1
|
Zhou Q, Guo X, Chen T, Liu Y, Ji H, Sun Y, Yang X, Ouyang C, Liu X, Lei M. The neuroprotective role of celastrol on hippocampus in diabetic rats by inflammation restraint, insulin signaling adjustment, Aβ reduction and synaptic plasticity alternation. Biomed Pharmacother 2024; 179:117397. [PMID: 39232386 DOI: 10.1016/j.biopha.2024.117397] [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: 07/09/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Celastrol, the primary constituent of Tripterygium wilfordii, has demonstrated neuroprotective properties in rats with dementia by reducing inflammation. A high-fat diet and streptozotocin injection were utilized to establish a diabetic rat model, which was then employed to investigate the possible protective effect of celastrol against the development of diabetes-induced learning and memory deficits. Afterwards, the experimental animals received a dose of celastrol by gavage (4 mg/kg/d). An animal study showed that celastrol enhanced insulin sensitivity and glucose tolerance in diabetic rats. In the Morris water maze test, rats with diabetes performed poorly in terms of spatial learning and memory; treatment with celastrol improved these outcomes. Additionally, administration of celastrol downregulated the expression of inflammatory-related proteins (NF-κB, IKKα, TNF-α, IL-1β, and IL-6) and greatly reduced the generation of Aβ in the diabetic hippocampus tissue. Moreover, the insulin signaling pathway-related proteins PI3K, AKT, and GSK-3β were significantly upregulated in diabetic rats after celastrol was administered. Also, celastrol prevented damage to the brain structures and increased the synthesis of synaptic proteins like PSD-95 and SYT1. In conclusion, celastrol exerts a neuroprotective effect by modulating the insulin signaling system and reducing inflammatory responses, which helps to ameliorate the cognitive impairment associated with diabetes.
Collapse
Affiliation(s)
- Qiaofeng Zhou
- Hubei Key Laboratory of Diabetes And Angiopathy, Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Xiying Guo
- Hubei Key Laboratory of Diabetes And Angiopathy, Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Tu Chen
- Xianning Public Inspection and Testing Center, Xianning 437100, China
| | - Yumin Liu
- Wuhan Huake Reproductive Specialist Hospital, Wuhan 430000, China
| | - Huimin Ji
- Hubei Key Laboratory of Diabetes And Angiopathy, Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Yixuan Sun
- Hubei Key Laboratory of Diabetes And Angiopathy, Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Xiaosong Yang
- Hubei Key Laboratory of Diabetes And Angiopathy, Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Changhan Ouyang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Xiufen Liu
- Hubei Key Laboratory of Diabetes And Angiopathy, Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China.
| | - Min Lei
- Hubei Key Laboratory of Diabetes And Angiopathy, Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China.
| |
Collapse
|
2
|
Sanganahalli BG, Mihailovic JM, Vekaria HJ, Coman D, Yackzan AT, Flemister A, Aware C, Wenger K, Hubbard WB, Sullivan PG, Hyder F, Lin AL. mTOR inhibition enhances synaptic and mitochondrial function in Alzheimer's disease in an APOE genotype-dependent manner. J Cereb Blood Flow Metab 2024; 44:1745-1758. [PMID: 38879800 PMCID: PMC11494852 DOI: 10.1177/0271678x241261942] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 04/11/2024] [Accepted: 05/24/2024] [Indexed: 10/18/2024]
Abstract
Apolipoprotein ε4 (APOE4) carriers develop brain metabolic dysfunctions decades before the onset of Alzheimer's disease (AD). A goal of the study is to identify if rapamycin, an inhibitor for the mammalian target of rapamycin (mTOR) inhibitor, would enhance synaptic and mitochondrial function in asymptomatic mice with human APOE4 gene (E4FAD) before they showed metabolic deficits. A second goal is to determine whether there may be genetic-dependent responses to rapamycin when compared to mice with human APOE3 alleles (E3FAD), a neutral AD genetic risk factor. We fed asymptomatic E4FAD and E3FAD mice with control or rapamycin diets for 16 weeks from starting from 3 months of age. Neuronal mitochondrial oxidative metabolism and excitatory neurotransmission rates were measured using in vivo 1H-[13C] proton-observed carbon-edited magnetic resonance spectroscopy, and isolated mitochondrial bioenergetic measurements using Seahorse. We found that rapamycin enhanced neuronal mitochondrial function, glutamate-glutamine cycling, and TCA cycle rates in the asymptomatic E4FAD mice. In contrast, rapamycin enhances glycolysis, non-neuronal activities, and inhibitory neurotransmission of the E3FAD mice. These findings indicate that rapamycin might be able to mitigate the risk for AD by enhancing brain metabolic functions for cognitively intact APOE4 carriers, and the responses to rapamycin are varied by APOE genotypes. Consideration of precision medicine may be needed for future rapamycin therapeutics.
Collapse
Affiliation(s)
- Basavaraju G Sanganahalli
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Jelena M Mihailovic
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Hemendra J Vekaria
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
- Lexington VA Health Care System, Lexington, KY, USA
| | - Daniel Coman
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Andrew T Yackzan
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | | | - Chetan Aware
- Department of Radiology, University of Missouri, Columbia, MO, USA
| | - Kathryn Wenger
- Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - W Brad Hubbard
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
- Lexington VA Health Care System, Lexington, KY, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Patrick G Sullivan
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
- Lexington VA Health Care System, Lexington, KY, USA
| | - Fahmeed Hyder
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ai-Ling Lin
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA
| |
Collapse
|
3
|
Guo X, Lei M, Ma G, Ouyang C, Yang X, Liu C, Chen Q, Liu X. Schisandrin A Alleviates Spatial Learning and Memory Impairment in Diabetic Rats by Inhibiting Inflammatory Response and Through Modulation of the PI3K/AKT Pathway. Mol Neurobiol 2024; 61:2514-2529. [PMID: 37910285 DOI: 10.1007/s12035-023-03725-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/19/2023] [Indexed: 11/03/2023]
Abstract
Clinical and epidemiological research shows that people with diabetes mellitus frequently experience diabetic cognitive impairment. Schisandrin A (SchA), one of the lignans found in the dried fruit of Schisandra chinensis, has a variety of pharmacological effects on immune system control, apoptosis suppression, anti-oxidation and anti-inflammation. The goal of the current investigation was to clarify the probable neuro-protective effects of SchA against streptozotocin-induced diabetes deficiencies of the spatial learning and memory in rats. The outcomes show that SchA therapy effectively improved impaired glucose tolerance, fasting blood glucose level and serum insulin level in diabetic rats. Additionally, in the Morris water maze test, diabetic rats showed deficits in spatial learning and memory that were ameliorated by SchA treatment. Moreover, giving diabetic rats SchA reduced damage to the hippocampus structure and increased the production of synaptic proteins. Further research revealed that SchA therapy reduced diabetic-induced hippocampus neuron damage and the generation of Aβ, as demonstrated by the upregulated phosphorylation levels of insulin signaling pathway connected proteins and by the decreased expression levels of inflammatory-related factors. Collectively, these results suggested that SchA could improve diabetes-related impairments in spatial learning and memory, presumably by reducing inflammatory responses and regulating the insulin signaling system.
Collapse
Affiliation(s)
- Xiying Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Min Lei
- Hubei Key Laboratory of Diabetes and Angiopathy, Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Guandi Ma
- Hubei Key Laboratory of Diabetes and Angiopathy, Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Changhan Ouyang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Xiaosong Yang
- Hubei Key Laboratory of Diabetes and Angiopathy, Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Chao Liu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China.
| | - Qingjie Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China.
| | - Xiufen Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China.
| |
Collapse
|
4
|
Wang W, Wang P, Zhu L, Liu B, Wei Q, Hou Y, Li X, Hu Y, Li W, Wang Y, Jiang C, Yang G, Wang J. An optimized fluorescent biosensor for monitoring long-chain fatty acyl-CoAs metabolism in vivo. Biosens Bioelectron 2024; 247:115935. [PMID: 38128319 DOI: 10.1016/j.bios.2023.115935] [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: 10/17/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Long-chain fatty acyl-CoAs (LCACoAs) are intermediates in lipid metabolism that exert a wide range of cellular functions. However, our knowledge about the subcellular distribution and regulatory impacts of LCACoAs is limited by a lack of methods for detecting LCACoAs in living cells and tissues. Here, we report our development of LACSerHR, a genetically encoded fluorescent biosensor that enables precise measurement of subtle fluctuations in the levels of endogenous LCACoAs in vivo. LACSerHR significantly improve the fluorescent brightness and analyte affinity, in vitro and in vivo testing showcased LACSerHR's large dynamic range. We demonstrate LACSerHR's capacity for real-time evaluation of LCACoA levels in specific subcellular compartments, for example in response to disruption of ACSL enzyme function in HEK293T cells. Moreover, we show the application of LACSerHR for sensitive measurement of elevated LCACoA levels in the livers of mouse models for two common metabolic diseases (NAFLD and type 2 diabetes). Thus, our LACSerHR sensor is a powerful, broadly applicable tool for studying LCACoAs metabolism and disease.
Collapse
Affiliation(s)
- Weibo Wang
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China; National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan, 430079, PR China
| | - Pengcheng Wang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, PR China
| | - Lixin Zhu
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Bingjie Liu
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Qingpeng Wei
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Yongkang Hou
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Xi Li
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Yufei Hu
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Wenzhe Li
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Yuan Wang
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, PR China
| | - Guangfu Yang
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan, 430079, PR China.
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China.
| |
Collapse
|
5
|
Nwakama CA, Durand-de Cuttoli R, Oketokoun ZM, Brown SO, Haller JE, Méndez A, Farshbaf MJ, Cho YZ, Ahmed S, Leng S, Ables JL, Sweis BM. Diabetes alters neuroeconomically dissociable forms of mental accounting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.04.574210. [PMID: 38260368 PMCID: PMC10802482 DOI: 10.1101/2024.01.04.574210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Those with diabetes mellitus are at high-risk of developing psychiatric disorders, yet the link between hyperglycemia and alterations in motivated behavior has not been explored in detail. We characterized value-based decision-making behavior of a streptozocin-induced diabetic mouse model on a naturalistic neuroeconomic foraging paradigm called Restaurant Row. Mice made self-paced choices while on a limited time-budget accepting or rejecting reward offers as a function of cost (delays cued by tone-pitch) and subjective value (flavors), tested daily in a closed-economy system across months. We found streptozocin-treated mice disproportionately undervalued less-preferred flavors and inverted their meal-consumption patterns shifted toward a more costly strategy that overprioritized high-value rewards. We discovered these foraging behaviors were driven by impairments in multiple decision-making systems, including the ability to deliberate when engaged in conflict and cache the value of the passage of time in the form of sunk costs. Surprisingly, diabetes-induced changes in behavior depended not only on the type of choice being made but also the salience of reward-scarcity in the environment. These findings suggest complex relationships between glycemic regulation and dissociable valuation algorithms underlying unique cognitive heuristics and sensitivity to opportunity costs can disrupt fundamentally distinct computational processes and could give rise to psychiatric vulnerabilities.
Collapse
|
6
|
Barutçu Ö, Süer C, Dursun N, Tufan E, Gülpınar EA, Tan B. Insulin-induced long-term potentiation in the dentate gyrus of hippocampal formation. Psychoneuroendocrinology 2023; 157:106343. [PMID: 37562098 DOI: 10.1016/j.psyneuen.2023.106343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023]
Abstract
The discovery that brain areas involving in learning and memory express receptors for insulin hormone, led to the idea that insulin signaling may have a role in regulating cognitive function. Although previous studies have shown a role for insulin in regulation of the threshold of plasticity induction, no study has addressed whether insulin can induce a chemical plasticity per se. Young-adult male rats that are fed with standard diets with or without carbohydrate syrup (sucrose or high-fructose corn syrups) were enrolled in this study. Extracellular field potentials were recorded from the dentate gyrus in response to perforant pathway stimulation at 0.033 Hz in anesthetized rats. The slope of field excitatory postsynaptic potentials (fEPSPs) and the amplitude of population spike (PS) were measured 15 min after a 60-min infusion of insulin (500 nM), NT157 (an IRS inhibitor, 6 μM), alone or together, or physiological saline. mRNA expressions of insulin signaling proteins were measured by rt-PCR in the whole hippocampus. We did not observe any appreciable change in the fEPSP slope and the PS amplitude before and after saline infusion. However, intra-hippocampal insulin application results in the induction of LTP of fEPSP and of PS in the dentate gyrus. Insulin infusion together with NT157 inhibited fEPSP-LTP, but not PS-LTP, and rats that are fed with carbohydrate syrup did not express synaptic LTP. In rats that additional carbohydrate syrup is not given, insulin-induced LTP was accompanied with an increase in PI3K-mRNA, AKT-mRNA, and GSK-3β-mRNA which was not observed when co-administered with NT157. The GSK-3β-mRNA and IRS1-mRNA levels were found to be lower in rats that received supplemental carbohydrate and that not express insulin-induced synaptic LTP, compared to the rats expressing synaptic LTP and fed by standard diet. The results obtained provide a mechanistic link between insulin and synaptic plasticity. We concluded that insulin not only functions as a modulator of synaptic plasticity but also acts as a chemical inducer of LTP.
Collapse
Affiliation(s)
- Özlem Barutçu
- University of Erciyes, Physiology department of Medical School, Türkiye; University of Erciyes, Institute of Health Science, Türkiye; Turkey, Council of Higher Education100/2000 PhD Scholarship Student, Türkiye
| | - Cem Süer
- University of Erciyes, Physiology department of Medical School, Türkiye; University of Erciyes, Institute of Health Science, Türkiye.
| | - Nurcan Dursun
- University of Erciyes, Physiology department of Medical School, Türkiye
| | - Esra Tufan
- University of Erciyes, Physiology department of Medical School, Türkiye; University of Erciyes, Institute of Health Science, Türkiye; Turkey, Council of Higher Education100/2000 PhD Scholarship Student, Türkiye
| | - Ezgi Aslan Gülpınar
- University of Erciyes, Physiology department of Medical School, Türkiye; University of Erciyes, Institute of Health Science, Türkiye; Turkey, Council of Higher Education100/2000 PhD Scholarship Student, Türkiye
| | - Burak Tan
- University of Erciyes, Physiology department of Medical School, Türkiye
| |
Collapse
|
7
|
Masliukov PM. Changes of Signaling Pathways in Hypothalamic Neurons with Aging. Curr Issues Mol Biol 2023; 45:8289-8308. [PMID: 37886966 PMCID: PMC10605528 DOI: 10.3390/cimb45100523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
The hypothalamus is an important regulator of autonomic and endocrine functions also involved in aging regulation. The aging process in the hypothalamus is accompanied by disturbed intracellular signaling including insulin/insulin-like growth factor-1 (IGF-1)/growth hormone (GH), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT)/the mammalian target of rapamycin (mTOR), mitogen activated protein kinase (MAPK), janus kinase (JAK)/signal transducer and activator of transcription (STAT), AMP-activated protein kinase (AMPK), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ĸB), and nitric oxide (NO). In the current review, I have summarized the current understanding of the changes in the above-mentioned pathways in aging with a focus on hypothalamic alterations.
Collapse
Affiliation(s)
- Petr M Masliukov
- Department Normal Physiology, Yaroslavl State Medical University, ul. Revoliucionnaya 5, 150000 Yaroslavl, Russia
| |
Collapse
|
8
|
Alberry B, Silveira PP. Brain insulin signaling as a potential mediator of early life adversity effects on physical and mental health. Neurosci Biobehav Rev 2023; 153:105350. [PMID: 37544390 DOI: 10.1016/j.neubiorev.2023.105350] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
In numerous brain structures, insulin signaling modulates the homeostatic processes, sensitivity to reward pathways, executive function, memory, and cognition. Through human studies and animal models, mounting evidence implicates central insulin signaling in the metabolic, physiological, and psychological consequences of early life adversity. In this review, we describe the consequences of early life adversity in the brain where insulin signaling is a key factor and how insulin may moderate the effects of adversity on psychiatric and cardio-metabolic health outcomes. Further understanding of how early life adversity and insulin signaling impact specific brain regions and mental and physical health outcomes will assist in prevention, diagnosis, and potential intervention following early life adversity.
Collapse
Affiliation(s)
- Bonnie Alberry
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Patricia Pelufo Silveira
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
| |
Collapse
|
9
|
Mazucanti CH, Kennedy V, Premathilake HU, Doyle ME, Tian J, Liu QR, O'Connell J, Camandola S, Egan JM. AAV5-mediated manipulation of insulin expression in choroid plexus has long-term metabolic and behavioral consequences. Cell Rep 2023; 42:112903. [PMID: 37515772 PMCID: PMC10529429 DOI: 10.1016/j.celrep.2023.112903] [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: 02/07/2023] [Revised: 06/06/2023] [Accepted: 07/13/2023] [Indexed: 07/31/2023] Open
Abstract
The choroid plexus (CP) is a source of trophic factors for the developing and mature brain. Insulin is produced in epithelial cells of the CP (EChPs), and its secretion is stimulated by Htr2c-mediated signaling. We modulated insulin expression in EChPs with intracerebroventricular injections of AAV5. Insulin overexpression in CP decelerates food intake, whereas its knockdown has the opposite effect. Insulin overexpression also results in reduced anxious behavior. Transcriptomic changes in the hypothalamus, especially in synapse-related processes, are also seen in mice overexpressing insulin in CP. Last, activation of Gq signaling in CP leads to acute Akt phosphorylation in neurons of the arcuate nucleus, indicating a direct action of CP-derived insulin on the hypothalamus. Taken together, our findings signify that CP is a relevant source of insulin in the central nervous system and that CP-derived insulin should be taken into consideration in future work pertaining to insulin actions in the brain.
Collapse
Affiliation(s)
- Caio Henrique Mazucanti
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Vernon Kennedy
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Hasitha U Premathilake
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Maire E Doyle
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Jane Tian
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Qing-Rong Liu
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Jennifer O'Connell
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Simonetta Camandola
- Translational Gerontology Branch, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Josephine M Egan
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA.
| |
Collapse
|
10
|
Shetti AU, Ramakrishnan A, Romanova L, Li W, Vo K, Volety I, Ratnayake I, Stephen T, Minshall RD, Cologna SM, Lazarov O. Reduced endothelial caveolin-1 underlies deficits in brain insulin signalling in type 2 diabetes. Brain 2023; 146:3014-3028. [PMID: 36731883 PMCID: PMC10316766 DOI: 10.1093/brain/awad028] [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: 08/05/2022] [Revised: 01/07/2023] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
Patients with type 2 diabetes exhibit severe impairments in insulin signalling in the brain and are five times more likely to develop Alzheimer's disease. However, what leads to these impairments is not fully understood. Here, we show reduced expression of endothelial cell caveolin-1 (Cav-1) in the db/db (Leprdb) mouse model of type 2 diabetes. This reduction correlated with alterations in insulin receptor expression and signalling in brain microvessels as well as brain parenchyma. These findings were recapitulated in the brains of endothelial cell-specific Cav-1 knock-out (Tie2Cre; Cav-1fl/fl) mice. Lack of Cav-1 in endothelial cells led to reduced response to insulin as well as reduced insulin uptake. Furthermore, we observed that Cav-1 was necessary for the stabilization of insulin receptors in lipid rafts. Interactome analysis revealed that insulin receptor interacts with Cav-1 and caveolae-associated proteins, insulin-degrading enzyme and the tight junction protein Zonula Occludence-1 in brain endothelial cells. Restoration of Cav-1 in Cav-1 knock-out brain endothelial cells rescued insulin receptor expression and localization. Overall, these results suggest that Cav-1 regulates insulin signalling and uptake by brain endothelial cells by modulating IR-α and IR-β localization and function in lipid rafts. Furthermore, depletion of endothelial cell-specific Cav-1 and the resulting impairment in insulin transport leads to alteration in insulin signalling in the brain parenchyma of type 2 diabetics.
Collapse
Affiliation(s)
- Aashutosh U Shetti
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Abhirami Ramakrishnan
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Liudmila Romanova
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Wenping Li
- Department of Chemistry, College of Liberal Arts and Sciences, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Khanh Vo
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Ipsita Volety
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Ishara Ratnayake
- Electron Microscopy Core, Research Resource Center, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Terilyn Stephen
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Richard D Minshall
- Department of Pharmacology and Regenerative Medicine, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Anesthesiology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Stephanie M Cologna
- Department of Chemistry, College of Liberal Arts and Sciences, The University of Illinois Chicago, Chicago, IL 60612, USA
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois Chicago, Chicago, IL 60612, USA
| |
Collapse
|
11
|
Chen W, Huang Q, Lazdon EK, Gomes A, Wong M, Stephens E, Royal TG, Frenkel D, Cai W, Kahn CR. Loss of insulin signaling in astrocytes exacerbates Alzheimer-like phenotypes in a 5xFAD mouse model. Proc Natl Acad Sci U S A 2023; 120:e2220684120. [PMID: 37186836 PMCID: PMC10214134 DOI: 10.1073/pnas.2220684120] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Brain insulin signaling controls peripheral energy metabolism and plays a key role in the regulation of mood and cognition. Epidemiological studies have indicated a strong connection between type 2 diabetes (T2D) and neurodegenerative disorders, especially Alzheimer's disease (AD), linked via dysregulation of insulin signaling, i.e., insulin resistance. While most studies have focused on neurons, here, we aim to understand the role of insulin signaling in astrocytes, a glial cell type highly implicated in AD pathology and AD progression. To this end, we created a mouse model by crossing 5xFAD transgenic mice, a well-recognized AD mouse model that expresses five familial AD mutations, with mice carrying a selective, inducible insulin receptor (IR) knockout in astrocytes (iGIRKO). We show that by age 6 mo, iGIRKO/5xFAD mice exhibited greater alterations in nesting, Y-maze performance, and fear response than those of mice with the 5xFAD transgenes alone. This was associated with increased Tau (T231) phosphorylation, increased Aβ plaque size, and increased association of astrocytes with plaques in the cerebral cortex as assessed using tissue CLARITY of the brain in the iGIRKO/5xFAD mice. Mechanistically, in vitro knockout of IR in primary astrocytes resulted in loss of insulin signaling, reduced ATP production and glycolic capacity, and impaired Aβ uptake both in the basal and insulin-stimulated states. Thus, insulin signaling in astrocytes plays an important role in the control of Aβ uptake, thereby contributing to AD pathology, and highlighting the potential importance of targeting insulin signaling in astrocytes as a site for therapeutics for patients with T2D and AD.
Collapse
Affiliation(s)
- Wenqiang Chen
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA02215
| | - Qian Huang
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY11568
| | - Ekaterina Katie Lazdon
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv69978, Israel
| | - Antonio Gomes
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA02215
| | - Marisa Wong
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY11568
| | - Emily Stephens
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA02215
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX79430
| | - Tabitha Grace Royal
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv69978, Israel
| | - Dan Frenkel
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA02215
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv69978, Israel
| | - Weikang Cai
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA02215
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY11568
| | - C. Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA02215
| |
Collapse
|
12
|
Loss of brain energy metabolism control as a driver for memory impairment upon insulin resistance. Biochem Soc Trans 2023; 51:287-301. [PMID: 36606696 DOI: 10.1042/bst20220789] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023]
Abstract
The pathophysiological mechanisms intersecting metabolic and neurodegenerative disorders include insulin resistance, which has a strong involvement of environmental factors. Besides central regulation of whole-body homeostasis, insulin in the central nervous system controls molecular signalling that is critical for cognitive performance, namely signalling through pathways that modulate synaptic transmission and plasticity, and metabolism in neurons and astrocytes. This review provides an overview on how insulin signalling in the brain might regulate brain energy metabolism, and further identified molecular mechanisms by which brain insulin resistance might impair synaptic fuelling, and lead to cognitive deterioration.
Collapse
|
13
|
Abedi A, Foroutan T, Mohaghegh Shalmani L, Dargahi L. Sex-specific effects of high-fat diet on rat brain glucose metabolism and early-onset dementia symptoms. Mech Ageing Dev 2023; 211:111795. [PMID: 36828273 DOI: 10.1016/j.mad.2023.111795] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/08/2023] [Accepted: 02/21/2023] [Indexed: 02/24/2023]
Abstract
Peripheral metabolic disturbances are associated with a variety of clinical health consequences and may contribute to the development of neurocognitive disorders. This study investigates whether long-term high-fat diet (HFD) consumption changes the brain glucose metabolism and impairs memory performance in a sex-dependent manner. Male and female rats, after weaning, were fed HFD or normal chow diet (NCD) for 16 weeks. Behavioral tests for spatial memory and an 18 F-FDG-PET scan were performed. Also, the expression of brain insulin resistance markers and Alzheimer's pathology-related genes was assessed by qPCR. The Morris water maze and Y-maze results showed, respectively, that memory retrieval and spatial working memory were impaired only in HFD male rats compared to NCD controls. In addition, measuring whole brain 18 F-FDG uptake indicated a significant reduction in glucose metabolism in male but not female HFD rats. Analysis of 15 genes related to glucose metabolism and Alzheimer's pathology, in the hippocampus, showed that expression of GLUT3, IRS2, and IDE is significantly reduced in HFD male rats. Our results suggest that sex affects the HFD-induced dysregulation of brain glucose metabolism and cognitive performance.
Collapse
Affiliation(s)
- Azam Abedi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Tahereh Foroutan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Leila Mohaghegh Shalmani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Nagao H, Cai W, Brandão BB, Wewer Albrechtsen NJ, Steger M, Gattu AK, Pan H, Dreyfuss JM, Wunderlich FT, Mann M, Kahn CR. Leucine-973 is a crucial residue differentiating insulin and IGF-1 receptor signaling. J Clin Invest 2023; 133:161472. [PMID: 36548088 PMCID: PMC9927934 DOI: 10.1172/jci161472] [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: 04/29/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Insulin and IGF-1 receptors (IR and IGF1R) are highly homologous and share similar signaling systems, but each has a unique physiological role, with IR primarily regulating metabolic homeostasis and IGF1R regulating mitogenic control and growth. Here, we show that replacement of a single amino acid at position 973, just distal to the NPEY motif in the intracellular juxtamembrane region, from leucine, which is highly conserved in IRs, to phenylalanine, the highly conserved homologous residue in IGF1Rs, resulted in decreased IRS-1/PI3K/Akt/mTORC1 signaling and increased Shc/Gab1/MAPK cell cycle signaling. As a result, cells expressing L973F-IR exhibited decreased insulin-induced glucose uptake, increased cell growth, and impaired receptor internalization. Mice with knockin of the L973F-IR showed similar alterations in signaling in vivo, and this led to decreased insulin sensitivity, a modest increase in growth, and decreased weight gain when mice were challenged with a high-fat diet. Thus, leucine-973 in the juxtamembrane region of the IR acts as a crucial residue differentiating IR signaling from IGF1R signaling.
Collapse
Affiliation(s)
- Hirofumi Nagao
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Weikang Cai
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, USA
| | - Bruna B Brandão
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicolai J Wewer Albrechtsen
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences,and.,Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Martin Steger
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Arijeet K Gattu
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA.,Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Hui Pan
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan M Dreyfuss
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - F Thomas Wunderlich
- Max Planck Institute for Metabolism Research, Center for Endocrinology, Diabetes and Preventive Medicine, University Hospital of Cologne, Center for Molecular Medicine Cologne, and.,Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, Cologne, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences,and
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Wu M, Liao W, Zhang R, Gao Y, Chen T, Hua L, Cai F. PTP1B Inhibitor Claramine Rescues Diabetes-Induced Spatial Learning and Memory Impairment in Mice. Mol Neurobiol 2023; 60:524-544. [PMID: 36319905 DOI: 10.1007/s12035-022-03079-9] [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: 05/25/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022]
Abstract
Accumulating clinical and epidemiological studies indicate that learning and memory impairment is more prevalent among people with diabetes mellitus (DM). PTP1B is a member of protein tyrosine phosphatase family and participates in a variety of pathophysiological effects including inflammatory, insulin signaling pathway, and learning and memory. This study was aimed to investigate the effects of CA, a specific inhibitor of PTP1B, on spatial learning and memory impairment in diabetic mice caused by high-fat diet and injection of streptozotocin. We found that the protein expressions of PTP1B increased in hippocampal CA1, CA3, and PFC regions of diabetic mice. Network pharmacology results showed that PTP1B might be one of the key targets between diabetes and cognitive dysfunction, and CA might alleviate DM-induced cognitive dysfunction. Animal experiments showed that CA ameliorated DM-induced spatial learning and memory impairment, and improved glucose and lipid metabolic disorders. Moreover, administration of CA alleviated hippocampal structure damage and enhanced the expressions of synaptic proteins, including PSD-95, SYN-1, and SYP in diabetic mice. Furthermore, CA treatment not only significantly down-regulated the expressions of PTP1B and NLRP3 inflammatory related proteins (NLRP3, ASC, Caspase-1, COX-2, IL-1β, and TNF-α), but also significantly up-regulated the expressions of insulin signaling pathway-related proteins (p-IRS1, p-PI3K, p-AKT, and p-GSK-3β) in diabetic mice. Taken together, these results suggested that PTP1B might be a targeted strategy to rescue learning and memory deficits in DM, possibly through inhibition of NLRP3 inflammasome and regulation of insulin signaling pathway.
Collapse
Affiliation(s)
- Mengyu Wu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Wenli Liao
- Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Ruyi Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Yuting Gao
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Tao Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Liangliang Hua
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Fei Cai
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China.
- Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China.
| |
Collapse
|
16
|
Modifiable risk factors of dementia linked to excitation-inhibition imbalance. Ageing Res Rev 2023; 83:101804. [PMID: 36410620 DOI: 10.1016/j.arr.2022.101804] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 11/04/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022]
Abstract
Recent evidence identifies 12 potentially modifiable risk factors for dementia to which 40% of dementia cases are attributed. While the recognition of these risk factors has paved the way for the development of new prevention measures, the link between these risk factors and the underlying pathophysiology of dementia is yet not well understood. A growing number of recent clinical and preclinical studies support a role of Excitation-Inhibition (E-I) imbalance in the pathophysiology of dementia. In this review, we aim to propose a conceptual model on the links between the modifiable risk factors and the E-I imbalance in dementia. This model, which aims to address the current gap in the literature, is based on 12 mediating common mechanisms: the hypothalamic-pituitary-adrenal (HPA) axis dysfunction, neuroinflammation, oxidative stress, mitochondrial dysfunction, cerebral hypo-perfusion, blood-brain barrier (BBB) dysfunction, beta-amyloid deposition, elevated homocysteine level, impaired neurogenesis, tau tangles, GABAergic dysfunction, and glutamatergic dysfunction. We believe this model serves as a framework for future studies in this field and facilitates future research on dementia prevention, discovery of new biomarkers, and developing new interventions.
Collapse
|
17
|
Insulin and IGF-1 elicit robust transcriptional regulation to modulate autophagy in astrocytes. Mol Metab 2022; 66:101647. [PMID: 36503893 PMCID: PMC9731889 DOI: 10.1016/j.molmet.2022.101647] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/08/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE Insulin is a principal metabolic hormone. It regulates a plethora of metabolic pathways in peripheral tissues. The highly homologous insulin-like growth factor 1 (IGF-1), on the other hand, is important for development and growth. Recent studies have shown that insulin and IGF-1 signaling plays fundamental roles in the brain. Loss of insulin or IGF-1 receptors in astrocytes leads to altered glucose handling, mitochondrial metabolism, neurovascular coupling, and behavioral abnormalities in mice. Here, we aim to investigate molecular mechanisms by which insulin and IGF-1 signaling regulates astrocyte functions. METHODS IR-flox and IRKO primary astrocytes were treated with 100 nM insulin or IGF-1 for 6 h, and their transcriptomes were analyzed. Astrocytes with either IR deletion, IGF1R deletion or both were used to examine receptor-dependent transcriptional regulations using qPCR. Additional immunoblotting and confocal imaging studies were performed to functionally validate pathways involved in protein homeostasis. RESULTS Using next-generation RNA sequencing, we show that insulin significantly regulates the expression of over 1,200 genes involved in multiple functional processes in primary astrocytes. Insulin-like growth factor 1 (IGF-1) triggers a similar robust transcriptional regulation in astrocytes. Thus, over 50% of the differentially expressed genes are regulated by both ligands. As expected, these commonly regulated genes are highly enriched in pathways involved in lipid and cholesterol biosynthesis. Additionally, insulin and IGF-1 induce the expression of genes involved in ribosomal biogenesis, while suppressing the expression of genes involved in autophagy, indicating a common role of insulin and IGF-1 on protein homeostasis in astrocytes. Insulin-dependent suppression of autophagy genes, including p62, Ulk1/2, and several Atg genes, is blunted only when both IR and IGF1R are deleted. CONCLUSIONS In summary, insulin and IGF-1 potently suppress autophagy in astrocytes through transcriptional regulation. Both IR and IGF1R can elicit ligand-dependent transcriptional suppression of autophagy. These results demonstrate an important role of astrocytic insulin/IGF-1 signaling on proteostasis. Impairment of this regulation in insulin resistance and diabetes may contribute to neurological complications related to diabetes.
Collapse
|
18
|
Pemberton S, Galindo DC, Schwartz MW, Banks WA, Rhea EM. Endocytosis of insulin at the blood-brain barrier. FRONTIERS IN DRUG DELIVERY 2022; 2:1062366. [PMID: 37936681 PMCID: PMC10629879 DOI: 10.3389/fddev.2022.1062366] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
For insulin to act within the brain, it is primarily transported from the blood across the blood-brain barrier (BBB). However, the endocytic machinery necessary for delivering insulin to the brain remains unknown. Additionally, there are processes within the brain endothelial cell that are designed to respond to insulin binding and elicit intracellular signaling. Using pharmacological inhibitors of different types of endocytosis (clathrin-vs. caveolin-mediated), we investigated molecular mediators of both insulin BBB binding in isolated mouse brain microvessels and BBB insulin transport in mice studied by brain perfusion. We found clathrin-mediated mechanisms responsible for insulin surface binding in isolated brain microvessels while caveolin-mediated endocytosis may mediate BBB insulin transport specifically in the hypothalamus. These results further define the molecular machinery necessary for transporting insulin into the CNS and highlight the distinction between insulin internalization for transendothelial transport vs. intracellular signaling.
Collapse
Affiliation(s)
- Sarah Pemberton
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States
| | - Demi C Galindo
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States
| | - Michael W Schwartz
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - William A Banks
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States
- Division of Gerontology and Geriatric Medicine, Department of Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States
- Division of Gerontology and Geriatric Medicine, Department of Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
19
|
Zakharova IO, Bayunova LV, Derkach KV, Ilyasov IO, Morina IY, Shpakov AO, Avrova NF. Effects of Intranasally Administered Insulin and Gangliosides on Hypothalamic Signaling and Expression of Hepatic Gluconeogenesis Genes in Rats with Type 2 Diabetes Mellitus. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022060072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
20
|
Chen W, Cai W, Hoover B, Kahn CR. Insulin action in the brain: cell types, circuits, and diseases. Trends Neurosci 2022; 45:384-400. [PMID: 35361499 PMCID: PMC9035105 DOI: 10.1016/j.tins.2022.03.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/10/2022] [Accepted: 03/03/2022] [Indexed: 10/18/2022]
Abstract
Since its discovery over 100 years ago, insulin has been recognized as a key hormone in control of glucose homeostasis. Deficiencies of insulin signaling are central to diabetes and many other disorders. The brain is among the targets of insulin action, and insulin resistance is a major contributor to many diseases, including brain disorders. Here, we summarize key roles of insulin action in the brain and how this involves different brain cell types. Disordered brain insulin signaling can also contribute to neuropsychiatric diseases, affecting brain circuits involved in mood and cognition. Understanding of insulin signaling in different brain cell types/circuits and how these are altered in disease may lead to the development of new therapeutic approaches to these challenging disorders.
Collapse
|
21
|
De Felice FG, Gonçalves RA, Ferreira ST. Impaired insulin signalling and allostatic load in Alzheimer disease. Nat Rev Neurosci 2022; 23:215-230. [DOI: 10.1038/s41583-022-00558-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2022] [Indexed: 12/14/2022]
|
22
|
Batra A, Latsko M, Portella AK, Silveira PP. Early adversity and insulin: neuroendocrine programming beyond glucocorticoids. Trends Endocrinol Metab 2021; 32:1031-1043. [PMID: 34635400 DOI: 10.1016/j.tem.2021.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/07/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023]
Abstract
Exposure to direct or contextual adversities during early life programs the functioning of the brain and other biological systems, contributing to the development of physical as well as mental health issues in the long term. While the role of glucocorticoids in mediating the outcomes of early adversity has been explored for many years, less attention has been given to insulin. Beyond its metabolic effects in the periphery, central insulin action affects synaptic plasticity, brain neurotransmission, and executive functions. Knowledge about the interactions between the peripheral metabolism and brain function from a developmental perspective can contribute to prevention and diagnosis programs, as well as early interventions for vulnerable populations.
Collapse
Affiliation(s)
- Aashita Batra
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
| | - Maeson Latsko
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Healthy Brains for Healthy Lives, McGill University, Montreal, QC, Canada
| | - Andre Krumel Portella
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Patricia P Silveira
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
| |
Collapse
|