Federico II", School of Medicine, Via Pansini 5, 80131, Naples, Italy
| | - Raffaele Napoli
- Department of Translational Medical Sciences, University of Naples "Federico II", Via S. Pansini 5, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medical Sciences, University of Naples "Federico II", Via S. Pansini 5, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Martina Billeci
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Odontostomatology University of Naples "Federico II", School of Medicine, Via Pansini 5, 80131, Naples, Italy
| | - Michele Fornaro
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Odontostomatology University of Naples "Federico II", School of Medicine, Via Pansini 5, 80131, Naples, Italy
| |
Collapse
|
14
|
Nakaya Y, Kosukegawa S, Kobayashi S, Hirose K, Kitano K, Mayahara K, Takei H, Motoyoshi M, Kobayashi M. Insulin potentiates inhibitory synaptic currents between fast-spiking and pyramidal neurons in the rat insular cortex. Neuropharmacology 2023:109649. [PMID: 37393988 DOI: 10.1016/j.neuropharm.2023.109649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 07/04/2023]
Abstract
Insulin plays roles in brain functions such as neural development and plasticity and is reported to be involved in dementia and depression. However, little information is available on the insulin-mediated modulation of electrophysiological activities, especially in the cerebral cortex. This study examined how insulin modulates the neural activities of inhibitory neurons and inhibitory postsynaptic currents (IPSCs) in rat insular cortex (IC; either sex) by multiple whole-cell patch-clamp recordings. We demonstrated that insulin increased the repetitive spike firing rate with a decrease in the threshold potential without changing the resting membrane potentials and input resistance of fast-spiking GABAergic neurons (FSNs). Next, we found a dose-dependent enhancement of unitary IPSCs (uIPSCs) by insulin in the connections from FSNs to pyramidal neurons (PNs). The insulin-induced enhancement of uIPSCs accompanied decreases in the paired-pulse ratio, suggesting that insulin increases GABA release from presynaptic terminals. The finding of miniature IPSC recordings of the increased frequency without changing the amplitude supports this hypothesis. Insulin had little effect on uIPSCs under the coapplication of S961, an insulin receptor antagonist, or lavendustin A, an inhibitor of tyrosine kinase. The PI3-K inhibitor wortmannin or the PKB/Akt inhibitors, deguelin and Akt inhibitor VIII, blocked the insulin-induced enhancement of uIPSCs. Intracellular application of Akt inhibitor VIII to presynaptic FSNs also blocked insulin-induced enhancement of uIPSCs. In contrast, uIPSCs were enhanced by insulin in combination with the MAPK inhibitor PD98059. These results suggest that insulin facilitates the inhibition of PNs by increases in FSN firing frequency and IPSCs from FSNs to PNs. (250 words).
Collapse
Affiliation(s)
- Yuka Nakaya
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Satoshi Kosukegawa
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Department of Orthodontics, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Satomi Kobayashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Department of Biology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Kensuke Hirose
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Department of Pedodontics, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Kouhei Kitano
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Kotoe Mayahara
- Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Department of Orthodontics, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Hiroki Takei
- Department of Dentistry, Saitama Prefectural Children's Medical Center, 1-2, Shintoshin, Chuo-ku, Saitama-shi, 3330-8777, Japan
| | - Mitsuru Motoyoshi
- Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Department of Orthodontics, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Masayuki Kobayashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.
| |
Collapse
|
15
|
Gupta M, Pandey S, Rumman M, Singh B, Mahdi AA. Molecular mechanisms underlying hyperglycemia associated cognitive decline. IBRO Neurosci Rep 2023; 14:57-63. [PMID: 36590246 PMCID: PMC9800261 DOI: 10.1016/j.ibneur.2022.12.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus (DM) is a metabolic disease characterized by chronic hyperglycemia. DM can lead to a number of secondary complications affecting multiple organs in the body including the eyes, kidney, heart, and brain. The most common effect of hyperglycemia on the brain is cognitive decline. It has been estimated that 20-70% of people with DM have cognitive deficits. High blood sugar affects key brain areas involved in learning, memory, and spatial navigation, and the structural complexity of the brain has made it prone to a variety of pathological disorders, including T2DM. Studies have reported that cognitive decline can occur in people with diabetes, which could go undetected for several years. Moreover, studies on brain imaging suggest extensive effects on different brain regions in patients with T2D. It remains unclear whether diabetes-associated cognitive decline is a consequence of hyperglycemia or a complication that co-occurs with T2D. The exact mechanism underlying cognitive impairment in diabetes is complex; however, impaired glucose metabolism and abnormal insulin function are thought to play important roles. In this review, we have tried to summarize the effect of hyperglycemia on the brain structure and functions, along with the potential mechanisms underlying T2DM-associated cognitive decline.
Collapse
Affiliation(s)
- Mrinal Gupta
- Department of Biochemistry, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Shivani Pandey
- Department of Biochemistry, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Mohammad Rumman
- Department of Biochemistry, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Babita Singh
- Department of Biochemistry, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Abbas Ali Mahdi
- Department of Biochemistry, King George’s Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
16
|
Villalobos N, Ramírez-Sánchez E, Mondragón-García A, Garduño J, Castillo-Rolón D, Trujeque-Ramos S, Hernández-López S. Insulin decreases epileptiform activity in rat layer 5/6 prefrontal cortex in vitro. Synapse 2023; 77:e22263. [PMID: 36732015 DOI: 10.1002/syn.22263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 02/04/2023]
Abstract
Accumulating evidence indicates that insulin-mediated signaling in the brain may play important roles in regulating neuronal function. Alterations to insulin signaling are associated with the development of neurological disorders including Alzheimer's disease and Parkinson's disease. Also, hyperglycemia and insulin resistance have been associated with seizure activity and brain injury. In recent work, we found that insulin increased inhibitory GABAA -mediated tonic currents in the prefrontal cortex (PFC). In this work, we used local field potential recordings and calcium imaging to investigate the effect of insulin on seizure-like activity in PFC slices. Seizure-like events (SLEs) were induced by perfusing the slices with magnesium-free artificial cerebrospinal fluid (ACSF) containing the proconvulsive compound 4-aminopyridine (4-AP). We found that insulin decreased the frequency, amplitude, and duration of SLEs as well as the synchronic activity of PFC neurons evoked by 4-AP. These insulin effects were mediated by the PI3K/Akt signaling pathway and mimicked by gaboxadol (THIP), a δ GABAA receptor agonist. The effect of insulin on the number of SLEs was partially blocked by L-655,708, an inverse agonist with high selectivity for GABAA receptors containing the α5 subunit. Our results suggest that insulin reduces neuronal excitability by an increase of GABAergic tonic currents. The physiological relevance of these findings is discussed.
Collapse
Affiliation(s)
- N Villalobos
- Academia de Fisiología, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Colonia Casco de Santo Tomás, Ciudad de México, México
- Sección de Estudios de Posgrado e Investigación de la Escuela Superior de Medicina del IPN, Plan de San Luis y Díaz Mirón, Colonia Casco de Santo Tomás, Ciudad de México, México
| | - E Ramírez-Sánchez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - A Mondragón-García
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - J Garduño
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - D Castillo-Rolón
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - S Trujeque-Ramos
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
- Translational Neurogenetics Unit, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - S Hernández-López
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
17
|
Zhou H, Rao Z, Zhang Z, Zhou J. Function of the GABAergic System in Diabetic Encephalopathy. Cell Mol Neurobiol 2023; 43:605-619. [PMID: 35460435 PMCID: PMC11415196 DOI: 10.1007/s10571-022-01214-7] [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: 08/25/2021] [Accepted: 03/17/2022] [Indexed: 11/03/2022]
Abstract
Diabetes is a common metabolic disease characterized by loss of blood sugar control and a high rate of complications. γ-Aminobutyric acid (GABA) functions as the primary inhibitory neurotransmitter in the adult mammalian brain. The normal function of the GABAergic system is affected in diabetes. Herein, we summarize the role of the GABAergic system in diabetic cognitive dysfunction, diabetic blood sugar control disorders, diabetes-induced peripheral neuropathy, diabetic central nervous system damage, maintaining diabetic brain energy homeostasis, helping central control of blood sugar and attenuating neuronal oxidative stress damage. We show the key regulatory role of the GABAergic system in multiple comorbidities in patients with diabetes and hope that further studies elucidating the role of the GABAergic system will yield benefits for the treatment and prevention of comorbidities in patients with diabetes.
Collapse
Affiliation(s)
- Hongli Zhou
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Zhili Rao
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, People's Republic of China
| | - Zuo Zhang
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Jiyin Zhou
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, 400037, People's Republic of China.
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, People's Republic of China.
| |
Collapse
|
18
|
Ezkurdia A, Ramírez MJ, Solas M. Metabolic Syndrome as a Risk Factor for Alzheimer's Disease: A Focus on Insulin Resistance. Int J Mol Sci 2023; 24:ijms24054354. [PMID: 36901787 PMCID: PMC10001958 DOI: 10.3390/ijms24054354] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Alzheimer's disease (AD) is the main type of dementia and is a disease with a profound socioeconomic burden due to the lack of effective treatment. In addition to genetics and environmental factors, AD is highly associated with metabolic syndrome, defined as the combination of hypertension, hyperlipidemia, obesity and type 2 diabetes mellitus (T2DM). Among these risk factors, the connection between AD and T2DM has been deeply studied. It has been suggested that the mechanism linking both conditions is insulin resistance. Insulin is an important hormone that regulates not only peripheral energy homeostasis but also brain functions, such as cognition. Insulin desensitization, therefore, could impact normal brain function increasing the risk of developing neurodegenerative disorders in later life. Paradoxically, it has been demonstrated that decreased neuronal insulin signalling can also have a protective role in aging and protein-aggregation-associated diseases, as is the case in AD. This controversy is fed by studies focused on neuronal insulin signalling. However, the role of insulin action on other brain cell types, such as astrocytes, is still unexplored. Therefore, it is worthwhile exploring the involvement of the astrocytic insulin receptor in cognition, as well as in the onset and/or development of AD.
Collapse
Affiliation(s)
- Amaia Ezkurdia
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - María J. Ramírez
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Maite Solas
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Correspondence:
| |
Collapse
|
19
|
Hamzé R, Delangre E, Tolu S, Moreau M, Janel N, Bailbé D, Movassat J. Type 2 Diabetes Mellitus and Alzheimer's Disease: Shared Molecular Mechanisms and Potential Common Therapeutic Targets. Int J Mol Sci 2022; 23:ijms232315287. [PMID: 36499613 PMCID: PMC9739879 DOI: 10.3390/ijms232315287] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
The global prevalence of diabetes mellitus and Alzheimer's disease is increasing alarmingly with the aging of the population. Numerous epidemiological data suggest that there is a strong association between type 2 diabetes and an increased risk of dementia. These diseases are both degenerative and progressive and share common risk factors. The amyloid cascade plays a key role in the pathophysiology of Alzheimer's disease. The accumulation of amyloid beta peptides gradually leads to the hyperphosphorylation of tau proteins, which then form neurofibrillary tangles, resulting in neurodegeneration and cerebral atrophy. In Alzheimer's disease, apart from these processes, the alteration of glucose metabolism and insulin signaling in the brain seems to induce early neuronal loss and the impairment of synaptic plasticity, years before the clinical manifestation of the disease. The large amount of evidence on the existence of insulin resistance in the brain during Alzheimer's disease has led to the description of this disease as "type 3 diabetes". Available animal models have been valuable in the understanding of the relationships between type 2 diabetes and Alzheimer's disease, but to date, the mechanistical links are poorly understood. In this non-exhaustive review, we describe the main molecular mechanisms that may link these two diseases, with an emphasis on impaired insulin and IGF-1 signaling. We also focus on GSK3β and DYRK1A, markers of Alzheimer's disease, which are also closely associated with pancreatic β-cell dysfunction and type 2 diabetes, and thus may represent common therapeutic targets for both diseases.
Collapse
Affiliation(s)
- Rim Hamzé
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Etienne Delangre
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Stefania Tolu
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Manon Moreau
- Team Degenerative Process, Stress and Aging, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Nathalie Janel
- Team Degenerative Process, Stress and Aging, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Danielle Bailbé
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Jamileh Movassat
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
- Correspondence: ; Tel.: +33-1-57-27-77-82; Fax: +33-1-57-27-77-91
| |
Collapse
|
20
|
Wang X, Cui X, Li Y, Li F, Li Y, Dai J, Hu H, Wang X, Sun J, Yang Y, Zhang S. MC4R Deficiency Causes Dysregulation of Postsynaptic Excitatory Synaptic Transmission as a Crucial Culprit for Obesity. Diabetes 2022; 71:2331-2343. [PMID: 35926095 DOI: 10.2337/db22-0162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 07/30/2022] [Indexed: 11/13/2022]
Abstract
Melanocortin 4 receptor (MC4R) in the paraventricular nucleus of the hypothalamus (PVH) shows bidirectional characterization in modulating food intake and energy homeostasis. We demonstrate that MC4R knockdown (KD) in the PVH can attenuate AMPA receptor (AMPAR)-mediated postsynaptic responses by altering the phosphorylation of AMPAR GluA1 subunit through the protein kinase A (PKA)-dependent signaling cascade and simultaneously lead to rapid body weight gain. Furthermore, PKA KD in the PVH engendered similar electrophysiological and behavioral phenotypes as in MC4R KD mice. Importantly, we observed that the reduction of AMPAR GluA1 expression not only led to attenuated synaptic responses but also caused body weight gain, suggesting that the aberration of synaptic responses may be one of the crucial pathogeny of obesity. Our study provides the synaptic and molecular explanations of how body weight is regulated by MC4R in the PVH.
Collapse
Affiliation(s)
- Xiaohui Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Cui
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yang Li
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yue Li
- Guangwai Community Health Service Center of Xicheng District, Beijing, China
| | - Jinye Dai
- Howard Hughes Medical Institute and Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA
| | - Han Hu
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuefeng Wang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianyuan Sun
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Beijing, China
| | - Yan Yang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuli Zhang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
21
|
Chan ES, Ge Y, So YW, Bai YF, Liu L, Wang YT. Allosteric potentiation of GABAA receptor single-channel conductance by netrin-1 during neuronal-excitation-induced inhibitory synaptic homeostasis. Cell Rep 2022; 41:111584. [DOI: 10.1016/j.celrep.2022.111584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/13/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
|
22
|
Chen H, Wang M, Xia L, Dong J, Xu G, Wang Z, Feng L, Zhou Y. New Evidence of Central Nervous System Damage in Diabetes: Impairment of Fine Visual Discrimination. Diabetes 2022; 71:1772-1784. [PMID: 35612428 DOI: 10.2337/db21-0715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 05/04/2022] [Indexed: 11/13/2022]
Abstract
Diabetes can damage both the peripheral sensory organs, causing retinopathy, and the central visual system, leading to contrast sensitivity and impaired color vision in patients without retinopathy. Orientation discrimination is important for shape recognition by the visual system. Our psychophysical findings in this study show diminished orientation discrimination in patients with diabetes without retinopathy. To reveal the underlying mechanism, we established a diabetic mouse model and recorded in vivo electrophysiological data in the dorsal lateral geniculate nucleus (dLGN) and primary visual cortex (V1). Reduced orientation selectivity was observed in both individual and populations of neurons in V1 and dLGN, which increased in severity with disease duration. This diabetes-associated neuronal dysfunction appeared earlier in the V1 than dLGN. Additionally, neuronal activity and signal-to-noise ratio are reduced in V1 neurons of diabetic mice, leading to a decreased capacity for information processing by V1 neurons. Notably, the V1 in diabetic mice exhibits reduced excitatory neuronal activity and lower levels of phosphorylated mammalian target of rapamycin (mTOR). Our findings show that altered responses of both populations of and single V1 neurons may impair fine vision, thus expanding our understanding of the underlying causes of diabetes-related impairment of the central nervous system.
Collapse
Affiliation(s)
- He Chen
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Menghan Wang
- School of Information Science and Technology, University of Science and Technology of China, Hefei, China
| | - Lin Xia
- Department of Ophthalmology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Branch of National Clinical Research Center for Ocular Diseases, Hefei, China
| | - Jiong Dong
- Department of Ophthalmology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- State Key Laboratory of Brain and Cognitive Science, Institute of Psychology, Chinese Academy of Sciences, Hefei, China
| | - Guangwei Xu
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Ziyi Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Lixia Feng
- Department of Ophthalmology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Branch of National Clinical Research Center for Ocular Diseases, Hefei, China
| | - Yifeng Zhou
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
- State Key Laboratory of Brain and Cognitive Science, Institute of Psychology, Chinese Academy of Sciences, Hefei, China
| |
Collapse
|
23
|
Grassi G, Figee M, Pozza A, Dell'Osso B. Obsessive-compulsive disorder, insulin signaling and diabetes - A novel form of physical health comorbidity: The sweet compulsive brain. Compr Psychiatry 2022; 117:152329. [PMID: 35679658 DOI: 10.1016/j.comppsych.2022.152329] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/03/2022] [Accepted: 05/17/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND While a growing body of research highlights a bi-directional link between diabetes and mood disorders, little is known about the relationship between diabetes and obsessive-compulsive disorder (OCD). The aim of the present review is to investigate current evidence linking OCD, insulin-signaling and diabetes. METHODS A PubMed search was conducted to review all the available studies assessing diabetes, glucose metabolism and insulin-signaling in OCD patients and vice versa. RESULTS Some clinical and epidemiological studies show a higher prevalence of diabetes in OCD and vice versa compared to the general population. Animal and genetic studies suggest a possible role of insulin-signaling in the pathophysiology of OCD. Deep brain stimulation (DBS) studies suggest that abnormal dopaminergic transmission in the striatum may contribute to impaired insulin sensitivity in OCD. While DBS seems to increase insulin sensitivity, a possible protective role of serotonin reuptake-inhibitors on diabetic risk needs further studies. CONCLUSION Despite their preliminary nature, these data highlight the importance of further investigations aimed at assessing metabolic features in OCD patients and OCD symptoms in diabetes patients to understand the impact of each condition on the pathophysiology and course of the other. Understanding the role of insulin in the obsessive-compulsive brain could open new treatment pathways for OCD.
Collapse
Affiliation(s)
| | - Martijn Figee
- Department of Psychiatry, Icahn Medical School at Mount Sinai, New York, NY, USA
| | | | - Bernardo Dell'Osso
- University of Milan, Department of Biomedical and Clinical Sciences Luigi Sacco, Ospedale Sacco-Polo Universitario, ASST Fatebenefratelli-Sacco, Milan, Italy
| |
Collapse
|
24
|
Mohseni-Moghaddam P, Ghobadian R, Khaleghzadeh-Ahangar H. Dementia in Diabetes mellitus and Atherosclerosis; Two Interrelated Systemic Diseases. Brain Res Bull 2022; 181:87-96. [DOI: 10.1016/j.brainresbull.2022.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/18/2021] [Accepted: 01/24/2022] [Indexed: 12/06/2022]
|
25
|
Alkasabera A, Onyali CB, Anim-Koranteng C, Shah HE, Ethirajulu A, Bhawnani N, Mostafa JA. The Effect of Type-2 Diabetes on Cognitive Status and the Role of Anti-diabetes Medications. Cureus 2021; 13:e19176. [PMID: 34877187 PMCID: PMC8642129 DOI: 10.7759/cureus.19176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/01/2021] [Indexed: 12/11/2022] Open
Abstract
Type-2 diabetes mellitus prevalence is constantly increasing; this is explained by the increase of its risk factors and the amelioration of its management. Therefore, people are living longer with diabetes mellitus, which, in turn, has revealed new complications of the disease. Dementia is represented mainly by Alzheimer's disease and is an interesting topic of study. Accordingly, statistics have shown that dementia incidence is doubled in diabetic patients. The establishment of a relation between type-2 diabetes mellitus was studied on several levels in both humans and animal subjects. First, insulin receptors were found in the brain, especially the hippocampus, and insulin transport to the brain is mainly accomplished through the blood-brain barrier. Secondly, several studies showed that insulin affects multiple neurotransmitters in favor of promoting memory and cognition status. Thirdly, multiple pathological studies showed that insulin and Alzheimer's disease share many common lesions in the brain, such as beta-amyloid plaques, amylin-Aβ plaques, hyper-phosphorylated tau protein, and brain atrophy, especially in the hippocampus. After recognizing the positive effect of insulin on cognitive status, and the harmful effect of insulin resistance on cognitive status, multiple studies were focused on the role of anti-diabetes medications in fighting dementia. Consequently, these studies showed a positive impact of oral anti-diabetes medication, as well as insulin in limiting the progression of dementia and promoting cognitive status. Moreover, their effects were also noticed on limiting the pathological lesions of Alzheimer's disease. Accordingly, we can consider type-2 diabetes mellitus as a risk factor for dementia and Alzheimer's disease. Therefore, this can be used on the pharmaceutical level by the promising implication of antidiabetics as a treatment of dementia and Alzheimer's disease or at least to limit its progression. However, multiple clinical studies should be dedicated to proving the true benefits of anti-diabetes medications in treating dementia before they can be used in reality.
Collapse
Affiliation(s)
- Almothana Alkasabera
- General Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | | | | | - Hira E Shah
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Aarthi Ethirajulu
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Nitin Bhawnani
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Jihan A Mostafa
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
26
|
Wagner K, Smylla TK, Lampe M, Krieg J, Huber A. Phospholipase D and retromer promote recycling of TRPL ion channel via the endoplasmic reticulum. Traffic 2021; 23:42-62. [PMID: 34719094 DOI: 10.1111/tra.12824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 12/27/2022]
Abstract
Plasma membrane protein trafficking is of fundamental importance for cell function and cell integrity of neurons and includes regulated protein recycling. In this work, we report a novel role of the endoplasmic reticulum (ER) for protein recycling as discovered in trafficking studies of the ion channel TRPL in photoreceptor cells of Drosophila. TRPL is located within the rhabdomeric membrane from where it is endocytosed upon light stimulation and stored in the cell body. Conventional immunohistochemistry as well as stimulated emission depletion super-resolution microscopy revealed TRPL storage at the ER after illumination, suggesting an unusual recycling route of TRPL. Our results also imply that both phospholipase D (PLD) and retromer complex are required for correct recycling of TRPL to the rhabdomeric membrane. Loss of PLD activity in PLD3.1 mutants results in enhanced degradation of TRPL. In the retromer mutant vps35MH20 , TRPL is trapped in a Rab5-positive compartment. Evidenced by epistatic analysis in the double mutant PLD3.1 vps35MH20 , PLD activity precedes retromer function. We propose a model in which PLD and retromer function play key roles in the transport of TRPL to an ER enriched compartment.
Collapse
Affiliation(s)
- Krystina Wagner
- Department of Biochemistry, University of Hohenheim, Institute of Biology, Stuttgart, Germany
| | - Thomas K Smylla
- Department of Biochemistry, University of Hohenheim, Institute of Biology, Stuttgart, Germany
| | - Marko Lampe
- European Molecular Biology Laboratory, Advanced Light Microscopy Core Facility, Heidelberg, Germany
| | - Jana Krieg
- Department of Biochemistry, University of Hohenheim, Institute of Biology, Stuttgart, Germany
| | - Armin Huber
- Department of Biochemistry, University of Hohenheim, Institute of Biology, Stuttgart, Germany
| |
Collapse
|
27
|
Wong YH, Wong SH, Wong XT, Yi Yap Q, Yip KY, Wong LZ, Chellappan DK, Bhattamisra SK, Candasamy M. Genetic associated complications of type 2 Diabetes Mellitus: a review. Panminerva Med 2021; 64:274-288. [PMID: 34609116 DOI: 10.23736/s0031-0808.21.04285-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
According to the International Diabetes Federation, the number of adults (age of 20-79) being diagnosed with Diabetes Mellitus (DM) have increased from 285 million in year 2009 to 463 million in year 2019 which comprises of 95% Type 2 DM patient (T2DM). Research have claimed that genetic predisposition could be one of the factors causing T2DM complications. In addition, T2DMcomplications cause an incremental risk to mortality. Therefore, this article aims to discuss some complications of T2DM in and their genetic association. The complications that are discussed in this article are diabetic nephropathy, diabetes induced cardiovascular disease, diabetic neuropathy, Diabetic Foot Ulcer (DFU) and Alzheimer's disease. According to the information obtained, genes associated with diabetic nephropathy (DN) are gene GABRR1 and ELMO1 that cause injury to glomerular. Replication of genes FRMD3, CARS and MYO16/IRS2 shown to have link with DN. The increase of gene THBS2, NGAL, PIP, TRAF6 polymorphism, ICAM-1 encoded for rs5498 polymorphism and C667T increase susceptibility towards DN in T2DM patient. Genes associated with cardiovascular diseases are Adiponectin gene (ACRP30) and Apolipoprotein E (APOE) polymorphism gene with ξ2 allele. Haptoglobin (Hp) 1-1 genotype and Mitochondria Superoxide Dismutase 2 (SOD2) plays a role in cardiovascular events. As for genes related to diabetic neuropathy, Janus Kinase (JAK), mutation of SCN9A and TRPA1 gene and destruction of miRNA contribute to pathogenesis of diabetic neuropathy among T2DM patients. Expression of cytokine IL-6, IL-10, miR-146a are found to cause diabetic neuropathy. Besides, A1a16Va1 gene polymorphism, an oxidative stress influence was found as one of the gene factors. Diabetic retinopathy (DR) is believed to have association with Monocyte Chemoattractant Protein-1 (MCP-1) and Insulin-like Growth Factor 1 (IGF1). Over-expression of gene ENPP1, IL-6 pro-inflammatory cytokine, ARHGAP22's protein rs3844492 polymorphism and TLR4 heterozygous genotype are contributing to significant pathophysiological process causing DR, while research found increases level of UCP1 gene protects retina cells from oxidative stress. Diabetic Foot Ulcer (DFU) is manifested by slowing in reepithelialisation of keratinocyte, persistence wound inflammation and healing impairment. Reepithelialisation disturbance was caused by E2F3 gene, reduction of Tacl gene encoded substance P causing persistence inflammation while expression of MMp-9 polymorphism contributes to healing impairment. A decrease in HIF-1a gene expression leads to increased risk of pathogenesis, while downregulation of TLR2 increases severity of wound in DFU patients. SNPs alleles has been shown to have significant association between the genetic dispositions of T2DM and Alzheimer's disease (AD). The progression of AD can be due to the change in DNA methylation of CLOCK gene, followed with worsening of AD by APOE4 gene due to dyslipidaemia condition in T2DM patients. Insulin resistance is also a factor that contributes to pathogenesis of AD.
Collapse
Affiliation(s)
- Yee H Wong
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Shen H Wong
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Xiao T Wong
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Qiao Yi Yap
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Khar Y Yip
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Liang Z Wong
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Subrat K Bhattamisra
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Mayuren Candasamy
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia -
| |
Collapse
|
28
|
Bruxel EM, do Canto AM, Bruno DCF, Geraldis JC, Lopes-Cendes I. Multi-omic strategies applied to the study of pharmacoresistance in mesial temporal lobe epilepsy. Epilepsia Open 2021; 7 Suppl 1:S94-S120. [PMID: 34486831 PMCID: PMC9340306 DOI: 10.1002/epi4.12536] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 12/19/2022] Open
Abstract
Mesial temporal lobe epilepsy (MTLE) is the most common type of focal epilepsy in adults, and hippocampal sclerosis (HS) is a frequent histopathological feature in patients with MTLE. Pharmacoresistance is present in at least one-third of patients with MTLE with HS (MTLE+HS). Several hypotheses have been proposed to explain the mechanisms of pharmacoresistance in epilepsy, including the effect of genetic and molecular factors. In recent years, the increased knowledge generated by high-throughput omic technologies has significantly improved the power of molecular genetic studies to discover new mechanisms leading to disease and response to treatment. In this review, we present and discuss the contribution of different omic modalities to understand the basic mechanisms determining pharmacoresistance in patients with MTLE+HS. We provide an overview and a critical discussion of the findings, limitations, new approaches, and future directions of these studies to improve the understanding of pharmacoresistance in MTLE+HS. However, it is important to point out that, as with other complex traits, pharmacoresistance to anti-seizure medications is likely a multifactorial condition in which gene-gene and gene-environment interactions play an important role. Thus, studies using multidimensional approaches are more likely to unravel these intricate biological processes.
Collapse
Affiliation(s)
- Estela M Bruxel
- Departments of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| | - Amanda M do Canto
- Departments of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| | - Danielle C F Bruno
- Departments of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| | - Jaqueline C Geraldis
- Departments of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| | - Iscia Lopes-Cendes
- Departments of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| |
Collapse
|
29
|
Flintoff J, Kesby JP, Siskind D, Burne TH. Treating cognitive impairment in schizophrenia with GLP-1RAs: an overview of their therapeutic potential. Expert Opin Investig Drugs 2021; 30:877-891. [PMID: 34213981 DOI: 10.1080/13543784.2021.1951702] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Schizophrenia is a neuropsychiatric disorder that affects approximately 1% of individuals worldwide. There are no available medications to treat cognitive impairment in this patient population currently. Preclinical evidence suggests that glucagon-like peptide-1 receptor agonists (GLP-1 RAs) improve cognitive function. There is a need to evaluate how GLP-1 RAs alter specific domains of cognition and whether they will be of therapeutic benefit in individuals with schizophrenia. AREAS COVERED This paper summarizes the effects of GLP-1 RAs on metabolic processes in the brain and how these mechanisms relate to improved cognitive function. We provide an overview of preclinical studies that demonstrate GLP-1 RAs improve cognition and comment on their potential therapeutic benefit in individuals with schizophrenia. EXPERT OPINION To understand the benefits of GLP-1 RAs in individuals with schizophrenia, further preclinical research with rodent models relevant to schizophrenia symptomology are needed. Moreover, preclinical studies must focus on using a wider range of behavioral assays to understand whether important aspects of cognition such as executive function, attention, and goal-directed behavior are improved using GLP-1 RAs. Further research into the specific mechanisms of how GLP-1 RAs affect cognitive function and their interactions with antipsychotic medication commonly prescribed is necessary.
Collapse
Affiliation(s)
- Jonathan Flintoff
- Queensland Brain Institute, the University of Queensland, St Lucia, QLD, Australia
| | - James P Kesby
- Queensland Brain Institute, the University of Queensland, St Lucia, QLD, Australia.,QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Dan Siskind
- Queensland Centre for Mental Health Research, Wacol, QLD, Australia.,Metro South Addiction and Mental Health Service, Woolloongabba, QLD, Australia
| | - Thomas Hj Burne
- Queensland Brain Institute, the University of Queensland, St Lucia, QLD, Australia.,Queensland Centre for Mental Health Research, Wacol, QLD, Australia
| |
Collapse
|
30
|
Hammoud H, Netsyk O, Tafreshiha AS, Korol SV, Jin Z, Li J, Birnir B. Insulin differentially modulates GABA signalling in hippocampal neurons and, in an age-dependent manner, normalizes GABA-activated currents in the tg-APPSwe mouse model of Alzheimer's disease. Acta Physiol (Oxf) 2021; 232:e13623. [PMID: 33559388 DOI: 10.1111/apha.13623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023]
Abstract
AIM We examined if tonic γ-aminobutyric acid (GABA)-activated currents in primary hippocampal neurons were modulated by insulin in wild-type and tg-APPSwe mice, an Alzheimer's disease (AD) model. METHODS GABA-activated currents were recorded in dentate gyrus (DG) granule cells and CA3 pyramidal neurons in hippocampal brain slices, from 8 to 10 weeks old (young) wild-type mice and in dorsal DG granule cells in adult, 5-6 and 10-12 (aged) months old wild-type and tg-APPSwe mice, in the absence or presence of insulin, by whole-cell patch-clamp electrophysiology. RESULTS In young mice, insulin (1 nmol/L) enhanced the total spontaneous inhibitory postsynaptic current (sIPSCT ) density in both dorsal and ventral DG granule cells. The extrasynaptic current density was only increased by insulin in dorsal CA3 pyramidal neurons. In absence of action potentials, insulin enhanced DG granule cells and dorsal CA3 pyramidal neurons miniature IPSC (mIPSC) frequency, consistent with insulin regulation of presynaptic GABA release. sIPSCT densities in DG granule cells were similar in wild-type and tg-APPSwe mice at 5-6 months but significantly decreased in aged tg-APPSwe mice where insulin normalized currents to wild-type levels. The extrasynaptic current density was increased in tg-APPSwe mice relative to wild-type littermates but, only in aged tg-APPSwe mice did insulin decrease and normalize the current. CONCLUSION Insulin effects on GABA signalling in hippocampal neurons are selective while multifaceted and context-based. Not only is the response to insulin related to cell-type, hippocampal axis-location, age of animals and disease but also to the subtype of neuronal inhibition involved, synaptic or extrasynaptic GABAA receptors-activated currents.
Collapse
Affiliation(s)
- Hayma Hammoud
- Department of Medical Cell Biology Uppsala University Uppsala Sweden
| | - Olga Netsyk
- Department of Medical Cell Biology Uppsala University Uppsala Sweden
| | | | - Sergiy V. Korol
- Department of Medical Cell Biology Uppsala University Uppsala Sweden
| | - Zhe Jin
- Department of Medical Cell Biology Uppsala University Uppsala Sweden
| | - Jin‐Ping Li
- Department of Medical Biochemistry and Microbiology Uppsala University Uppsala Sweden
| | - Bryndis Birnir
- Department of Medical Cell Biology Uppsala University Uppsala Sweden
| |
Collapse
|
31
|
Erichsen JM, Calva CB, Reagan LP, Fadel JR. Intranasal insulin and orexins to treat age-related cognitive decline. Physiol Behav 2021; 234:113370. [PMID: 33621561 PMCID: PMC8053680 DOI: 10.1016/j.physbeh.2021.113370] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/19/2021] [Indexed: 02/06/2023]
Abstract
The intranasal (IN) administration of neuropeptides, such as insulin and orexins, has been suggested as a treatment strategy for age-related cognitive decline (ARCD). Because dysfunctional neuropeptide signaling is an observed characteristic of ARCD, it has been suggested that IN delivery of insulin and/or orexins may restore endogenous peptide signaling and thereby preserve cognition. IN administration is particularly alluring as it is a relatively non-invasive method that directly targets peptides to the brain. Several laboratories have examined the behavioral effects of IN insulin in young, aged, and cognitively impaired rodents and humans. These studies demonstrated improved performance on various cognitive tasks following IN insulin administration. Fewer laboratories have assessed the effects of IN orexins; however, this peptide also holds promise as an effective treatment for ARCD through the activation of the cholinergic system and/or the reduction of neuroinflammation. Here, we provide a brief overview of the advantages of IN administration and the delivery pathway, then summarize the current literature on IN insulin and orexins. Additional preclinical studies will be useful to ultimately uncover the mechanisms underlying the pro-cognitive effects of IN insulin and orexins, whereas future clinical studies will aid in the determination of the most efficacious dose and dosing paradigm. Eventually, IN insulin and/or orexin administration may be a widely used treatment strategy in the clinic for ARCD.
Collapse
Affiliation(s)
- Jennifer M Erichsen
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States.
| | - Coleman B Calva
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States
| | - Lawrence P Reagan
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States; Columbia VA Health Care System, Columbia, SC, 29208, United States
| | - Jim R Fadel
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States
| |
Collapse
|
32
|
Kuboki A, Kikuta S, Otori N, Kojima H, Matsumoto I, Reisert J, Yamasoba T. Insulin-Dependent Maturation of Newly Generated Olfactory Sensory Neurons after Injury. eNeuro 2021; 8:ENEURO.0168-21.2021. [PMID: 33906971 PMCID: PMC8143024 DOI: 10.1523/eneuro.0168-21.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/24/2022] Open
Abstract
Loss of olfactory sensory neurons (OSNs) after injury to the olfactory epithelium (OE) triggers the generation of OSNs that are incorporated into olfactory circuits to restore olfactory sensory perception. This study addresses how insulin receptor-mediated signaling affects the functional recovery of OSNs after OE injury. Insulin levels were reduced in mice by ablating the pancreatic β cells via streptozotocin (STZ) injections. These STZ-induced diabetic and control mice were then intraperitoneally injected with the olfactotoxic drug methimazole to selectively ablate OSNs. The OE of diabetic and control mice regenerated similarly until day 14 after injury. Thereafter, the OE of diabetic mice contained fewer mature and more apoptotic OSNs than control mice. Functionally, diabetic mice showed reduced electro-olfactogram (EOG) responses and their olfactory bulbs (OBs) had fewer c-Fos-active cells following odor stimulation, as well as performed worse in an odor-guided task compared with control mice. Insulin administered intranasally during days 8-13 after injury was sufficient to rescue recovery of OSNs in diabetic mice compared with control levels, while insulin administration between days 1 and 6 did not. During this critical time window on days 8-13 after injury, insulin receptors are highly expressed and intranasal application of an insulin receptor antagonist inhibits regeneration. Furthermore, an insulin-enriched environment could facilitate regeneration even in non-diabetic mice. These results indicate that insulin facilitates the regeneration of OSNs after injury and suggest a critical stage during recovery (8-13 d after injury) during which the maturation of newly generated OSNs is highly dependent on and promoted by insulin.
Collapse
Affiliation(s)
- Akihito Kuboki
- Department of Otolaryngology, Jikei University School of Medicine, Tokyo 105-8461, Japan
- Monell Chemical Senses Center, Philadelphia, PA 19104
| | - Shu Kikuta
- Department of Otolaryngology, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Nobuyoshi Otori
- Department of Otolaryngology, Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Hiromi Kojima
- Department of Otolaryngology, Jikei University School of Medicine, Tokyo 105-8461, Japan
| | | | | | - Tatsuya Yamasoba
- Department of Otolaryngology, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
33
|
Brain Insulin Resistance: Focus on Insulin Receptor-Mitochondria Interactions. Life (Basel) 2021; 11:life11030262. [PMID: 33810179 PMCID: PMC8005009 DOI: 10.3390/life11030262] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Current hypotheses implicate insulin resistance of the brain as a pathogenic factor in the development of Alzheimer’s disease and other dementias, Parkinson’s disease, type 2 diabetes, obesity, major depression, and traumatic brain injury. A variety of genetic, developmental, and metabolic abnormalities that lead to disturbances in the insulin receptor signal transduction may underlie insulin resistance. Insulin receptor substrate proteins are generally considered to be the node in the insulin signaling system that is critically involved in the development of insulin insensitivity during metabolic stress, hyperinsulinemia, and inflammation. Emerging evidence suggests that lower activation of the insulin receptor (IR) is another common, while less discussed, mechanism of insulin resistance in the brain. This review aims to discuss causes behind the diminished activation of IR in neurons, with a focus on the functional relationship between mitochondria and IR during early insulin signaling and the related roles of oxidative stress, mitochondrial hypometabolism, and glutamate excitotoxicity in the development of IR insensitivity to insulin.
Collapse
|
34
|
Wingrove JO, O'Daly O, Forbes B, Swedrowska M, Amiel SA, Zelaya FO. Intranasal insulin administration decreases cerebral blood flow in cortico-limbic regions: A neuropharmacological imaging study in normal and overweight males. Diabetes Obes Metab 2021; 23:175-185. [PMID: 33026175 DOI: 10.1111/dom.14213] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/17/2020] [Accepted: 09/27/2020] [Indexed: 12/13/2022]
Abstract
AIM To assess and compare the effects of 160 IU intranasal insulin (IN-INS) administration on regional cerebral blood flow (rCBF) in healthy male individuals with normal weight and overweight phenotypes. METHODS Thirty young male participants (mean age 25.9 years) were recruited and stratified into two cohorts based on body mass index: normal weight (18.5-24.9 kg/m2 ) and overweight (25.0-29.9 kg/m2 ). On separate mornings participants received 160 IU of IN-INS using an intranasal protocol and intranasal placebo as part of a double-blind crossover design. Thirty minutes following administration rCBF data were collected using a magnetic resonance imaging method called pseudocontinuous arterial spin labelling. Blood samples were collected to assess insulin sensitivity and changes over time in peripheral glucose, insulin and C-peptide. RESULTS Insulin sensitivity did not significantly differ between groups. Compared with placebo, IN-INS administration reduced rCBF in parts of the hippocampus, insula, putamen, parahippocampal gyrus and fusiform gyrus in the overweight group. No effect was seen in the normal weight group. Insula rCBF was greater in the overweight group versus normal weight only under placebo conditions. Peripheral glucose and insulin levels were not affected by IN-INS. C-peptide levels in the normal weight group decreased significantly over time following IN-INS administration but not placebo. CONCLUSION Insulin-induced changes within key regions of the brain involved in gustation, memory and reward were observed in overweight healthy male individuals. Following placebo administration, differences in gustatory rCBF were observed between overweight and normal weight healthy individuals.
Collapse
Affiliation(s)
- Jed O Wingrove
- Centre for Neuroimaging Sciences, King's College London, London, UK
- Centre for Obesity Research, University College London, London, UK
- Diabetes Research Group, King's College Hospital Campus, Weston Education Central, King's College London, London, UK
| | - Owen O'Daly
- Centre for Neuroimaging Sciences, King's College London, London, UK
| | - Ben Forbes
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Magda Swedrowska
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Stephanie A Amiel
- Diabetes Research Group, King's College Hospital Campus, Weston Education Central, King's College London, London, UK
- Institute of Diabetes and Obesity, King's Health Partners, London, UK
| | | |
Collapse
|
35
|
Morris JK, John CS, Green ZD, Wilkins HM, Wang X, Kamat A, Swerdlow RS, Vidoni ED, Petersen ME, O’Bryant SE, Honea RA, Burns JM. Characterization of the Meal-Stimulated Incretin Response and Relationship With Structural Brain Outcomes in Aging and Alzheimer's Disease. Front Neurosci 2020; 14:608862. [PMID: 33328877 PMCID: PMC7734152 DOI: 10.3389/fnins.2020.608862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/02/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Individuals with Alzheimer's Disease (AD) are often characterized by systemic markers of insulin resistance; however, the broader effects of AD on other relevant metabolic hormones, such as incretins that affect insulin secretion and food intake, remains less clear. METHODS Here, we leveraged a physiologically relevant meal tolerance test to assess diagnostic differences in these metabolic responses in cognitively healthy older adults (CH; n = 32) and AD (n = 23) participants. All individuals also underwent a comprehensive clinical examination, cognitive evaluation, and structural magnetic resonance imaging. RESULTS The meal-stimulated response of glucose, insulin, and peptide tyrosine tyrosine (PYY) was significantly greater in individuals with AD as compared to CH. Voxel-based morphometry revealed negative relationships between brain volume and the meal-stimulated response of insulin, C-Peptide, and glucose-dependent insulinotropic polypeptide (GIP) in primarily parietal brain regions. CONCLUSION Our findings are consistent with prior work that shows differences in metabolic regulation in AD and relationships with cognition and brain structure.
Collapse
Affiliation(s)
- Jill K. Morris
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Casey S. John
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Zachary D. Green
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Heather M. Wilkins
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Xiaowan Wang
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Ashwini Kamat
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Russell S. Swerdlow
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Eric D. Vidoni
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Melissa E. Petersen
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, United States
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, United states
| | - Sid E. O’Bryant
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, United states
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Robyn A. Honea
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Jeffrey M. Burns
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| |
Collapse
|
36
|
Xu Y, Chen F. Factors and Molecular Mechanisms Influencing the Protein Synthesis, Degradation and Membrane Trafficking of ASIC1a. Front Cell Dev Biol 2020; 8:596304. [PMID: 33195276 PMCID: PMC7644914 DOI: 10.3389/fcell.2020.596304] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/05/2020] [Indexed: 12/31/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are members of the degenerin/epithelial sodium channel superfamily. They are extracellular pH sensors that are activated by protons. Among all ASICs, ASIC1a is one of the most intensively studied isoforms because of its unique ability to be permeable to Ca2+. In addition, it is considered to contribute to various pathophysiological conditions. As a membrane proton receptor, the number of ASIC1a present on the cell surface determines its physiological and pathological functions, and this number partially depends on protein synthesis, degradation, and membrane trafficking processes. Recently, several studies have shown that various factors affect these processes. Therefore, this review elucidated the major factors and underlying molecular mechanisms affecting ASIC1a protein expression and membrane trafficking.
Collapse
Affiliation(s)
- Yayun Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Feihu Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| |
Collapse
|
37
|
Huo J, Ren S, Gao P, Wan D, Rong S, Li X, Liu S, Xu S, Sun K, Guo B, Wang P, Yu B, Wu J, Wang F, Sun T. ALG13 participates in epileptogenesis via regulation of GABA A receptors in mouse models. Cell Death Discov 2020; 6:87. [PMID: 33014431 PMCID: PMC7499177 DOI: 10.1038/s41420-020-00319-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/09/2020] [Accepted: 09/01/2020] [Indexed: 12/23/2022] Open
Abstract
ALG13 (asparagine-linked glycosylation 13) plays crucial roles in the process of N-linked glycosylation. Mutations of the ALG13 gene underlie congenital disorders of glycosylation type I (CDG-I), a rare human genetic disorder with defective glycosylation. Epilepsy is commonly observed in congenital disorders of glycosylation type I (CDG-I). In our study, we found that about 20% of adult ALG13KO knockout mice display spontaneous seizures, which were identified in a simultaneous video and intracranial EEG recording. However, the mechanisms of ALG13 by which deficiency leads to epilepsy are unknown. Whole-cell patch-clamp recordings demonstrated that ALG13KO mice show a marked decrease in gamma-aminobutyric acid A receptor (GABAAR)-mediated inhibitory synaptic transmission. Furthermore, treatment with low-dose diazepam (a positive allosteric modulator of GABAA receptors), which enhances GABAAR function, also markedly ameliorates severity of epileptic seizures in ALG13KO mice. Moreover, ALG13 may influenced the expression of GABAARα2 membrane and total protein by changing transcription level of GABAARα2. Furthermore, protein interactions between ALG13 and GABAARα2 were observed in the cortex of wild-type mice. Overall, these results reveal that ALG13 may be involved in the occurrence of epilepsy through the regulation of GABAAR function, and may provide new insight into epilepsy prevention and treatment.
Collapse
Affiliation(s)
- Junming Huo
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
| | - Shuanglai Ren
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
| | - Peng Gao
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
- Department of Neurosurgery, General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, 750001 Ningxia China
| | - Ding Wan
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
- Department of Neurosurgery, General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, 750001 Ningxia China
| | - Shikuo Rong
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
| | - Xinxiao Li
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
| | - Shenhai Liu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
| | - Siying Xu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
| | - Kuisheng Sun
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
| | - Baorui Guo
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
| | - Peng Wang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
| | - Baoli Yu
- Renji Hospital Shanghai Jiaotong University School of Medicine, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240 China
| | - Ji Wu
- Renji Hospital Shanghai Jiaotong University School of Medicine, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240 China
- Ningxia Key Laboratory of Reproduction and Genetics, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
| | - Feng Wang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
- Department of Neurosurgery, General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, 750001 Ningxia China
| | - Tao Sun
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001 Ningxia China
| |
Collapse
|
38
|
Hölscher C. Evidence for pathophysiological commonalities between metabolic and neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 155:65-89. [PMID: 32854859 DOI: 10.1016/bs.irn.2020.01.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Diabetes mellitus is a risk factor for developing neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases. This relationship seems counter-intuitive as these pathological syndromes appear to be very different. However, they share underlying mechanisms such as desensitization of insulin signaling. Insulin not only regulates blood glucose levels, but also acts as a growth factor that is important for neuronal activity and repair. Insulin signaling desensitization has been found in the brains of people with progressive neurodegenerative diseases, which is most likely driven by chronic inflammation. Based on this, insulin has been tested in patients with Alzheimer's disease, and it was found that memory formation was improved and brain pathology reduced. Glucagon-like peptide-1 (GLP-1) is an incretin hormone, and numerous drugs that mimic this peptide are on the market to treat type 2 diabetes mellitus. Preclinical studies have provided robust evidence that some of these drugs, such as liraglutide or lixisenatide can enter the brain and improve key pathological parameters, such as memory loss, impairment of motor activity, synapse loss, reduced energy utilization by neurons and chronic inflammation in the brain. First clinical trials with a GLP-1 mimetic show good effects in patients with Parkinson's disease, improving motor control and insulin signaling in the brain. This is a proof of concept that this approach is viable and that drug treatment affects the main drivers of the disease and does not just modify the symptoms. It demonstrates that this new research area is a promising and fertile space for the development of novel treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Christian Hölscher
- Neurology Department of the Second Associated Hospital of Shanxi Medical University, Taiyuan, Shanxi, PR China; Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan, PR China.
| |
Collapse
|
39
|
Spinelli M, Fusco S, Grassi C. Brain insulin resistance impairs hippocampal plasticity. VITAMINS AND HORMONES 2020; 114:281-306. [PMID: 32723548 DOI: 10.1016/bs.vh.2020.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nutrient-related signals have been demonstrated to influence brain development and cognitive functions. In particular, insulin signaling has been shown to impact on molecular cascades underlying hippocampal plasticity, learning and memory. Alteration of brain insulin signaling interferes with the maintenance of neural stem cell niche and neuronal activity in the hippocampus. Brain insulin resistance is also emerging as key factor causing the cognitive impairment observed in metabolic and neurodegenerative diseases. Here, we review the molecular mechanisms involved in the insulin modulation of both adult neurogenesis and synaptic activity in the hippocampus. We also summarize the effects of altered insulin sensitivity on hippocampal plasticity. Finally, we reassume the experimental and epidemiological evidence highlighting the critical role of brain insulin resistance at the crossroad between type 2 diabetes and Alzheimer's disease.
Collapse
Affiliation(s)
- Matteo Spinelli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Salvatore Fusco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
40
|
Gupta S, Singhal NK, Ganesh S, Sandhir R. Extending Arms of Insulin Resistance from Diabetes to Alzheimer's Disease: Identification of Potential Therapeutic Targets. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 18:172-184. [PMID: 30430949 DOI: 10.2174/1871527317666181114163515] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/08/2018] [Accepted: 11/08/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND & OBJECTIVE Type 3 diabetes (T3D) is chronic insulin resistant state of brain which shares pathology with sporadic Alzheimer's disease (sAD). Insulin signaling is a highly conserved pathway in the living systems that orchestrate cell growth, repair, maintenance, energy homeostasis and reproduction. Although insulin is primarily studied as a key molecule in diabetes mellitus, its role has recently been implicated in the development of Alzheimer's disease (AD). Severe complications in brain of diabetic patients and metabolically compromised status is evident in brain of AD patients. Underlying shared pathology of two disorders draws a trajectory from peripheral insulin resistance to insulin unresponsiveness in the central nervous system (CNS). As insulin has a pivotal role in AD, it is not an overreach to address diabetic condition in AD brain as T3D. Insulin signaling is indispensable to nervous system and it is vital for neuronal growth, repair, and maintenance of chemical milieu at synapses. Downstream mediators of insulin signaling pathway work as a regulatory hub for aggregation and clearance of unfolded proteins like Aβ and tau. CONCLUSION In this review, we discuss the regulatory roles of insulin as a pivotal molecule in brain with the understanding of defective insulin signaling as a key pathological mechanism in sAD. This article also highlights ongoing trials of targeting insulin signaling as a therapeutic manifestation to treat diabetic condition in brain.
Collapse
Affiliation(s)
- Smriti Gupta
- Department of Biochemistry, Basic Medical Science Block II, Sector 25, Panjab University, Chandigarh 160014, India
| | - Nitin Kumar Singhal
- National Agri-Food Biotechnology Institute, Sector 81, S.A.S. Nagar, Mohali, Punjab 140306, India
| | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Rajat Sandhir
- Department of Biochemistry, Basic Medical Science Block II, Sector 25, Panjab University, Chandigarh 160014, India
| |
Collapse
|
41
|
Madhusudhanan J, Suresh G, Devanathan V. Neurodegeneration in type 2 diabetes: Alzheimer's as a case study. Brain Behav 2020; 10:e01577. [PMID: 32170854 PMCID: PMC7218246 DOI: 10.1002/brb3.1577] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/04/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Rigorous research in the last few years has shown that in addition to the classical mechanism of neurodegeneration, certain unconventional mechanisms may also lead to neurodegenerative disease. One of them is a widely studied metabolic disorder: type 2 diabetes mellitus (T2DM). We now have a clear understanding of glucose-mediated neurodegeneration, mostly from studies in Alzheimer's disease (AD) models. AD is recognized to be significantly associated with hyperglycemia, even earning the term "type 3 diabetes." Here, we review first the pathophysiology of AD, both from the perspective of classical protein accumulation, as well as the newer T2DM-dependent mechanisms supported by findings from patients with T2DM. Secondly, we review the different pathways through which neurodegeneration is aggravated in hyperglycemic conditions taking AD as a case study. Finally, some of the current advances in AD management as a result of recent research developments in metabolic disorders-driven neurodegeneration are also discussed. METHODS Relevant literatures found from PubMed search were reviewed. RESULTS Apart from the known causes of AD, type 2 diabetes opens a new window to the AD pathology in several ways. It is a bidirectional interaction, of which, the molecular and signaling mechanisms are recently studied. This is our attempt to connect all of them to draw a complete mechanistic explanation for the neurodegeneration in T2DM. Refer to Figure 3. CONCLUSION The perspective of AD as a classical neurodegenerative disease is changing, and it is now being looked at from a zoomed-out perspective. The correlation between T2DM and AD is something observed and studied extensively. It is promising to know that there are certain advances in AD management following these studies.
Collapse
Affiliation(s)
- Jalaja Madhusudhanan
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Gowthaman Suresh
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Vasudharani Devanathan
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| |
Collapse
|
42
|
Induction of LTM following an Insulin Injection. eNeuro 2020; 7:ENEURO.0088-20.2020. [PMID: 32291265 PMCID: PMC7218004 DOI: 10.1523/eneuro.0088-20.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/21/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
The pond snail Lymnaea stagnalis learns conditioned taste aversion (CTA) and consolidates it into long-term memory (LTM). One-day food-deprived snails (day 1 snails) show the best CTA learning and memory, whereas more severely food-deprived snails (5 d) do not express good memory. However, previous studies showed that CTA-LTM was indeed formed in 5-d food-deprived snails (day 5 snails), but its recall was prevented by the effects of food deprivation. CTA-LTM recall in day 5 snails was expressed following 7 d of feeding and then 1 d of food deprivation (day 13 snails). In the present study, we thus hypothesized that memory recall occurs because day 13 snails are in an optimal internal state. One day of food deprivation before the memory test in day 13 snails increased the mRNA level of molluscan insulin-related peptide (MIP) in the CNS. Thus, we further hypothesized that an injection of insulin into day 5 snails following seven additional days with access to food (day 12 snails) activates CTA neurons and mimics the food deprivation state before the memory test in day 13 snails. Day 12 snails injected with insulin could recall the memory. In addition, the simultaneous injection of an anti-insulin receptor antibody and insulin into day 12 snails did not allow memory recall. Insulin injection also decreased the hemolymph glucose concentration. Together, the results suggest that an optimal internal state (i.e., a spike in insulin release and specific glucose levels) are necessary for LTM recall following CTA training in snails.
Collapse
|
43
|
Di J, Li J, O’Hara B, Alberts I, Xiong L, Li J, Li X. The role of GABAergic neural circuits in the pathogenesis of autism spectrum disorder. Int J Dev Neurosci 2020; 80:73-85. [DOI: 10.1002/jdn.10005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 12/13/2019] [Indexed: 12/21/2022] Open
Affiliation(s)
- Jing Di
- Department of Neurology David Geffen School of Medicine at UCLA Los Angeles CA USA
| | - Jian Li
- Department of Pediatrics the Second Xiangya HospitalCentral South University Changsha P.R. China
| | - Bruce O’Hara
- Department of Biology University of Kentucky Lexington KY USA
| | - Ian Alberts
- Department of Natural Sciences LaGuardia CCCUNY New York NY USA
| | - Lei Xiong
- Department of Clinical Medicine Yunnan University of Chinese Medicine Kunming P.R. China
| | - Jijun Li
- Department of Integrative Medicine on Pediatrics Shanghai Children’s Medical Center Shanghai Jiao Tong University School of Medicine Shanghai P.R. China
| | - Xiaohong Li
- Department of Neurochemistry New York State Institute for Basic Research in Developmental Disabilities New York NY USA
| |
Collapse
|
44
|
Hölscher C. Brain insulin resistance: role in neurodegenerative disease and potential for targeting. Expert Opin Investig Drugs 2020; 29:333-348. [PMID: 32175781 DOI: 10.1080/13543784.2020.1738383] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: This review evaluates the novel strategy of treating Alzheimer's and Parkinson's disease (AD and PD) withdrugs that initially have been developed to treat type 2 diabetes. As insulin signalling has been found to be de-sensitized in the brains of patients, drugs that can re-sensitize insulin signalling have been tested to evaluate if this strategy can alter disease progression.Areas covered: The review will give an overview of preclinical and clinical tests in AD and PD of drugs activating insulin receptors, glucagon-like peptide -1 (GLP-1) receptors, and glucose-dependent insulinotropic polypeptide (GIP) receptors.Expert opinion: Insulin, GLP-1 and GIP receptor agonists have shown good effects in preclinical studies. First clinical trials in MCI/AD patients have shown that insulin can improve on key pathological symptoms of AD such as memory impairment, brain activity, neuronal energy utilization, and inflammation markers. A GLP-1 receptor agonist has shown disease-modifying effects in PD patients, and first pilot studies have shown encouraging effects of a GLP-1 receptor agonist in AD patients. Novel dual GLP-1/GIP receptor agonists that cross the blood brain barrier show superior neuroprotective effects compared to single GLP-1 or GIP receptor agonists, and show great promise as novel treatments of AD and PD.
Collapse
Affiliation(s)
- Christian Hölscher
- Second Hospital, Neurology Department, Shanxi Medical University, Taiyuan, Shanxi, PR China.,Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan, PR China
| |
Collapse
|
45
|
Slankster E, Kollala S, Baria D, Dailey-Krempel B, Jain R, Odell SR, Mathew D. Mechanism underlying starvation-dependent modulation of olfactory behavior in Drosophila larva. Sci Rep 2020; 10:3119. [PMID: 32080342 PMCID: PMC7033209 DOI: 10.1038/s41598-020-60098-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/05/2020] [Indexed: 12/14/2022] Open
Abstract
Starvation enhances olfactory sensitivity that encourage animals to search for food. The molecular mechanisms that enable sensory neurons to remain flexible and adapt to a particular internal state remain poorly understood. Here, we study the roles of GABA and insulin signaling in starvation-dependent modulation of olfactory sensory neuron (OSN) function in the Drosophila larva. We show that GABAB-receptor and insulin-receptor play important roles during OSN modulation. Using an OSN-specific gene expression analysis, we explore downstream targets of insulin signaling in OSNs. Our results suggest that insulin and GABA signaling pathways interact within OSNs and modulate OSN function by impacting olfactory information processing. We further show that manipulating these signaling pathways specifically in the OSNs impact larval feeding behavior and its body weight. These results challenge the prevailing model of OSN modulation and highlight opportunities to better understand OSN modulation mechanisms and their relationship to animal physiology.
Collapse
Affiliation(s)
- Eryn Slankster
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - Sai Kollala
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - Dominique Baria
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | | | - Roshni Jain
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
- Cell and Molecular Biology Graduate Program, University of Nevada, Reno, NV, 89557, USA
| | - Seth R Odell
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
- Integrated Neuroscience Graduate Program, University of Nevada, Reno, NV, 89557, USA
| | - Dennis Mathew
- Department of Biology, University of Nevada, Reno, NV, 89557, USA.
- Cell and Molecular Biology Graduate Program, University of Nevada, Reno, NV, 89557, USA.
- Integrated Neuroscience Graduate Program, University of Nevada, Reno, NV, 89557, USA.
| |
Collapse
|
46
|
Cheong JL, de Pablo-Fernandez E, Foltynie T, Noyce AJ. The Association Between Type 2 Diabetes Mellitus and Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2020; 10:775-789. [PMID: 32333549 PMCID: PMC7458510 DOI: 10.3233/jpd-191900] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/31/2020] [Indexed: 02/07/2023]
Abstract
In recent years, an emerging body of evidence has forged links between Parkinson's disease (PD) and type 2 diabetes mellitus (T2DM). In observational studies, those with T2DM appear to be at increased risk of developing PD, as well as experiencing faster progression and a more severe phenotype of PD, with the effects being potentially mediated by several common cellular pathways. The insulin signalling pathway, for example, may be responsible for neurodegeneration via insulin dysregulation, aggregation of amyloids, neuroinflammation, mitochondrial dysfunction and altered synaptic plasticity. In light of these potential shared disease mechanisms, clinical trials are now investigating the use of established diabetes drugs targeting insulin resistance in the management of PD. This review will discuss the epidemiological links between T2DM and PD, the potential shared cellular mechanisms, and assess the relevant treatment options for disease modification of PD.
Collapse
Affiliation(s)
- Julia L.Y. Cheong
- Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Eduardo de Pablo-Fernandez
- Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, UK
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London, UK
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London, UK
| | - Alastair J. Noyce
- Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, UK
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London, UK
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| |
Collapse
|
47
|
Abstract
Addiction to substances such as alcohol, cocaine, opioids, and methamphetamine poses a continuing clinical and public challenge globally. Despite progress in understanding substance use disorders, challenges remain in their treatment. Some of these challenges include limited ability of therapeutics to reach the brain (blood-brain barrier), adverse systemic side effects of current medications, and importantly key aspects of addiction not addressed by currently available treatments (such as cognitive impairment). Inability to sustain abstinence or seek treatment due to cognitive deficits such as poor decision-making and impulsivity is known to cause poor treatment outcomes. In this review, we provide an evidenced-based rationale for intranasal drug delivery as a viable and safe treatment modality to bypass the blood-brain barrier and target insulin to the brain to improve the treatment of addiction. Intranasal insulin with improvement of brain cell energy and glucose metabolism, stress hormone reduction, and improved monoamine transmission may be an ideal approach for treating multiple domains of addiction including memory and impulsivity. This may provide additional benefits to enhance current treatment approaches.
Collapse
Affiliation(s)
- Bhavani Kashyap
- HealthPartners Neuroscience Center, 295 Phalen Blvd, St Paul, Minnesota, 55130, USA.
- HealthPartners Institute, Bloomington, Minnesota, USA.
| | - Leah R Hanson
- HealthPartners Neuroscience Center, 295 Phalen Blvd, St Paul, Minnesota, 55130, USA
- HealthPartners Institute, Bloomington, Minnesota, USA
| | - William H Frey Ii
- HealthPartners Neuroscience Center, 295 Phalen Blvd, St Paul, Minnesota, 55130, USA
- HealthPartners Institute, Bloomington, Minnesota, USA
| |
Collapse
|
48
|
Γ-Aminobutyric acid in adult brain: an update. Behav Brain Res 2019; 376:112224. [DOI: 10.1016/j.bbr.2019.112224] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 01/21/2023]
|
49
|
Nasoohi S, Parveen K, Ishrat T. Metabolic Syndrome, Brain Insulin Resistance, and Alzheimer's Disease: Thioredoxin Interacting Protein (TXNIP) and Inflammasome as Core Amplifiers. J Alzheimers Dis 2019; 66:857-885. [PMID: 30372683 DOI: 10.3233/jad-180735] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Empirical evidence indicates a strong association between insulin resistance and pathological alterations related to Alzheimer's disease (AD) in different cerebral regions. While cerebral insulin resistance is not essentially parallel with systemic metabolic derangements, type 2 diabetes mellitus (T2DM) has been established as a risk factor for AD. The circulating "toxic metabolites" emerging in metabolic syndrome may engage several biochemical pathways to promote oxidative stress and neuroinflammation leading to impair insulin function in the brain or "type 3 diabetes". Thioredoxin-interacting protein (TXNIP) as an intracellular amplifier of oxidative stress and inflammasome activation may presumably mediate central insulin resistance. Emerging data including those from our recent studies has demonstrated a sharp TXNIP upregulation in stroke, aging and AD and well underlining the significance of this hypothesis. With the main interest to illustrate TXNIP place in type 3 diabetes, the present review primarily briefs the potential mechanisms contributing to cerebral insulin resistance in a metabolically deranged environment. Then with a particular focus on plausible TXNIP functions to drive and associate with AD pathology, we present the most recent evidence supporting TXNIP as a promising therapeutic target in AD as an age-associated dementia.
Collapse
|
50
|
Lei Y, Zhang ZF, Lei RX, Wang S, Zhuang Y, Liu AC, Wu Y, Chen J, Tang JC, Pan MX, Liu R, Liao WJ, Feng YG, Wan Q, Zheng M. DJ-1 Suppresses Cytoplasmic TDP-43 Aggregation in Oxidative Stress-Induced Cell Injury. J Alzheimers Dis 2019; 66:1001-1014. [PMID: 30372676 DOI: 10.3233/jad-180460] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
DJ-1 (also called PARK7) is a multifunctional redox-sensitive protein that is protective against oxidative stress-induced cell death. TAR DNA-binding protein 43 (TDP-43) is a major protein component of pathological inclusions in amyotrophic lateral sclerosis and frontotemporal dementia. Reducing aberrant aggregation of TDP-43 is a potential approach to prevent cell death. To investigate whether DJ-1 might inhibit TDP-43 aggregation to exert a protective effect in oxidative stress-induced injury, we tested the protein level and subcellular localization of TDP-43 and DJ-1 in SH-SY5Y cells transfected with wild-type DJ-1, DJ-1 mutant (L166P) cDNA, or DJ-1 siRNA. We show that oxidative stress induced by paraquat leads to the formation of cytosolic TDP-43 aggregation in SH-SY5Y cells. DJ-1 overexpression decreases paraquat-induced cytoplasmic accumulation of TDP-43 in SH-SY5Y cells and protects against paraquat-induced cell death. Transfection of DJ-1 L166P mutant or DJ-1 siRNA leads to increased cytosolic aggregation of TDP-43 in paraquat-treated SH-SY5Y cells and promotes cell death. These data suggest that DJ-1 may protect against oxidative stress-induced cell death through the suppression of cytoplasmic TDP-43 aggregation.
Collapse
Affiliation(s)
- Yang Lei
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, Wuhan, China
| | - Zhi-Feng Zhang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, Wuhan, China.,Department of Physiology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
| | - Rui-Xue Lei
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, Wuhan, China
| | - Shu Wang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, Wuhan, China
| | - Yang Zhuang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, Wuhan, China
| | - An-Chun Liu
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, Wuhan, China
| | - Yan Wu
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, Wuhan, China
| | - Juan Chen
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, Wuhan, China
| | - Jun-Chun Tang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, Wuhan, China
| | - Meng-Xian Pan
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, Wuhan, China
| | - Rui Liu
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, Wuhan, China
| | - Wei-Jing Liao
- Center for Brain Clinic, Zhongnan Hospital, Wuhan University School of Medicine, Wuhan, China
| | - Yu-Gong Feng
- Research Institute of Neuroregeneration & Neurorehabilitation, and Department of Neurosurgery, Qingdao University, Qingdao, China
| | - Qi Wan
- Research Institute of Neuroregeneration & Neurorehabilitation, and Department of Neurosurgery, Qingdao University, Qingdao, China
| | - Mei Zheng
- Department of Neurology, Beijing University Third Hospital, Beijing, China
| |
Collapse
|