1
|
Baranykova S, Gupta RK, Kajdasz A, Wasilewska I, Macias M, Szybinska A, Węgierski T, Nahia KA, Mondal SS, Winata CL, Kuźnicki J, Majewski L. Loss of Stim2 in zebrafish induces glaucoma-like phenotype. Sci Rep 2024; 14:24442. [PMID: 39424970 PMCID: PMC11489432 DOI: 10.1038/s41598-024-74909-0] [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: 05/12/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024] Open
Abstract
Calcium is involved in vision processes in the retina and implicated in various pathologies, including glaucoma. Rod cells rely on store-operated calcium entry (SOCE) to safeguard against the prolonged lowering of intracellular calcium ion concentrations. Zebrafish that lacked the endoplasmic reticulum Ca2+ sensor Stim2 (stim2 knockout [KO]) exhibited impaired vision and lower light perception-related gene expression. We sought to understand mechanisms that are responsible for vision impairment in stim2 KO zebrafish. The single-cell RNA (scRNA) sequencing of neuronal cells from brains of 5 days postfertilization larvae distinguished 27 cell clusters, 10 of which exhibited distinct gene expression patterns, including amacrine and γ-aminobutyric acid (GABA)ergic retinal interneurons and GABAergic optic tectum cells. Five clusters exhibited significant changes in cell proportions between stim2 KO and controls, including GABAergic diencephalon and optic tectum cells. Transmission electron microscopy of stim2 KO zebrafish revealed decreases in width of the inner plexiform layer, ganglion cells, and their dendrites numbers (a hallmark of glaucoma). GABAergic neuron densities in the inner nuclear layer, including amacrine cells, as well as photoreceptors significantly decreased in stim2 KO zebrafish. Our study suggests a novel role for Stim2 in the regulation of neuronal insulin expression and GABAergic-dependent vision causing glaucoma-like retinal pathology.
Collapse
Affiliation(s)
- Sofiia Baranykova
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland
| | - Rishikesh Kumar Gupta
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, 201313, India
| | - Arkadiusz Kajdasz
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Zygmunta Noskowskiego 12/14, 61-704, Poznan, Poland
- Xenstats sp. z o.o., Otwarta 1, 60-008, Poznan, Poland
| | - Iga Wasilewska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Adolfa Pawińskiego 5, 02-106, Warsaw, Poland
| | - Matylda Macias
- Microscopy and Cytometry Facility, International Institute of Molecular and Cell Biology, Ks. Trojdena 4, 02-109, WarsawWarsaw, Poland
| | - Aleksandra Szybinska
- Microscopy and Cytometry Facility, International Institute of Molecular and Cell Biology, Ks. Trojdena 4, 02-109, WarsawWarsaw, Poland
| | - Tomasz Węgierski
- Microscopy and Cytometry Facility, International Institute of Molecular and Cell Biology, Ks. Trojdena 4, 02-109, WarsawWarsaw, Poland
| | - Karim Abu Nahia
- Laboratory of Zebrafish Developmental Genomics, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland
| | - Shamba S Mondal
- Laboratory of Zebrafish Developmental Genomics, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland
| | - Cecilia L Winata
- Laboratory of Zebrafish Developmental Genomics, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland
| | - Jacek Kuźnicki
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland
| | - Lukasz Majewski
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland.
| |
Collapse
|
2
|
El Hajji S, Shiga Y, Belforte N, Solorio YC, Tastet O, D’Onofrio P, Dotigny F, Prat A, Arbour N, Fortune B, Di Polo A. Insulin restores retinal ganglion cell functional connectivity and promotes visual recovery in glaucoma. SCIENCE ADVANCES 2024; 10:eadl5722. [PMID: 39110798 PMCID: PMC11305393 DOI: 10.1126/sciadv.adl5722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 06/28/2024] [Indexed: 08/10/2024]
Abstract
Dendrite pathology and synaptic loss result in neural circuit dysfunction, a common feature of neurodegenerative diseases. There is a lack of strategies that target dendritic and synaptic regeneration to promote neurorecovery. We show that daily human recombinant insulin eye drops stimulate retinal ganglion cell (RGC) dendrite and synapse regeneration during ocular hypertension, a risk factor to develop glaucoma. We demonstrate that the ribosomal protein p70S6 kinase (S6K) is essential for insulin-dependent dendritic regrowth. Furthermore, S6K phosphorylation of the stress-activated protein kinase-interacting protein 1 (SIN1), a link between the mammalian target of rapamycin complexes 1 and 2 (mTORC1/2), is required for insulin-induced dendritic regeneration. Using two-photon microscopy live retinal imaging, we show that insulin rescues single-RGC light-evoked calcium (Ca2+) dynamics. We further demonstrate that insulin enhances neuronal survival and retina-brain connectivity leading to improved optomotor reflex-elicited behaviors. Our data support that insulin is a compelling pro-regenerative strategy with potential clinical implications for the treatment and management of glaucoma.
Collapse
Affiliation(s)
- Sana El Hajji
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Yukihiro Shiga
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Nicolas Belforte
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Yves Carpentier Solorio
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Olivier Tastet
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Philippe D’Onofrio
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Florence Dotigny
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Alexandre Prat
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Nathalie Arbour
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Health, Portland, OR, USA
| | - Adriana Di Polo
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| |
Collapse
|
3
|
Qeva E, Sollazzo C, Bilotta F. Insulin signaling in the central nervous system, a possible pathophysiological mechanism of anesthesia-induced delayed neurocognitive recovery/postoperative neurocognitive disorder: a narrative review. Expert Rev Neurother 2022; 22:839-847. [PMID: 36332201 DOI: 10.1080/14737175.2022.2144234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
INTRODUCTION Impairment in neurocognitive functions ranges between delayed neurocognitive recovery (DNR) and postoperative neurocognitive disorders (pNCD). Incidence varies from 11% after noncardiac surgery to 60% after cardiac surgery. AREAS COVERED Insulin receptors (IRs) signaling pathway in the central nervous system (CNS) could be a possible pathophysiological mechanism of anesthesia-induced DNR/pNCD and perioperative intranasal insulin administration could be a preventive approach. This hypothesis is supported by the following evidence: effects of IRs-CNS signaling pathway on neuromodulation; higher incidence of DNR/pNCD in patients with insulin resistance; neurotoxicity of IRs signaling pathways after anesthetic exposure; improvement of neurocognitive impairment after insulin exposure. This narrative review was conducted after a literature search of PubMed, EMBASE and SCOPUS online medical data performed in May 2022. EXPERT OPINION Perioperative intranasal insulin is shown to be protective and future studies should address: the role of insulin as a neuromodulator; its integration into neuroprotection approaches; patient populations that might benefit from this approach; a well-defined protocol of intranasal insulin administration in a perioperative background and other disciplines; and possible collateral effects.
Collapse
Affiliation(s)
- Ega Qeva
- Department of Anesthesia and Intensive Care Medicine, "Sapienza" University of Rome, 'Policlinico Umberto I' Hospital, 00161 Rome, Italy.,Department of Anesthesia, Intensive Care and Emergency, University of Turin, 'Città Della Salute e Della Scienza' Hospital, 10126 Turin, Italy
| | - Camilla Sollazzo
- Department of Anesthesia and Intensive Care Medicine, "Sapienza" University of Rome, 'Policlinico Umberto I' Hospital, 00161 Rome, Italy
| | - Federico Bilotta
- Department of Anesthesia and Intensive Care Medicine, "Sapienza" University of Rome, 'Policlinico Umberto I' Hospital, 00161 Rome, Italy
| |
Collapse
|
4
|
Effects of Chronic Arginase Inhibition with Norvaline on Tau Pathology and Brain Glucose Metabolism in Alzheimer's Disease Mice. Neurochem Res 2022; 47:1255-1268. [DOI: 10.1007/s11064-021-03519-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 12/11/2021] [Accepted: 12/27/2021] [Indexed: 12/23/2022]
|
5
|
Al Hussein Al Awamlh S, Wareham LK, Risner ML, Calkins DJ. Insulin Signaling as a Therapeutic Target in Glaucomatous Neurodegeneration. Int J Mol Sci 2021; 22:4672. [PMID: 33925119 PMCID: PMC8124776 DOI: 10.3390/ijms22094672] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 01/28/2023] Open
Abstract
Glaucoma is a multifactorial disease that is conventionally managed with treatments to lower intraocular pressure (IOP). Despite these efforts, many patients continue to lose their vision. The degeneration of retinal ganglion cells (RGCs) and their axons in the optic tract that characterizes glaucoma is similar to neurodegeneration in other age-related disorders of the central nervous system (CNS). Identifying the different molecular signaling pathways that contribute to early neuronal dysfunction can be utilized for neuroprotective strategies that prevent degeneration. The discovery of insulin and its receptor in the CNS and retina led to exploration of the role of insulin signaling in the CNS. Historically, insulin was considered a peripherally secreted hormone that regulated glucose homeostasis, with no obvious roles in the CNS. However, a growing number of pre-clinical and clinical studies have demonstrated the potential of modulating insulin signaling in the treatment of neurodegenerative diseases. This review will highlight the role that insulin signaling plays in RGC neurodegeneration. We will focus on how this pathway can be therapeutically targeted to promote RGC axon survival and preserve vision.
Collapse
Affiliation(s)
- Sara Al Hussein Al Awamlh
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.H.A.A.); (L.K.W.); (M.L.R.)
| | - Lauren K. Wareham
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.H.A.A.); (L.K.W.); (M.L.R.)
| | - Michael L. Risner
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.H.A.A.); (L.K.W.); (M.L.R.)
| | - David J. Calkins
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.H.A.A.); (L.K.W.); (M.L.R.)
- Department of Ophthalmology & Visual Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
6
|
Shaughness M, Acs D, Brabazon F, Hockenbury N, Byrnes KR. Role of Insulin in Neurotrauma and Neurodegeneration: A Review. Front Neurosci 2020; 14:547175. [PMID: 33100956 PMCID: PMC7546823 DOI: 10.3389/fnins.2020.547175] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022] Open
Abstract
Insulin is a hormone typically associated with pancreatic release and blood sugar regulation. The brain was long thought to be “insulin-independent,” but research has shown that insulin receptors (IR) are expressed on neurons, microglia and astrocytes, among other cells. The effects of insulin on cells within the central nervous system are varied, and can include both metabolic and non-metabolic functions. Emerging data suggests that insulin can improve neuronal survival or recovery after trauma or during neurodegenerative diseases. Further, data suggests a strong anti-inflammatory component of insulin, which may also play a role in both neurotrauma and neurodegeneration. As a result, administration of exogenous insulin, either via systemic or intranasal routes, is an increasing area of focus in research in neurotrauma and neurodegenerative disorders. This review will explore the literature to date on the role of insulin in neurotrauma and neurodegeneration, with a focus on traumatic brain injury (TBI), spinal cord injury (SCI), Alzheimer’s disease (AD) and Parkinson’s disease (PD).
Collapse
Affiliation(s)
- Michael Shaughness
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Deanna Acs
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Fiona Brabazon
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Nicole Hockenbury
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Kimberly R Byrnes
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
7
|
Wu S, Yang S, Bloe CB, Zhuang R, Huang J, Zhang W. Identification of Key Genes and Pathways in Mouse Spinal Cord Involved in ddC-Induced Neuropathic Pain by Transcriptome Sequencing. J Mol Neurosci 2020; 71:651-661. [PMID: 32812184 DOI: 10.1007/s12031-020-01686-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/11/2020] [Indexed: 01/12/2023]
Abstract
Highly active antiretroviral therapy (HAART) works effectively in inhibiting HIV replication in patients. However, the use of nucleoside reverse transcriptase inhibitors (NRTIs) often causes side effects of neuropathic pain, and its mechanism remains to be elucidated. Therefore, we aim to explore the mechanism of NRTIs-induced neuropathic pain at the transcriptome level. C57BL/6 J mice were given intraperitoneal injection of zalcitabine (ddC) or saline (control) for 2 weeks, during which the mechanical pain threshold of the mice was detected by von Frey test. Then the L3~L5 spinal segments of the mice were isolated and subsequently used for RNA sequencing (RNA-seq) on the last day of treatment. The mechanical pain threshold of mice given ddC decreased significantly. Compared with the control group, ddC caused significant changes in the expression of 135 genes, of which 66 upregulated and 69 downregulated. Enrichment analysis showed that the functions of these genes are mainly enriched in regulation of transcription, multicellular organism development, and cell differentiation, and the pathway is mainly enriched in the cGMP-PKG signaling pathway and AMPK signaling pathway. Furthermore, key genes such as Gabrd, Kcnd3, Npcd, Insr, Lypd6, Scd2, and Mef2d were also identified. These may serve as drug targets for the prevention or treatment of NRTI-induced neuropathic pain.
Collapse
Affiliation(s)
- Shengjun Wu
- Clinical Laboratory of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Su Yang
- Clinical Laboratory of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Chris Bloe Bloe
- College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Renjie Zhuang
- The Affiliated Hospital of Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Jian Huang
- College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Wenping Zhang
- College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| |
Collapse
|
8
|
Byun MS, Kim HJ, Yi D, Choi HJ, Baek H, Lee JH, Choe YM, Lee SH, Ko K, Sohn BK, Lee JY, Lee Y, Kim YK, Lee YS, Lee DY. Region-specific association between basal blood insulin and cerebral glucose metabolism in older adults. NEUROIMAGE-CLINICAL 2019; 22:101765. [PMID: 30904824 PMCID: PMC6434096 DOI: 10.1016/j.nicl.2019.101765] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 12/31/2018] [Accepted: 03/10/2019] [Indexed: 01/30/2023]
Abstract
Background Although previous studies have suggested that insulin plays a role in brain function, it still remains unclear whether or not insulin has a region-specific association with neuronal and synaptic activity in the living human brain. We investigated the regional pattern of association between basal blood insulin and resting-state cerebral glucose metabolism (CMglu), a proxy for neuronal and synaptic activity, in older adults. Method A total of 234 nondiabetic, cognitively normal (CN) older adults underwent comprehensive clinical assessment, resting-state 18F-fluodeoxyglucose (FDG)-positron emission tomography (PET) and blood sampling to determine overnight fasting blood insulin and glucose levels, as well as apolipoprotein E (APOE) genotyping. Results An exploratory voxel-wise analysis of FDG-PET without a priori hypothesis demonstrated a positive association between basal blood insulin levels and resting-state CMglu in specific cerebral cortices and hippocampus, rather than in non-specific overall cerebral regions, even after controlling for the effects of APOE e4 carrier status, vascular risk factor score, body mass index, fasting blood glucose, and demographic variables. Particularly, a positive association of basal blood insulin with CMglu in the right posterior hippocampus and adjacent parahippocampal region as well as in the right inferior parietal region remained significant after multiple comparison correction. Conversely, no region showed negative association between basal blood insulin and CMglu. Conclusions Our finding suggests that basal fasting blood insulin may have association with neuronal and synaptic activity in specific cerebral regions, particularly in the hippocampal/parahippocampal and inferior parietal regions. We investigated regional pattern of association between basal blood insulin and resting-state cerebral glucose metabolism. Significant clusters with positive associations were found mainly in the hippocampal and inferior parietal regions. Our finding suggests a region-specific association of basal blood insulin with resting-state cerebral glucose metabolism. Further studies to elucidate underlying mechanism and implication of this region-specific association will be necessary.
Collapse
Affiliation(s)
- Min Soo Byun
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Hyun Jung Kim
- Department of Psychiatry, Changsan Convalescent Hospital, Changwon, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Hyo Jung Choi
- Department of Psychiatry, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Hyewon Baek
- Department of Neuropsychiatry, Kyunggi Provincial Hospital for the Elderly, Yongin, Republic of Korea
| | - Jun Ho Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Young Min Choe
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Republic of Korea
| | - Seung Hoon Lee
- Department of Neuropsychiatry, Bucheon Geriatric Medical Center, Bucheon, Republic of Korea
| | - Kang Ko
- Department of Neuropsychiatry, National Center for Mental Health, Seoul, Republic of Korea
| | - Bo Kyung Sohn
- Department of Psychiatry, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Republic of Korea
| | - Jun-Young Lee
- Department of Psychiatry, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea; Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Younghwa Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dong Young Lee
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea; Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea; Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | | |
Collapse
|
9
|
Dakic TB, Jevdjovic TV, Peric MI, Bjelobaba IM, Markelic MB, Milutinovic BS, Lakic IV, Jasnic NI, Djordjevic JD, Vujovic PZ. Short-term fasting promotes insulin expression in rat hypothalamus. Eur J Neurosci 2017; 46:1730-1737. [PMID: 28544147 DOI: 10.1111/ejn.13607] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 04/26/2017] [Accepted: 05/14/2017] [Indexed: 11/30/2022]
Abstract
In the hypothalamus, insulin takes on many roles involved in energy homoeostasis. Therefore, the aim of this study was to examine hypothalamic insulin expression during the initial phase of the metabolic response to fasting. Hypothalamic insulin content was assessed by both radioimmunoassay and Western blot. The relative expression of insulin mRNA was examined by qPCR. Immunofluorescence and immunohistochemistry were used to determine the distribution of insulin immunopositivity in the hypothalamus. After 6-h fasting, both glucose and insulin levels were decreased in serum but not in the cerebrospinal fluid. Our study showed for the first time that, while the concentration of circulating glucose and insulin decreased, both insulin mRNA expression and insulin content in the hypothalamic parenchyma were increased after short-term fasting. Increased insulin immunopositivity was detected specifically in the neurons of the hypothalamic periventricular nucleus and in the ependymal cells of fasting animals. These novel findings point to the complexity of mechanisms regulating insulin expression in the CNS in general and in the hypothalamus in particular.
Collapse
Affiliation(s)
- Tamara B Dakic
- Department for Comparative Physiology and Ecophysiology, Faculty of Biology, Institute for Physiology and Biochemistry, University of Belgrade, Studentski trg 16, 11000, Belgrade, Serbia
| | - Tanja V Jevdjovic
- Department for Comparative Physiology and Ecophysiology, Faculty of Biology, Institute for Physiology and Biochemistry, University of Belgrade, Studentski trg 16, 11000, Belgrade, Serbia
| | - Mina I Peric
- Centre for Laser Microscopy, Faculty of Biology, Institute for Physiology and Biochemistry, University of Belgrade, Belgrade, Serbia
| | - Ivana M Bjelobaba
- Institute for Biological Research 'Sinisa Stankovic', University of Belgrade, Belgrade, Serbia
| | - Milica B Markelic
- Chair for Cell and Tissue Biology, Faculty of Biology, Institute for Zoology, University of Belgrade, Belgrade, Serbia
| | - Bojana S Milutinovic
- Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Iva V Lakic
- Department for Comparative Physiology and Ecophysiology, Faculty of Biology, Institute for Physiology and Biochemistry, University of Belgrade, Studentski trg 16, 11000, Belgrade, Serbia
| | - Nebojsa I Jasnic
- Department for Comparative Physiology and Ecophysiology, Faculty of Biology, Institute for Physiology and Biochemistry, University of Belgrade, Studentski trg 16, 11000, Belgrade, Serbia
| | - Jelena D Djordjevic
- Department for Comparative Physiology and Ecophysiology, Faculty of Biology, Institute for Physiology and Biochemistry, University of Belgrade, Studentski trg 16, 11000, Belgrade, Serbia
| | - Predrag Z Vujovic
- Department for Comparative Physiology and Ecophysiology, Faculty of Biology, Institute for Physiology and Biochemistry, University of Belgrade, Studentski trg 16, 11000, Belgrade, Serbia
| |
Collapse
|
10
|
Liu J, Saponjian Y, Mahoney MM, Staley KJ, Berdichevsky Y. Epileptogenesis in organotypic hippocampal cultures has limited dependence on culture medium composition. PLoS One 2017; 12:e0172677. [PMID: 28225808 PMCID: PMC5321418 DOI: 10.1371/journal.pone.0172677] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 02/08/2017] [Indexed: 01/10/2023] Open
Abstract
Rodent organotypic hippocampal cultures spontaneously develop epileptiform activity after approximately 2 weeks in vitro and are increasingly used as a model of chronic post-traumatic epilepsy. However, organotypic cultures are maintained in an artificial environment (culture medium), which contains electrolytes, glucose, amino acids and other components that are not present at the same concentrations in cerebrospinal fluid (CSF). Therefore, it is possible that epileptogenesis in organotypic cultures is driven by these components. We examined the influence of medium composition on epileptogenesis. Epileptogenesis was evaluated by measurements of lactate and lactate dehydrogenase (LDH) levels (biomarkers of ictal activity and cell death, respectively) in spent culture media, immunohistochemistry and automated 3-D cell counts, and extracellular recordings from CA3 regions. Changes in culture medium components moderately influenced lactate and LDH levels as well as electrographic seizure burden and cell death. However, epileptogenesis occurred in any culture medium that was capable of supporting neural survival. We conclude that medium composition is unlikely to be the cause of epileptogenesis in the organotypic hippocampal culture model of chronic post-traumatic epilepsy.
Collapse
Affiliation(s)
- Jing Liu
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, Pennsylvania, United States of America
| | - Yero Saponjian
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mark M. Mahoney
- Bioengineering Program, Lehigh University, Bethlehem, Pennsylvania, United States of America
| | - Kevin J. Staley
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yevgeny Berdichevsky
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, Pennsylvania, United States of America
- Bioengineering Program, Lehigh University, Bethlehem, Pennsylvania, United States of America
| |
Collapse
|
11
|
Benni JM, Patil PA. Non-diabetic clinical applications of insulin. J Basic Clin Physiol Pharmacol 2016; 27:445-456. [PMID: 27235672 DOI: 10.1515/jbcpp-2015-0101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 04/12/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND Introducing a new drug to the market is a time-consuming process, is complex, and involves consumption of a lot of resources. Therefore, discovering new uses for the old drugs (i.e. drug repurposing) benefits the patients by providing them time-tested drugs. With developments in insulin therapy still happening, it is worth keeping up to date on trends in the use of this powerful glucose-lowering agent. The aim of this article is to explore the potential non-diabetic clinical applications of insulin. METHODS Literature survey was carried out through the various scientific journals publishing experimental and clinical research papers regarding the diverse applications of insulin other than in diabetes mellitus. These applications include both therapeutic as well as diagnostic uses of insulin. The relevant information collected from these publications was paraphrased in the present paper. RESULTS On studying the literature, the non-diabetic uses of insulin include the following: wound healing, parenteral nutrition, antiaging, body building, cardioprotection in acute coronary syndromes, insulin tolerance test to test the hypothalamo-pituitary-adrenal axis functioning, cell culture, cancer treatment, organ preservation, and management of septic shock, calcium channel, β-blocker overdose and other critical illnesses in intensive care units. CONCLUSIONS This review attempts to survey some interesting new applications of insulin other than in diabetes mellitus.
Collapse
|
12
|
Bilotta F, Lauretta MP, Tewari A, Haque M, Hara N, Uchino H, Rosa G. Insulin and the Brain: A Sweet Relationship With Intensive Care. J Intensive Care Med 2015; 32:48-58. [PMID: 26168800 DOI: 10.1177/0885066615594341] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/28/2015] [Accepted: 05/15/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Insulin receptors (IRs) in the brain have unique molecular features and a characteristic pattern of distribution. Their possible functions extend beyond glucose utilization. In this systematic review, we explore the interactions between insulin and the brain and its implications for anesthesiologists, critical care physicians, and other medical disciplines. METHODS A literature search of published preclinical and clinical studies between 1978 and 2014 was conducted, yielding 5996 articles. After applying inclusion and exclusion criteria, 92 studies were selected for this systematic review. RESULTS The IRs have unique molecular features, pattern of distribution, and mechanism of action. It has effects on neuronal function, metabolism, and neurotransmission. The IRs are involved in neuronal apoptosis and neurodegenerative processes. CONCLUSION In this systematic review, we present a close relationship between insulin and the brain, with discernible effects on memory, learning abilities, and motor functions. The potential therapeutic effects extend from acute brain insults such as traumatic brain injury, brain ischemia, and hemorrhage, to chronic neurodegenerative diseases such as Alzheimer and Parkinson disease. An understanding of the wider effects of insulin conveyed in this review will prompt anaesthesiologists and critical care physicians to consider its therapeutic potential and guide future studies.
Collapse
Affiliation(s)
- F Bilotta
- Department of Anesthesiology, Critical Care and Pain Medicine, "Sapienza" University of Rome, Rome, Italy
| | - M P Lauretta
- Anesthesia and Intensive Care Department, "La Sapienza" University of Rome, Rome, Italy .,Critical Care Department, University College London Hospital, London, United Kingdom
| | - A Tewari
- Department of Pediatric Neuroanesthesia and IONM, Cincinnati Children Hospital & Medical Center, Cincinnati, OH, USA
| | - M Haque
- Anesthesia and Critical Care Department, University College London Hospital, London, United Kingdom
| | - N Hara
- Department of Anesthesiology, Tokyo Medical University, Tokyo, Japan
| | - H Uchino
- Department of Anesthesiology, Tokyo Medical University, Tokyo, Japan
| | - G Rosa
- Department of Anesthesiology, Critical Care and Pain Medicine, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
13
|
Akintola AA, van Heemst D. Insulin, aging, and the brain: mechanisms and implications. Front Endocrinol (Lausanne) 2015; 6:13. [PMID: 25705204 PMCID: PMC4319489 DOI: 10.3389/fendo.2015.00013] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/21/2015] [Indexed: 12/12/2022] Open
Abstract
There is now an impressive body of literature implicating insulin and insulin signaling in successful aging and longevity. New information from in vivo and in vitro studies concerning insulin and insulin receptors has extended our understanding of the physiological role of insulin in the brain. However, the relevance of these to aging and longevity remains to be elucidated. Here, we review advances in our understanding of the physiological role of insulin in the brain, how insulin gets into the brain, and its relevance to aging and longevity. Furthermore, we examine possible future therapeutic applications and implications of insulin in the context of available models of delayed and accelerated aging.
Collapse
Affiliation(s)
- Abimbola A. Akintola
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: Abimbola A. Akintola, Department of Gerontology and Geriatrics, Leiden University Medical Center, C7-124, Albinusdreef 2, Leiden 2333 ZA, Netherlands e-mail:
| | - Diana van Heemst
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
14
|
Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis. Med Hypotheses 2014; 82:535-46. [DOI: 10.1016/j.mehy.2014.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 01/27/2014] [Accepted: 02/03/2014] [Indexed: 12/12/2022]
|
15
|
Insulin deprivation decreases caspase-dependent apoptotic signaling in cultured rat sertoli cells. ISRN UROLOGY 2013; 2013:970370. [PMID: 24228182 PMCID: PMC3817687 DOI: 10.1155/2013/970370] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/12/2013] [Indexed: 11/30/2022]
Abstract
Insulin is essential for the regulation of glucose homeostasis. Insulin dysfunction occurs in several pathologies, such as diabetes mellitus, which is associated with fertility problems. Somatic Sertoli cells (SCs) not only metabolize glucose to lactate, which is the central energy source used by developing germ cells, but also determine the germ cell population size. If a deregulation in SCs apoptosis occurs, it will affect germ cells, compromising spermatogenesis. As SCs apoptotic signaling is a hormonally regulated process, we hypothesized that the lack of insulin could lead to alterations in apoptotic signaling. Therefore, we examined the effect of insulin deprivation on several markers of apoptotic signaling in cultured rat SCs. We determined mRNA and protein expression of apoptotic markers as well as caspase-3 activity. SCs cultured in insulin deprivation demonstrated a significant decrease on mRNA levels of p53, Bax, caspase-9, and caspase-3 followed by a significant increase of Bax and decrease of caspase-9 protein levels relatively to the control. Caspase-3 activity was also decreased in SCs cultured in insulin deprivation conditions. Our results show that insulin deprivation decreases caspase-dependent apoptotic signaling in cultured rat SCs evidencing a possible mechanism by which lack of insulin can affect spermatogenesis and fertility.
Collapse
|
16
|
Haghir H, Rezaee AAR, Sankian M, Kheradmand H, Hami J. The effects of induced type-I diabetes on developmental regulation of insulin & insulin like growth factor-1 (IGF-1) receptors in the cerebellum of rat neonates. Metab Brain Dis 2013; 28:397-410. [PMID: 23397157 DOI: 10.1007/s11011-013-9386-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 02/01/2013] [Indexed: 12/24/2022]
Abstract
Diabetes during pregnancy impairs brain development in offspring, leading to behavioral problems, motor dysfunction and learning deficits. Insulin and insulin-like growth factor-1 (IGF-1) are important regulators of developmental and cognitive functions in the central nervous system. Aim of the present study was to examine the effects of maternal diabetes on insulin receptor (InsR) and IGF-1 receptor (IGF-1R) expression in the developing rat cerebellum. Wistar female rats were maintained diabetic from a week before pregnancy through parturition and male offspring was killed at P0, P7, and P14, an active neurogenesis period in brain development equivalent to the third trimester in human. The expression of InsR and IGF-1R in cerebelli was evaluated using real-time PCR and western blot analysis. We found a significant upregulation of both IGF-1R and InsR transcripts in cerebellum of pups born to diabetic mothers at P0, compared to controls. However, at the same time point, the results of western blot analysis revealed only a slight change in their protein levels. In contrast to InsR, which does not show any difference, there was a markedly reduction in cerebellar expression of IGF-1R mRNA and protein level in the diabetic group of newborns at P7. Moreover, 2 weeks after birth, mRNA expression and protein levels of both InsR and IGF-1R in cerebellum of the diabetic group was significantly downregulated. Compared to controls, we did not find any difference in cerebellar InsR or IGF-1R mRNA and protein levels in the insulin treated group. The present study revealed that diabetes during pregnancy strongly influences the regulation of both InsR and IGF-1R in the developing cerebellum. Furthermore, optimal maternal glycaemia control by insulin administration normalized these effects.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- Animals, Newborn
- Blood Glucose/metabolism
- Blotting, Western
- Cerebellum/metabolism
- DNA, Complementary/biosynthesis
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/metabolism
- Female
- Gene Expression/drug effects
- Male
- Pregnancy
- RNA/biosynthesis
- RNA/isolation & purification
- Rats
- Rats, Wistar
- Real-Time Polymerase Chain Reaction
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
Collapse
Affiliation(s)
- Hossein Haghir
- Department of Anatomy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | | | | |
Collapse
|
17
|
Ghasemi R, Haeri A, Dargahi L, Mohamed Z, Ahmadiani A. Insulin in the brain: sources, localization and functions. Mol Neurobiol 2012; 47:145-71. [PMID: 22956272 DOI: 10.1007/s12035-012-8339-9] [Citation(s) in RCA: 215] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 08/20/2012] [Indexed: 02/07/2023]
Abstract
Historically, insulin is best known for its role in peripheral glucose homeostasis, and insulin signaling in the brain has received less attention. Insulin-independent brain glucose uptake has been the main reason for considering the brain as an insulin-insensitive organ. However, recent findings showing a high concentration of insulin in brain extracts, and expression of insulin receptors (IRs) in central nervous system tissues have gathered considerable attention over the sources, localization, and functions of insulin in the brain. This review summarizes the current status of knowledge of the peripheral and central sources of insulin in the brain, site-specific expression of IRs, and also neurophysiological functions of insulin including the regulation of food intake, weight control, reproduction, and cognition and memory formation. This review also considers the neuromodulatory and neurotrophic effects of insulin, resulting in proliferation, differentiation, and neurite outgrowth, introducing insulin as an attractive tool for neuroprotection against apoptosis, oxidative stress, beta amyloid toxicity, and brain ischemia.
Collapse
Affiliation(s)
- Rasoul Ghasemi
- Department of Physiology, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | | | |
Collapse
|
18
|
Accardi G, Caruso C, Colonna-Romano G, Camarda C, Monastero R, Candore G. Can Alzheimer disease be a form of type 3 diabetes? Rejuvenation Res 2012; 15:217-21. [PMID: 22533436 DOI: 10.1089/rej.2011.1289] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Alzheimer disease (AD) and metabolic syndrome are two highly prevalent pathological conditions of Western society due to incorrect diet, lifestyle, and vascular risk factors. Recent data have suggested metabolic syndrome as an independent risk factor for AD and pre-AD syndrome. Furthermore, biological plausibility for this relationship has been framed within the "metabolic cognitive syndrome" concept. Due to the increasing aging of populations, prevalence of AD in Western industrialized countries will rise in the near future. Thus, new knowledge in the area of molecular biology and epigenetics will probably help to make an early molecular diagnosis of dementia. An association between metabolic syndrome and specific single-nucleotide polymorphisms (SNPs) in the gene INPPL1, encoding for SHIP2, a SH2 domain-containing inositol 5-phosphatase involved in insulin signaling, has been described. According to recent data suggesting that Type 2 diabetes represents an independent risk factor for AD and pre-AD, preliminary results of a case-control study performed to test the putative association between three SNPs in the SHIP2 gene and AD show a trend toward association of these SNPs with AD.
Collapse
Affiliation(s)
- Giulia Accardi
- Department of Biopathology and Medical and Forensic Biotechnologies, University of Palermo, Palermo, Italy
| | | | | | | | | | | |
Collapse
|
19
|
Duarte AI, Moreira PI, Oliveira CR. Insulin in central nervous system: more than just a peripheral hormone. J Aging Res 2012; 2012:384017. [PMID: 22500228 PMCID: PMC3303591 DOI: 10.1155/2012/384017] [Citation(s) in RCA: 213] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 10/12/2011] [Accepted: 11/23/2011] [Indexed: 12/14/2022] Open
Abstract
Insulin signaling in central nervous system (CNS) has emerged as a novel field of research since decreased brain insulin levels and/or signaling were associated to impaired learning, memory, and age-related neurodegenerative diseases. Thus, besides its well-known role in longevity, insulin may constitute a promising therapy against diabetes- and age-related neurodegenerative disorders. More interestingly, insulin has been also faced as the potential missing link between diabetes and aging in CNS, with Alzheimer's disease (AD) considered as the "brain-type diabetes." In fact, brain insulin has been shown to regulate both peripheral and central glucose metabolism, neurotransmission, learning, and memory and to be neuroprotective. And a future challenge will be to unravel the complex interactions between aging and diabetes, which, we believe, will allow the development of efficient preventive and therapeutic strategies to overcome age-related diseases and to prolong human "healthy" longevity. Herewith, we aim to integrate the metabolic, neuromodulatory, and neuroprotective roles of insulin in two age-related pathologies: diabetes and AD, both in terms of intracellular signaling and potential therapeutic approach.
Collapse
Affiliation(s)
- Ana I. Duarte
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Paula I. Moreira
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal
| | - Catarina R. Oliveira
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Institute of Biochemistry, Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal
| |
Collapse
|
20
|
Loganathan G, Dawra RK, Pugazhenthi S, Guo Z, Soltani SM, Wiseman A, Sanders MA, papas KK, Kumaravel V, Saluja AK, Sutherland DE, Hering BJ, Balamurugan AN. Insulin degradation by acinar cell proteases creates a dysfunctional environment for human islets before/after transplantation: benefits of α-1 antitrypsin treatment. Transplantation 2011; 92:1222-30. [PMID: 22089666 PMCID: PMC3587768 DOI: 10.1097/tp.0b013e318237585c] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Pancreatic acinar cells are commonly cotransplanted along with islets during auto- and allotransplantations. The aims of this study were to identify how acinar cell proteases cause human islet cell loss before and after transplantation of impure islet preparations and to prevent islet loss and improve function with supplementation of α-1 antitrypsin (A1AT). METHODS Acinar cell protease activity, insulin levels, and percent islet loss were measured after culture of pure and impure clinical islet preparations. The effect of proteases on ultrastructure of islets and β-cell insulin granules were examined by transmission electron microscopy. The number of insulin granules and insulin-labeled immunogold particles were counted. The in vivo effect of proteases on islet function was studied by transplanting acinar cells adjacent to islet grafts in diabetic mice. The effects of A1AT culture supplementation on protease activity, insulin levels, and islet function were assessed in pure and impure islets. RESULTS Islet loss after culture was significantly higher in impure relative to pure preparations (30% vs. 14%, P<0.04). Lower islet purity was associated with increased protease activity and decreased insulin levels in culture supernatants. Reduced β-cell insulin granules and insulin degradation by proteases were confirmed by transmission electron microscopy. Transplantations in mice showed delayed islet graft function when acinar cells were transplanted adjacent to the islets under the kidney capsule. Supplementation of A1AT to impure islet cultures maintained islet cell mass, restored insulin levels, and preserved islet functional integrity. CONCLUSION Culture of impure human islet fractions in the presence of A1AT prevents insulin degradation and improves islet recovery.
Collapse
Affiliation(s)
| | - Rajinder K. Dawra
- Department of Surgery, Basic and Translational Research, University of Minnesota, Minneapolis, MN.
| | | | - Zhiguang Guo
- Sanford Project, Sanford Health/University of South Dakota, Sioux Falls, SD.
| | - Sajjad M. Soltani
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN.
| | | | | | - Klearchos K. papas
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN.
| | - V. Kumaravel
- Alpha Hospital and Research Center, Institute of Diabetes and Endocrinology, Madurai-9, Tamilnadu, India.
| | - Ashok K. Saluja
- Department of Surgery, Basic and Translational Research, University of Minnesota, Minneapolis, MN.
| | - David E.R. Sutherland
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN.
| | - Bernhard J. Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN.
| | - A. N. Balamurugan
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN.
- Corresponding author: Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA. Phone: 651-253-0656, Fax: 612-626-5855
| |
Collapse
|
21
|
Burns JM, Honea RA, Vidoni ED, Hutfles LJ, Brooks WM, Swerdlow RH. Insulin is differentially related to cognitive decline and atrophy in Alzheimer's disease and aging. Biochim Biophys Acta Mol Basis Dis 2011; 1822:333-9. [PMID: 21745566 DOI: 10.1016/j.bbadis.2011.06.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Revised: 06/21/2011] [Accepted: 06/23/2011] [Indexed: 01/21/2023]
Abstract
We assessed the relationship of insulin resistance with cognitive decline and brain atrophy over two years in early Alzheimer's disease (AD, n=48) and nondemented controls (n=61). Intravenous glucose tolerance tests were conducted at baseline to determine insulin area-under-the-curve (AUC). A standard battery of cognitive tasks and MRI were conducted at baseline and 2-year follow-up. In nondemented controls, higher baseline insulin AUC was associated with 2-year decline in global cognitive performance (beta=-0.36, p=0.005). In early AD, however, higher insulin AUC was associated with less decline in global cognitive performance (beta=0.26, p=0.06), slower global brain atrophy (beta=0.40, p=0.01) and less regional atrophy in the bilateral hippocampi and cingulate cortices. While insulin resistance is associated with cognitive decline in nondemented aging, higher peripheral insulin may have AD-specific benefits or insulin signaling may be affected by systemic physiologic changes associated with AD. This article is part of a Special Issue entitled: Imaging Brain Aging and Neurodegenerative disease.
Collapse
Affiliation(s)
- Jeffrey M Burns
- Department of Neurology, 3599 Rainbow Blvd, University of Kansas School of Medicine Kansas City, KS 66160, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Keuper M, Blüher M, Schön MR, Möller P, Dzyakanchuk A, Amrein K, Debatin KM, Wabitsch M, Fischer-Posovszky P. An inflammatory micro-environment promotes human adipocyte apoptosis. Mol Cell Endocrinol 2011; 339:105-13. [PMID: 21501656 DOI: 10.1016/j.mce.2011.04.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 03/30/2011] [Accepted: 04/01/2011] [Indexed: 02/08/2023]
Abstract
Obesity-associated macrophage infiltration into adipose tissue is responsible for both local and systemic inflammation. Recent findings suggest fat cell apoptosis as an initiator of macrophage recruitment. Here, we investigated the effects of an inflammatory micro-environment on fat cells using human THP-1 macrophages and SGBS adipocytes. Macrophage-secreted factors induced insulin resistance, inhibited insulin-stimulated Akt phosphorylation, and induced apoptosis of adipocytes. The apoptosis-inducing effect was even more pronounced in direct co-cultures of adipocytes and macrophages. Our data suggest a link between insulin resistance and apoptosis sensitivity. Accordingly, pharmacological and genetic inhibition of insulin signaling at the level of Akt2 sensitized adipocytes to apoptosis induction by macrophage-secreted factors. In conclusion, we describe here a novel interaction of macrophages and fat cells, i.e. induction of apoptosis. Our data suggest a feed-forward cycle in which macrophages further drive the inflammatory process by inducing insulin resistance and concomitant apoptosis of adipocytes.
Collapse
Affiliation(s)
- Michaela Keuper
- Division of Pediatric Endocrinology, Diabetes and Obesity Unit, Department of Pediatrics and Adolescent Medicine, Ulm University, Eythstr. 24, 89075 Ulm, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Schuh AF, Rieder CM, Rizzi L, Chaves M, Roriz-Cruz M. Mechanisms of brain aging regulation by insulin: implications for neurodegeneration in late-onset Alzheimer's disease. ISRN NEUROLOGY 2011; 2011:306905. [PMID: 22389813 PMCID: PMC3263551 DOI: 10.5402/2011/306905] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Accepted: 04/09/2011] [Indexed: 01/21/2023]
Abstract
Insulin and IGF seem to be important players in modulating brain aging. Neurons share more similarities with islet cells than any other human cell type. Insulin and insulin receptors are diffusely found in the brain, especially so in the hippocampus. Caloric restriction decreases insulin resistance, and it is the only proven mechanism to expand lifespan. Conversely, insulin resistance increases with age, obesity, and sedentarism, all of which have been shown to be risk factors for late-onset Alzheimer's disease (AD). Hyperphagia and obesity potentiate the production of oxidative reactive species (ROS), and chronic hyperglycemia accelerates the formation of advanced glucose end products (AGEs) in (pre)diabetes—both mechanisms favoring a neurodegenerative milieu. Prolonged high cerebral insulin concentrations cause microvascular endothelium proliferation, chronic hypoperfusion, and energy deficit, triggering β-amyloid oligomerization and tau hyperphosphorylation. Insulin-degrading enzyme (IDE) seems to be the main mechanism in clearing β-amyloid from the brain. Hyperinsulinemic states may deviate IDE utilization towards insulin processing, decreasing β-amyloid degradation.
Collapse
Affiliation(s)
- Artur F Schuh
- Division of Geriatric Neurology, Department of Neurology, Clinicas Hospital (HCPA), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street 2.350, 90035-903 Porto Alegre, RS, Brazil
| | | | | | | | | |
Collapse
|
24
|
Gaddameedi RR, Burgula S, Sairam M, Singh SS. Role of insulin in Cr(VI)-mediated genotoxicity in Neurospora crassa. Lett Appl Microbiol 2011; 53:14-21. [PMID: 21488912 DOI: 10.1111/j.1472-765x.2011.03058.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
AIMS Chromium (III) is an insulinomimetic agent whose biological and/or environmental availability is frequently in the form of Cr(VI), which is known to be toxic. Wall-less mutant of Neurospora crassa (FGSC stock no. 4761) is known to possess insulin receptor in its cell membrane and hence is a good model for Cr toxicity studies. This study explores the toxicity of Cr(VI) and the possible consequences on simultaneous exposure to insulin in N. crassa. METHODS AND RESULTS Comet assay of N. crassa cells treated with 100 μmol l⁻¹ Cr(VI) showed up to 50% reduction in comet tail lengths when incubated simultaneously with 0.4 U insulin. Fluorescence measurement in Cr(VI)-treated cells using DCFH-DA showed six- to eightfold increase in free radical generation, which was reduced to fourfold by 0.4 U insulin. Annexin-V/PI Flow cytometry analysis indicated necrotic cell death up to 28.7 ± 3.6% and 68.6 ± 2.5% on Cr(VI) exposure at concentrations 100 and 500 μmol l⁻¹ which was reduced by 68.3 ± 3.2% and 48.9 ± 3.6%, respectively, upon addition of insulin. CONCLUSION Insulin-mediated protection from DNA damage by Cr(VI) is because of scavenging of free radicals liberated during exposure to Cr(VI). SIGNIFICANCE AND IMPACT OF THE STUDY Overall, Cr(VI) toxicity depends upon available insulin, indicating that Cr(VI) toxicity may be a serious issue in insulin-deficient individuals with diabetes.
Collapse
Affiliation(s)
- R R Gaddameedi
- Department of Biochemistry, Osmania University, Hyderabad, India
| | | | | | | |
Collapse
|
25
|
Burns JM, Johnson DK, Watts A, Swerdlow RH, Brooks WM. Reduced lean mass in early Alzheimer disease and its association with brain atrophy. ACTA ACUST UNITED AC 2010; 67:428-33. [PMID: 20385908 DOI: 10.1001/archneurol.2010.38] [Citation(s) in RCA: 288] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To examine body composition in individuals with early AD and without dementia and its relation to cognition and brain volume. DESIGN Cross-sectional case-control study. PARTICIPANTS Individuals without dementia (Clinical Dementia Rating, 0; n = 70) and with early-stage AD (Clinical Dementia Rating, 0.5 or 1; n = 70) in the Alzheimer and Memory Program at the University of Kansas School of Medicine. MAIN OUTCOME MEASURES Participants were evaluated with brain magnetic resonance imaging (MRI), neuropsychological testing, and dual-energy x-ray absorptiometry to determine whole-body fat and lean masses. Body mass index was calculated as weight in kilograms divided by height in meters squared. RESULTS Lean mass was reduced in persons with early AD compared with controls without dementia (F = 7.73; P = .006) after controlling for sex. Whole-brain volume (beta = .20; P < .001), white matter volume (beta = .19; P < .001), and global cognitive performance (beta = .12; P = .007) were associated with lean mass (dependent variable) when controlling for age and sex. The total body fat and percentage of body fat values were not different across groups or related to cognition and brain volume. CONCLUSION Loss of lean mass is accelerated in AD and is associated with brain atrophy and cognitive performance, perhaps as a direct or indirect consequence of AD pathophysiology or through shared mechanisms common to both AD and sarcopenia.
Collapse
Affiliation(s)
- Jeffrey M Burns
- Department of Neurology, University of Kansas School of Medicine, 3599 Rainbow Blvd, Kansas City, KS 66160, USA.
| | | | | | | | | |
Collapse
|
26
|
Levin BE. Developmental gene x environment interactions affecting systems regulating energy homeostasis and obesity. Front Neuroendocrinol 2010; 31:270-83. [PMID: 20206200 PMCID: PMC2903638 DOI: 10.1016/j.yfrne.2010.02.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 02/25/2010] [Accepted: 02/25/2010] [Indexed: 11/25/2022]
Abstract
Most human obesity is inherited as a polygenic trait which is largely refractory to medical therapy because obese individuals avidly defend their elevated body weight set-point. This set-point is mediated by an integrated neural network that controls energy homeostasis. Epidemiological studies suggest that perinatal and pre-pubertal environmental factors can promote offspring obesity. Rodent studies demonstrate the important interactions between genetic predisposition and environmental factors in promoting obesity. This review covers issues of development and function of neural systems involved in the regulation of energy homeostasis and the roles of leptin and insulin in these processes, the ways in which interventions at various phases from gestation, lactation and pre-pubertal stages of development can favorably and unfavorably alter the development of obesity n offspring. These studies suggest that early identification of obesity-prone humans and of the factors that can prevent them from becoming obese could provide an effective strategy for preventing the world-wide epidemic of obesity.
Collapse
Affiliation(s)
- Barry E Levin
- Neurology Service, VA Medical Center, E. Orange, NJ 07018-1095, USA.
| |
Collapse
|
27
|
Levin BE. Interaction of perinatal and pre-pubertal factors with genetic predisposition in the development of neural pathways involved in the regulation of energy homeostasis. Brain Res 2010; 1350:10-7. [PMID: 20059985 DOI: 10.1016/j.brainres.2009.12.085] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 12/28/2009] [Accepted: 12/29/2009] [Indexed: 12/26/2022]
Abstract
A majority of human obesity is inherited as a polygenic trait. Once obesity develops, over 90% of individuals repeatedly regain lost weight after dieting. Only surgical interventions offer long lasting weight loss. Thus, clinical data suggest that some individuals have a predisposition to develop and maintain an elevated body weight set-point once they are provided with sufficient calories to gain weight. This set-point is mediated by an integrated neural network that controls energy homeostasis. Unfortunately, currently available tools for identifying obesity-prone individuals and examining the functioning of these neural systems have insufficient resolution to identify specific neural factors that cause humans to develop and maintain the obese state. However, rodent models of polygenically inherited obesity allow us to investigate the factors that both predispose them to become obese and that prevent or enhance the development of such obesity. Maternal obesity during gestation and lactation in obesity-prone rodents enhances offspring obesity and alters their neural pathways involved in energy homeostasis regulation. Early postnatal exposure of obesity-resistant offspring to the milk of genetically obese dams alters their hypothalamic pathways involved in energy homeostasis causing them to become obese when fed a high fat diet as adults. Finally, short-term exercise begun in the early post-weaning period increases the sensitivity to the anorectic effects of leptin and protects obesity-prone offspring from becoming obese for months exercise cessation. Such studies suggest that early identification of obesity-prone humans and of the factors that can prevent them from becoming obese could provide an effective strategy for preventing the world wide epidemic of obesity.
Collapse
Affiliation(s)
- Barry E Levin
- Neurology Service (127C), Veterans Administration Medical Center, and Department of Neurology and Neurosciences, New Jersey Medical School, E. Orange, Newark, NJ 07018-1095, USA.
| |
Collapse
|
28
|
Roy S, Trudeau K, Roy S, Behl Y, Dhar S, Chronopoulos A. New Insights into Hyperglycemia-induced Molecular Changes in Microvascular Cells. J Dent Res 2009; 89:116-27. [DOI: 10.1177/0022034509355765] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Hyperglycemia is the most prevalent characteristic of diabetes and plays a central role in mediating adverse effects on vascular cells during the progression of diabetic vascular complications. In diabetic microangiopathy, hyperglycemia induces biochemical and molecular changes in microvascular cells that ultimately progress to retinal, renal, and neural complications and extends to other complications, including advanced periodontal disease. In this review, we describe changes involving basement membrane thickening, tissue remodeling, gap junctions, inflammation, cytokines, and transcription factors, and their effects on the pathogenesis of diabetic microvascular complications. The majority of the changes described relate to retinal microangiopathy, since ultrastructural, structural, and biochemical alterations have been well-characterized in this tissue.
Collapse
Affiliation(s)
- S. Roy
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston University, 650 Albany Street, Boston, MA 02118, USA
| | - K. Trudeau
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston University, 650 Albany Street, Boston, MA 02118, USA
| | - S. Roy
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston University, 650 Albany Street, Boston, MA 02118, USA
| | - Y. Behl
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston University, 650 Albany Street, Boston, MA 02118, USA
| | - S. Dhar
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston University, 650 Albany Street, Boston, MA 02118, USA
| | - A. Chronopoulos
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston University, 650 Albany Street, Boston, MA 02118, USA
| |
Collapse
|
29
|
Abstract
During postnatal cerebellar development, Purkinje cells form the most elaborate dendritic trees among neurons in the brain, which have been of great interest to many investigators. This article overviews various examples of cellular and molecular mechanisms of formation of Purkinje cell dendrites as well as the methodological aspects of investigating those mechanisms.
Collapse
Affiliation(s)
- Masahiko Tanaka
- Department of Cellular Biophysics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan.
| |
Collapse
|
30
|
Effects of Intracisternal Administration of Insulin on the Time Dynamics of Behavior in Rats. ACTA ACUST UNITED AC 2009; 39:671-82. [DOI: 10.1007/s11055-009-9181-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Indexed: 11/26/2022]
|
31
|
Chan CB, Ye K, Chan CB, Ye K. PIKE GTPase are phosphoinositide-3-kinase enhancers, suppressing programmed cell death. J Cell Mol Med 2007; 11:39-53. [PMID: 17367500 PMCID: PMC4401219 DOI: 10.1111/j.1582-4934.2007.00014.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Accepted: 01/05/2007] [Indexed: 01/05/2023] Open
Abstract
Phosphoinositide-3-kinase enhancers (PIKE) are GTP-binding proteins that posses anti-apoptotic functions. The PIKE family includes three members, PIKE-L, PIKE-S and PIKE-A, which are originated from a single gene (CENTG1) through alternative splicing or differential transcription initiation. Both PIKE-S and PIKE-L bind to phosphoinositide-3-kinase (PI3K) and enhance its activity. PIKE-A does not interplay with PI3K. Instead, it interacts with the downstream effector Akt and promotes its activity. These actions are mediated by their GTPase activity. Because both PI3K and Akt are important effectors in the growth factor-mediated signaling which triggers cellular growth and acts against apoptosis, PIKEs therefore serve as the molecular switch that their activation are crucial for growth factors to exert their physiological functions. In this review, the current understanding of different PIKE isoforms in growth factors-induced anti-apoptotic function will be discussed. Moreover, the role of PIKE in the survival and invasion activity of cancer cells will also be introduced.
Collapse
Affiliation(s)
- Chi Bun Chan
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Chi Bun Chan
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
32
|
Cole AR, Astell A, Green C, Sutherland C. Molecular connexions between dementia and diabetes. Neurosci Biobehav Rev 2007; 31:1046-63. [PMID: 17544131 DOI: 10.1016/j.neubiorev.2007.04.004] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 03/27/2007] [Accepted: 04/11/2007] [Indexed: 12/29/2022]
Abstract
Recent evidence suggests that the molecular defects associated with the development of diabetes also contribute to an increased risk of all types of dementia, including Alzheimer's disease, vascular dementia and Pick's disease. Indeed, the presence of type II diabetes mellitus results in a two to three fold higher risk of developing dementia [Fontbonne et al., 2001. Changes in cognitive abilities over a 4-year period are unfavourably affected in elderly diabetic subjects: results of the Epidemiology of Vascular Aging Study. Diabetes Care 24, 366-370; Gregg et al., 2000. Is diabetes associated with cognitive impairment and cognitive decline among older women? Study of Osteoporotic Fractures Research Group. Archives of Internal Medicine 160, 174-180; Peila et al., 2002. Type 2 diabetes, APOE gene, and the risk for dementia and related pathologies: The Honolulu-Asia Aging Study. Diabetes 51, 1256-1262]. There are currently 250 million people worldwide (>2 million in the UK) diagnosed with diabetes, and this number is predicted to double within the next 20 years, therefore the associated risk translates into a potential explosion in the appearance of dementia in the population. This review primarily focuses on the proposed molecular links between insulin action, Diabetes and Alzheimer's disease, while discussing the potential for therapeutic intervention to alleviate these disorders. In particular, we will review the regulation of glycogen synthase kinase-3 (GSK-3) and its neuronal substrates.
Collapse
Affiliation(s)
- Adam R Cole
- Division of Pathology and Neurosciences, University of Dundee, Ninewells Hospital, Dundee, Scotland, UK
| | | | | | | |
Collapse
|
33
|
van der Heide LP, Ramakers GMJ, Smidt MP. Insulin signaling in the central nervous system: Learning to survive. Prog Neurobiol 2006; 79:205-21. [PMID: 16916571 DOI: 10.1016/j.pneurobio.2006.06.003] [Citation(s) in RCA: 299] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2006] [Revised: 06/19/2006] [Accepted: 06/23/2006] [Indexed: 12/23/2022]
Abstract
Insulin is best known for its role in peripheral glucose homeostasis. Less studied, but not less important, is its role in the central nervous system. Insulin and its receptor are located in the central nervous system and are both implicated in neuronal survival and synaptic plasticity. Interestingly, over the past few years it has become evident that the effects of insulin, on neuronal survival and synaptic plasticity, are mediated by a common signal transduction cascade, which has been identified as "the PI3K route". This route has turned out to be a major integrator of insulin signaling in the brain. A pronounced feature of this insulin-activated route is that it promotes survival by directly inactivating the pro-apoptotic machinery. Interestingly, it is this same route that is required for the induction of long-term potentiation and depression, basic processes underlying learning and memory. This leads to the hypothesis that the PI3K route forms a direct link between learning and memory and neuronal survival. The implications of this hypothesis are far reaching, since it provides an explanation why insulin has beneficial effects on learning and memory and how synaptic activity can prevent cellular degeneration. Applying this knowledge may provide novel therapeutic approaches in the treatment of neurodegenerative diseases such as Alzheimer's disease.
Collapse
Affiliation(s)
- Lars P van der Heide
- Rudolf Magnus Institute of Neuroscience, Department of Pharmacology and Anatomy, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands.
| | | | | |
Collapse
|
34
|
Fischer-Posovszky P, Tornqvist H, Debatin KM, Wabitsch M. Inhibition of death-receptor mediated apoptosis in human adipocytes by the insulin-like growth factor I (IGF-I)/IGF-I receptor autocrine circuit. Endocrinology 2004; 145:1849-59. [PMID: 14691011 DOI: 10.1210/en.2003-0985] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adipose tissue mass is reflected by the volume and the number of adipocytes and is subject to homeostatic regulation involving cell death mechanisms. In this study we have investigated the mechanisms of apoptosis in human preadipocytes and adipocytes that may play a role in the regulation of adipose tissue mass. We found that death receptors (CD95, TNF-related apoptosis-inducing ligand receptors 1 and 2, and TNF receptor 1) are expressed in human fat cells and that apoptosis can be induced by specific ligands. Sensitivity to apoptosis could be stimulated by an inhibitor of biosynthesis. In addition, inhibition of auto-/paracrine action of IGF-I dramatically sensitizes human adipocytes for death ligand-induced apoptosis. Phosphoinositide 3-kinase and, to a weaker extent, p38 MAPK are involved in IGF-I-mediated survival. IGF-I protects human fat cells from apoptosis by maintaining the expression of antiapoptotic proteins, Bcl-x(L) and Fas-associated death domain-like IL-1-converting enzyme inhibitory protein. In conclusion, we identified mechanisms of apoptosis induction in human fat cells. We furthermore demonstrate that human fat cells protect themselves from apoptosis by IGF-I in an auto-/paracrine manner.
Collapse
|
35
|
Schubert M, Gautam D, Surjo D, Ueki K, Baudler S, Schubert D, Kondo T, Alber J, Galldiks N, Küstermann E, Arndt S, Jacobs AH, Krone W, Kahn CR, Brüning JC. Role for neuronal insulin resistance in neurodegenerative diseases. Proc Natl Acad Sci U S A 2004; 101:3100-5. [PMID: 14981233 PMCID: PMC365750 DOI: 10.1073/pnas.0308724101] [Citation(s) in RCA: 474] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Impairment of insulin signaling in the brain has been linked to neurodegenerative diseases. To test the hypothesis that neuronal insulin resistance contributes to defects in neuronal function, we have performed a detailed analysis of brain/neuron-specific insulin receptor knockout (NIRKO) mice. We find that NIRKO mice exhibit a complete loss of insulin-mediated activation of phosphatidylinositol 3-kinase and inhibition of neuronal apoptosis. In intact animals, this loss results in markedly reduced phosphorylation of Akt and GSK3 beta, leading to substantially increased phosphorylation of the microtubule-associated protein Tau, a hallmark of neurodegenerative diseases. Nevertheless, these animals exhibit no alteration in neuronal proliferation/survival, memory, or basal brain glucose metabolism. Thus, lack of insulin signaling in the brain may lead to changes in Akt and GSK3 beta activity and Tau hyperphosphorylation but must interact with other mechanisms for development of Alzheimer's disease.
Collapse
Affiliation(s)
- Markus Schubert
- Institute for Genetics, University of Cologne, D-50931 Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Varela-Nieto I, de la Rosa EJ, Valenciano AI, León Y. Cell death in the nervous system: lessons from insulin and insulin-like growth factors. Mol Neurobiol 2003; 28:23-50. [PMID: 14514984 DOI: 10.1385/mn:28:1:23] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2002] [Accepted: 02/28/2003] [Indexed: 12/11/2022]
Abstract
Programmed cell death is an essential process for proper neural development. Cell death, with its similar regulatory and executory mechanisms, also contributes to the origin or progression of many or even all neurodegenerative diseases. An understanding of the mechanisms that regulate cell death during neural development may provide new targets and tools to prevent neurodegeneration. Many studies that have focused mainly on insulin-like growth factor-I (IGF-I), have shown that insulin-related growth factors are widely expressed in the developing and adult nervous system, and positively modulate a number of processes during neural development, as well as in adult neuronal and glial physiology. These factors also show neuroprotective effects following neural damage. Although some specific actions have been demonstrated to be anti-apoptotic, we propose that a broad neuroprotective role is the foundation for many of the observed functions of the insulin-related growth factors, whose therapeutical potential for nervous system disorders may be greater than currently accepted.
Collapse
Affiliation(s)
- Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Arturo Duperier 4, E-28029 Madrid, Spain.
| | | | | | | |
Collapse
|
37
|
Ursø B, Niesler CU, O'Rahilly S, Siddle K. Comparison of anti-apoptotic signalling by the insulin receptor and IGF-I receptor in preadipocytes and adipocytes. Cell Signal 2001; 13:279-85. [PMID: 11306245 DOI: 10.1016/s0898-6568(01)00130-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We compared the effectiveness of insulin receptor (IR) and type I insulin-like growth factor (IGF) receptor (IGFR) cytoplasmic domains in mediating anti-apoptotic effects in 3T3-L1 preadipocytes and adipocytes. We used TrkC/IR and TrkC/IGFR chimeras, stably expressed in these cells and stimulated with neurotrophin-3 (NT-3), so as to avoid interference from endogenous receptors. After 24-h serum deprivation, 10% of preadipocytes and 2% of adipocytes appeared apoptotic as determined by fluorescence-activated cell sorter (FACS) analysis of cells stained with propidium iodide (PI) and Annexin V. When NT-3 was added, the two chimeras inhibited apoptosis to the same extent by 80% in preadipocytes and 50% in adipocytes. Mutation of juxtamembrane tyrosines (IR Y960F, IGFR Y950F) abrogated these anti-apoptotic effects. Qualitatively similar results were obtained by determination of viable rather than apoptotic cells. We conclude that IR and IGFR have equal potential to inhibit apoptosis in cell backgrounds, which are normally responsive to either IGF-I or insulin.
Collapse
Affiliation(s)
- B Ursø
- University of Cambridge, Department of Clinical Biochemistry, Addenbrooke's Hospital, Hills Road, CB2 2QR, Cambridge, UK
| | | | | | | |
Collapse
|
38
|
Abstract
The objective of the present study was to examine the effect of antenatal or postnatal treatment with corticosteroids on the NMDA receptor, one of the mediators of both normal brain development and hypoxic-ischemic injury, by determining the characteristics of the receptor MK-801 binding site in untreated and corticosteroid-treated fetal and newborn lambs. (3)H-MK-801 binding was performed in cerebral cortical cell membranes from fetal sheep at 88, 120, and 136 d gestation (term = 150 d), and from 5-d-old lambs and adult ewes. Animals were randomized to receive dexamethasone [fetuses: 6 mg, i.m. every 12 hr for four doses to mother; lambs: 0.01 mg/kg (low dose) or 0.25 mg/kg (high dose) every 12 hr for four doses] or placebo. During development, B(max) (apparent number of receptors) increased, reaching a maximum in 5-d-old lambs (p < 0.05) and decreasing in the adult brain. K(d) (dissociation constant) did not change, suggesting that receptor affinity was not altered during maturation. Dexamethasone treatment had no effect on MK-801 binding in the fetus or adult, but in lambs was associated with a significant decrease in B(max) from 2.17 +/- 0.18 pmol/mg protein in placebo-treated animals to 1.65 +/- 0.8 and 1.62 +/- 0.07 pmol/mg protein in low-dose and high-dose animals, respectively. Affinity for (3)H-MK-801 decreased 20% after dexamethasone treatment in lambs only (p < 0.05). Thus, dexamethasone treatment appears to modify the NMDA receptor only during a specific period of brain development.
Collapse
|
39
|
Clayton H, Turner J, Swift S, James R, Bell P. Supplementation of islet culture medium with insulin may have a beneficial effect on islet secretory function. Pancreas 2001; 22:72-4. [PMID: 11138975 DOI: 10.1097/00006676-200101000-00013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent reports suggest that apoptosis resulting from the disruption of the normal cell-matrix relationship (anoikis) during islet isolation could lead to a loss of islet tissue in culture. Insulin is known to have a role in cell growth and survival, and this study was undertaken to assess any beneficial effect on islets by supplementing the islet culture medium with insulin. Human and porcine islets were cultured in medium supplemented with 0, 10, 100, and 1,000 ng x mL(-1) insulin. Secretory function was assessed by perifusion at days 1 and 8. The results demonstrated a significant variation in stimulation index between isolations for human islets, but there was no effect relating to the concentration of insulin in the medium or time in culture. For porcine islets, there was a significant (p < 0.001) improvement in secretory function for islets cultured in 10 and 100 ng x mL(-1) insulin, relative to 0 and 1,000 ng x mL(-1) insulin. There was no interisolation variation or effect of time in culture. In conclusion, the secretory function of porcine islets benefited from the addition of 10 to 100 ng x mL(-1) insulin to the culture medium, but interisolation variation in human islet secretory function did not allow any specific effect of the insulin to be determined.
Collapse
Affiliation(s)
- H Clayton
- Department of Surgery, University of Leicester, Leicester Royal Infirmary, United Kingdom.
| | | | | | | | | |
Collapse
|
40
|
Abstract
There is an obesity epidemic in the industrialized world that is not simply explained by excess energy intake and decreased energy expenditure. Persistent obesity develops when genetically predisposed individuals are in a chronic state of positive energy balance. Once established, the obese body weight is avidly defended against both over- and underfeeding. Animal studies have shown that lean individuals who are genetically predisposed toward obesity have abnormalities of neural function that prime them to become obese when caloric density of the diet is raised. These neural abnormalities are gradually "corrected" as obesity becomes fully developed, suggesting that obesity is the normal state for such individuals. Thus, defense of the obese body weight may be perpetuated by the formation of new neural circuits involved in energy-homeostasis pathways that are not then easily abolished. Such neural plasticity can occur in both adult life and during nervous-system development. Early pre- and postnatal metabolic conditions (maternal diabetes, obesity, undernutrition) can lead genetically predisposed offspring to become even more obese as adults. This enhanced obesity is associated with altered brain neural circuitry, and these changes can then be passed on to subsequent generations in a feed-forward cycle of ever-increasing body weight. Thus, the metabolic perturbations associated with obesity during both brain development and adult life can produce "metabolic imprinting" on genetically predisposed neural circuits involved in energy homeostasis. Drugs that reduce body weight decrease the defended body weight and alter neural pathways involved in energy homeostasis but have no permanent effect on body weight or neural function in most individuals. Thus, early intervention in mothers, infants, children, and adults may be the only way to prevent the formation of permanent neural connections that promote and perpetuate obesity in genetically predisposed individuals.
Collapse
Affiliation(s)
- B E Levin
- Department of Neurosciences, New Jersey Medical School, Newark, New Jersey, USA.
| |
Collapse
|
41
|
Brunstrom JE, Pearlman AL. Growth factor influences on the production and migration of cortical neurons. Results Probl Cell Differ 2000; 30:189-215. [PMID: 10857190 DOI: 10.1007/978-3-540-48002-0_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- J E Brunstrom
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | |
Collapse
|
42
|
Levin BE. The obesity epidemic: metabolic imprinting on genetically susceptible neural circuits. OBESITY RESEARCH 2000; 8:342-7. [PMID: 10933311 DOI: 10.1038/oby.2000.41] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The apparent obesity epidemic in the industrialized world is not explained completely by increased food intake or decreased energy expenditure. Once obesity develops in genetically predisposed individuals, their obese body weight is avidly defended against chronic caloric restriction. In animals genetically predisposed toward obesity, there are multiple abnormalities of neural function that prime them to become obese when dietary caloric density and quantity are raised. Once obesity is fully developed, these abnormalities largely disappear. This suggests that obesity might be the normal state for such individuals. Formation of new neural circuits involved in energy homeostasis might underlie the near permanence of the obese body weight. Such neural plasticity can occur during both nervous system development and in adult life. Maternal diabetes, obesity, and undernutrition have all been associated with obesity in the offspring of such mothers, especially in genetically predisposed individuals. Altered brain neural circuitry and function often accompanies such obesity. This enhanced obesity may then be passed on to subsequent generations in a feed-forward, upward spiral of increasing body weight across generations. Such findings suggest a form of "metabolic imprinting" upon genetically predisposed neural circuits involved in energy homeostasis. Centrally acting drugs used for obesity treatment lower the defended body weight and alter the function of neural pathways involved in energy homeostasis. But they generally have no permanent effect on body weight or neural function. Thus, early identification of obesity-prone mothers, infants, and adults and treatment of early obesity may be the only way to prevent the formation of permanent neural connections that promote and perpetuate obesity in genetically predisposed individuals.
Collapse
Affiliation(s)
- B E Levin
- Department of Neurosciences, NJ Medical School, Newark 07103, USA.
| |
Collapse
|
43
|
Tanaka M, Momoi T, Marunouchi T. In situ detection of activated caspase-3 in apoptotic granule neurons in the developing cerebellum in slice cultures and in vivo. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2000; 121:223-8. [PMID: 10876036 DOI: 10.1016/s0165-3806(00)00041-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Some granule neurons naturally undergo apoptosis in the external granular layer (EGL) of the postnatally developing cerebellum. In the present study, we examined the involvement of caspase-3 in this apoptosis using an organotypic slice culture system of postnatal rat cerebellum and an antibody specific for the active form of caspase-3 (p20/17). Double staining by immunohistochemistry against p20/17 and in situ nick-end labeling showed that p20/17 was present in some of the apoptotic EGL neurons. A similar staining pattern was also observed in the postnatal cerebellum in vivo. Double positive cells were observed more frequently when T7 DNA polymerase was used for the DNA fragmentation labeling in place of terminal deoxynucleotidyl transferase, by which apoptotic cells at earlier stages were thought to be labeled. Taken together, whereas caspase-3 was shown to be activated in some of the apoptotic EGL neurons in the developing cerebellum, activation of caspase-3 in some apoptotic EGL neurons may occur before they become positive on DNA fragmentation labeling. In addition, there may be another mechanism of EGL neuron apoptosis that is independent of caspase-3.
Collapse
Affiliation(s)
- M Tanaka
- Division of Cell Biology, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, 470-1192, Aichi, Japan.
| | | | | |
Collapse
|
44
|
Contestabile A. Roles of NMDA receptor activity and nitric oxide production in brain development. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2000; 32:476-509. [PMID: 10760552 DOI: 10.1016/s0165-0173(00)00018-7] [Citation(s) in RCA: 281] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
The concept that neural activity is important for brain maturation has focused much research interest on the developmental role of the NMDA receptor, a key mediator of experience-dependent synaptic plasticity. However, a mechanism able to link spatial and temporal parameters of synaptic activity during development emerged as a necessary condition to explain how axons segregate into a common brain region and make specific synapses on neuronal sub-populations. To comply with this developmental constraint, it was proposed that nitric oxide (NO), or other substances having similar chemical and biological characteristics, could act as short-lived, activity-dependent spatial signals, able to stabilize active synapses by diffusing through a local volume of tissue. The present article addresses this issue, by reviewing the experimental evidence for a correlated role of the activity of the NMDA receptor and the production of NO in key steps of neural development. Evidence for such a functional coupling emerges not only concerning synaptogenesis and formation of neural maps, for which it was originally proposed, but also for some earlier phases of neurogenesis, such as neural cell proliferation and migration. Regarding synaptogenesis and neural map formation in some cases, there is so far no conclusive experimental evidence for a coupled functional role of NMDA receptor activation and NO production. Some technical problems related to the use of inhibitors of NO formation and of gene knockout animals are discussed. It is also suggested that other substances, known to act as spatial signals in adult synaptic plasticity, could have a role in developmental plasticity. Concerning the crucial developmental phase of neuronal survival or elimination through programmed cell death, the well-documented survival role related to NMDA receptor activation also starts to find evidence for a concomitant requirement of downstream NO production. On the basis of the reviewed literature, some of the major controversial issues are addressed and, in some cases, suggestions for possible future experiments are proposed.
Collapse
Affiliation(s)
- A Contestabile
- Department of Biology, University of Bologna, Via Selmi 3, 40126, Bologna, Italy.
| |
Collapse
|
45
|
Abstract
Apoptosis is now recognized as a normal feature in the development of the nervous system and may also play a role in neurodegenerative diseases and aging. This phenomenon has been investigated intensively during the last 6-7 years, and the progress made in this field is reviewed here. Besides a few in vivo studies, a variety of neuronal preparations from various parts of the brain, the majority of which were primary cultures, and some cell lines have been investigated. Several apoptosis-inducing agents have been identified, and these include lack of neurotrophic support, neurotransmitters, neurotoxicants, modulators of protein phosphorylation and calcium homeostasis, DNA-damaging agents, oxidative stress, nitric oxide, and ceramides. The precise signaling cascade is not well established, and there are lacunae in many suggested pathways. However, it appears certain that the Bcl family of proteins is involved in the apoptotic pathway, and these proteins in turn affect the processing of interleukin-1beta converting enzyme (ICE)/caspases. The available evidence suggests that there may be several apoptotic pathways that may depend on the cell type and the inducing agent, and most of the pathways may converge at the ICE/caspases step.
Collapse
Affiliation(s)
- P S Sastry
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, India
| | | |
Collapse
|
46
|
|
47
|
Hamada Y, Kadokawa Y, Okabe M, Ikawa M, Coleman JR, Tsujimoto Y. Mutation in ankyrin repeats of the mouse Notch2 gene induces early embryonic lethality. Development 1999; 126:3415-24. [PMID: 10393120 DOI: 10.1242/dev.126.15.3415] [Citation(s) in RCA: 197] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Notch family genes encode transmembrane proteins involved in cell-fate determination. Using gene targeting procedures, we disrupted the mouse Notch2 gene by replacing all but one of the ankyrin repeat sequences in the cytoplasmic domain with the E. coli (beta)-galactosidase gene. The mutant Notch2 gene encodes a 380 kDa Notch2-(beta)-gal fusion protein with (beta)-galactosidase activity. Notch2 homozygous mutant mice die prior to embryonic day 11.5, whereas heterozygotes show no apparent abnormalities and are fully viable. Analysis of Notch2 expression patterns, revealed by X-gal staining, demonstrated that the Notch2 gene is expressed in a wide variety of tissues including neuroepithelia, somites, optic vesicles, otic vesicles, and branchial arches, but not heart. Histological studies, including in situ nick end labeling procedures, showed earlier onset and higher incidence of apoptosis in homozygous mutant mice than in heterozygotes or wild type mice. Dying cells were particularly evident in neural tissues, where they were seen as early as embryonic day 9.5 in Notch2-deficient mice. Cells from Notch2 mutant mice attach and grow normally in culture, demonstrating that Notch2 deficiency does not interfere with cell proliferation and that expression of the Notch2-(beta)-gal fusion protein is not toxic per se. In contrast to Notch1-deficient mice, Notch2 mutant mice did not show disorganized somitogenesis, nor did they fail to properly regulate the expression of neurogenic genes such as Hes-5 or Mash1. In situ hybridization studies show no indication of altered Notch1 expression patterns in Notch2 mutant mice. The results indicate that Notch2 plays an essential role in postimplantation development in mice, probably in some aspect of cell specification and/or differentiation, and that the ankyrin repeats are indispensable for its function.
Collapse
Affiliation(s)
- Y Hamada
- National Institute for Basic Biology, Okazaki, Aichi 444-8585, Japan.
| | | | | | | | | | | |
Collapse
|
48
|
DeCoster MA, Schabelman E, Tombran-Tink J, Bazan NG. Neuroprotection by pigment epithelial-derived factor against glutamate toxicity in developing primary hippocampal neurons. J Neurosci Res 1999; 56:604-10. [PMID: 10374815 DOI: 10.1002/(sici)1097-4547(19990615)56:6<604::aid-jnr6>3.0.co;2-b] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Pigment epithelial-derived factor (PEDF) has been shown to be a survival factor for cerebellar granule neurons. Here we investigated the ability of PEDF to enhance the survival of hippocampal neurons in culture, and to protect these neurons against acute glutamate toxicity. Hippocampal neurons prepared from 1- to 3-day postnatal rat brain were cultured for either 7 or 14 days in vitro (DIV). At 14 DIV, neurons were only slightly protected (13% +/- 4%) against 50 microM glutamate toxicity when treated with 1 microg/ml of PEDF for 3 successive days before glutamate exposure as measured by lactate dehydrogenase (LDH) release. In comparison, basic fibroblast growth factor (bFGF) at 10 ng/ml for the same treatment period protected 58% +/- 8% of the neurons against glutamate. Using quantitative image analysis of digitized micrographs, we found that the average size of neurons in young, developing hippocampal cultures (7 DIV), was greatly decreased by treatment with 50 microM glutamate. Treatment for up to 5 successive days with 1 microg/ml of PEDF before glutamate addition dramatically increased the average hippocampal neuron soma size, compared to cells treated with glutamate alone. Thus, PEDF may promote the growth of hippocampal neurons, and, if added to developing hippocampal neurons, can also protect these cells from subsequent injury, such as the excitotoxicity of glutamate.
Collapse
Affiliation(s)
- M A DeCoster
- Louisiana State University Medical Center, Neuroscience Center, New Orleans 70112, USA
| | | | | | | |
Collapse
|
49
|
Loftus TM, Kuhajda FP, Lane MD. Insulin depletion leads to adipose-specific cell death in obese but not lean mice. Proc Natl Acad Sci U S A 1998; 95:14168-72. [PMID: 9826672 PMCID: PMC24345 DOI: 10.1073/pnas.95.24.14168] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mutation of the obese gene produces obesity, hyperinsulinemia, and compensatory "overexpression" of the defective gene. As insulin activates obese gene expression, it seemed possible that hyperinsulinemia might be responsible for overexpression of the gene. To address this question we rapidly neutralized circulating insulin by injection of an insulin antibody. Unexpectedly, insulin depletion in obese (ob/ob or db/db) mice caused massive adipose RNA degradation confirmed by histological analysis to result from adipocyte cell death by a largely necrotic mechanism. This effect was not observed in lean littermates and was completely corrected by coadministration of insulin. Comparison of multiple tissues demonstrated that the effect was restricted to adipose tissue. Insulin depletion in obese mice by administration of streptozotocin also led to cell death, but this death was less extensive and appeared to be apoptotic in mechanism. Thus insulin may promote the survival side of the physiological balance between adipocyte survival and death.
Collapse
Affiliation(s)
- T M Loftus
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
50
|
Tanaka M, Sawada M, Miura M, Marunouchi T. Insulin-like growth factor-I analogue prevents apoptosis mediated through an interleukin-1 beta converting enzyme (caspase-1)-like protease of cerebellar external granular layer neurons: developmental stage-specific mechanisms of neuronal cell death. Neuroscience 1998; 84:89-100. [PMID: 9522365 DOI: 10.1016/s0306-4522(97)00518-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Using an organotypic slice culture system of neonatal rat cerebellum, we examined developmental stage-specific mechanisms of cell death of granule neurons. This culture system allows a serial process of granule neuron development including their proliferation during the early culture period and the proceeding migration from the external granular layer to the internal granular layer in the presence of a supraphysiological concentration (5 micrograms/ml) of insulin. Insulin deprivation induced apoptosis of granule neurons in external granular layer but not in internal granular layer. A truncated analogue of insulin-like growth factor-I (des (1-3) insulin-like growth factor-I) prevented this apoptosis at a concentration of 65-650 ng/ml. Some apoptotic granule neurons expressed proliferating cell nuclear antigen but not TAG-1, a marker protein of the postmitotic and premigratory granule neurons. Thus, this apoptosis occurred at a specific stage in granule neuron development: at the stage before TAG-1 expression and at least partly at the proliferative state. Ac-YVAD-CHO, an inhibitor of interleukin-1 beta converting enzyme (caspase-1)-like proteases, had a protective effect on this apoptosis. Interleukin-1 beta converting enzyme (caspase-1)-like protease activity increased under the apoptosis-induced condition. High concentration of K+, which is known to prevent granule neuron apoptosis in dissociated cultures, had a partial protective effect on this apoptosis. These findings suggest that (i) cerebellar granule neurons fall into apoptosis at the specific developmental stage unless stimulated by insulin-like growth factor-I (analogue), (ii) this apoptosis is mediated through an interleukin-1 beta converting enzyme-like protease, and (iii) this apoptosis consists of K(+)-sensitive and K(+)-insensitive components.
Collapse
Affiliation(s)
- M Tanaka
- Division of Cell Biology, Fujita Health University, Toyoake, Japan
| | | | | | | |
Collapse
|