1
|
Mirabelli M, Chiefari E, Arcidiacono B, Salatino A, Pascarella A, Morelli M, Credendino SC, Brunetti FS, Di Vito A, Greco A, Huin V, Nicoletti F, Pierantoni GM, Fedele M, Aguglia U, Foti DP, Brunetti A. HMGA1 deficiency: a pathogenic link between tau pathology and insulin resistance. EBioMedicine 2025; 115:105700. [PMID: 40233659 PMCID: PMC12019291 DOI: 10.1016/j.ebiom.2025.105700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Growing evidence links tau-related neurodegeneration with insulin resistance and type 2 diabetes (T2D), though the underlying mechanisms remain unclear. Our previous research identified HMGA1 as crucial for insulin receptor (INSR) expression, with defects in the HMGA1 gene associated with insulin resistance and T2D. Here, we explore HMGA1 deficiency as a potential contributor to tauopathies, such as Alzheimer's disease (AD), and its connection to insulin resistance. METHODS Immunoblot analyses, protein-DNA interaction studies, ChIP-qPCR, and reporter gene assays were conducted in human and mouse neuronal cell models. Tau immunohistochemistry, behavioural studies, and brain glucose metabolism were analysed in Hmga1-knockout mice. Additionally, a case-control study investigated the relationship between HMGA1 and tau pathology in patients with tauopathy, carrying or not the HMGA1 rs146052672 variant, known to reduce HMGA1 protein levels and increase the risk of insulin resistance and T2D. FINDINGS We show that HMGA1 regulates tau protein expression primarily through the specific repression of MAPT gene transcription. In both human neuronal cells and primary mouse neurons, tau mRNA and protein levels were inversely correlated with HMGA1 expression. This inverse relationship was further confirmed in the brain of Hmga1-knockout mice, where tau was overexpressed, INSR was downregulated, and brain glucose uptake was impaired. Additionally, the rs146052672 variant was more common in patients with tauopathy (12/69, 17.4%) than in controls (10/200, 5.0%) (p = 0.001), and carriers of this variant exhibited more severe disease progression and poorer therapeutic outcomes. INTERPRETATION These findings suggest that HMGA1 deficiency may drive tau pathology, linking tauopathies to insulin resistance and providing new insights into the relationship between metabolic and neurodegenerative disorders. Furthermore, our observation that over 17% of individuals with tauopathy exhibit a deficit in HMGA1 protein production could have significant clinical implications, potentially guiding the development of therapeutic strategies targeting this specific defect. FUNDING See acknowledgements section.
Collapse
Affiliation(s)
- Maria Mirabelli
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Biagio Arcidiacono
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Alessandro Salatino
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Angelo Pascarella
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Maurizio Morelli
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Sara C Credendino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Francesco S Brunetti
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Anna Di Vito
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Adelaide Greco
- Interdepartmental Centre of Veterinary Radiology, University of Naples "Federico II", Naples, Italy
| | - Vincent Huin
- University of Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, UMR-S1172, Team Alzheimer & Tauopathies, F-59000, Lille, France
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University of Rome "Sapienza", Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Giovanna M Pierantoni
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Monica Fedele
- Institute of Experimental Endocrinology and Oncology, CNR, Naples, Italy
| | - Umberto Aguglia
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Daniela P Foti
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Græcia", Catanzaro, Italy.
| | - Antonio Brunetti
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy.
| |
Collapse
|
2
|
Chen Y, Chen X, Luo Z, Kang X, Ge Y, Wan R, Wang Q, Han Z, Li F, Fan Z, Xie Y, Qi B, Zhang X, Yang Z, Zhang JH, Liu D, Xu Y, Wu D, Chen S. Exercise-Induced Reduction of IGF1R Sumoylation Attenuates Neuroinflammation in APP/PS1 Transgenic Mice. J Adv Res 2025; 69:279-297. [PMID: 38565402 PMCID: PMC11954827 DOI: 10.1016/j.jare.2024.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/03/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION Alzheimer's Disease (AD), a progressive neurodegenerative disorder, is marked by cognitive deterioration and heightened neuroinflammation. The influence of Insulin-like Growth Factor 1 Receptor (IGF1R) and its post-translational modifications, especially sumoylation, is crucial in understanding the progression of AD and exploring novel therapeutic avenues. OBJECTIVES This study investigates the impact of exercise on the sumoylation of IGF1R and its role in ameliorating AD symptoms in APP/PS1 mice, with a specific focus on neuroinflammation and innovative therapeutic strategies. METHODS APP/PS1 mice were subjected to a regimen of moderate-intensity exercise. The investigation encompassed assessments of cognitive functions, alterations in hippocampal protein expressions, neuroinflammatory markers, and the effects of exercise on IGF1R and SUMO1 nuclear translocation. Additionally, the study evaluated the efficacy of KPT-330, a nuclear export inhibitor, as an alternative to exercise. RESULTS Exercise notably enhanced cognitive functions in AD mice, possibly through modulations in hippocampal proteins, including Bcl-2 and BACE1. A decrease in neuroinflammatory markers such as IL-1β, IL-6, and TNF-α was observed, indicative of reduced neuroinflammation. Exercise modulated the nuclear translocation of SUMO1 and IGF1R in the hippocampus, thereby facilitating neuronal regeneration. Mutant IGF1R (MT IGF1R), lacking SUMO1 modification sites, showed reduced SUMOylation, leading to diminished expression of pro-inflammatory cytokines and apoptosis. KPT-330 impeded the formation of the IGF1R/RanBP2/SUMO1 complex, thereby limiting IGF1R nuclear translocation, inflammation, and neuronal apoptosis, while enhancing cognitive functions and neuron proliferation. CONCLUSION Moderate-intensity exercise effectively mitigates AD symptoms in mice, primarily by diminishing neuroinflammation, through the reduction of IGF1R Sumoylation. KPT-330, as a potential alternative to physical exercise, enhances the neuroprotective role of IGF1R by inhibiting SUMOylation through targeting XPO1, presenting a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Yisheng Chen
- Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaofeng Chen
- Department of Orthopaedics, National Regional Medical Center, Jinjiang Municipal Hospital,Shanghai Sixth People's Hospital, Fujian, Jinjiang,China.
| | - Zhiwen Luo
- Huashan Hospital, Fudan University, Shanghai, China
| | - Xueran Kang
- Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, China
| | - Yunshen Ge
- Huashan Hospital, Fudan University, Shanghai, China
| | - Renwen Wan
- Huashan Hospital, Fudan University, Shanghai, China
| | - Qian Wang
- Department of Central Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Zhihua Han
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Fangqi Li
- Huashan Hospital, Fudan University, Shanghai, China
| | - Zhongcheng Fan
- Department of Orthopaedic Surgery, Hainan Province Clinical Medical Center, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, China
| | - Yuchun Xie
- Jiangsu Province Geriatric Hospital, China
| | - Beijie Qi
- Huashan Hospital, Fudan University, Shanghai, China
| | - Xintao Zhang
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital Lianhua Road, Shenzhen City, Guangdong Province, China
| | - Zhenwei Yang
- Department of Orthopaedics, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - John H Zhang
- Department of Neurosurgery, Department of Physiology and Pharmacology, Department of Neurosurgery and Anesthesiology, School of Medicine, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92354, USA.
| | - Danping Liu
- Department of Orthopaedics, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China.
| | - Yuzhen Xu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China.
| | - Dongyan Wu
- Huashan Hospital, Fudan University, Shanghai, China.
| | - Shiyi Chen
- Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Hansman DS, Du J, Casson RJ, Peet DJ. Eye on the horizon: The metabolic landscape of the RPE in aging and disease. Prog Retin Eye Res 2025; 104:101306. [PMID: 39433211 PMCID: PMC11833275 DOI: 10.1016/j.preteyeres.2024.101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
To meet the prodigious bioenergetic demands of the photoreceptors, glucose and other nutrients must traverse the retinal pigment epithelium (RPE), a polarised monolayer of cells that lie at the interface between the outer retina and the choroid, the principal vascular layer of the eye. Recent investigations have revealed a metabolic ecosystem in the outer retina where the photoreceptors and RPE engage in a complex exchange of sugars, amino acids, and other metabolites. Perturbation of this delicate metabolic balance has been identified in the aging retina, as well as in age-related macular degeneration (AMD), the leading cause of blindness in the Western world. Also common in the aging and diseased retina are elevated levels of cytokines, oxidative stress, advanced glycation end-products, increased growth factor signalling, and biomechanical stress - all of which have been associated with metabolic dysregulation in non-retinal cell types and tissues. Herein, we outline the role of these factors in retinal homeostasis, aging, and disease. We discuss their effects on glucose, mitochondrial, lipid, and amino acid metabolism in tissues and cell types outside the retina, highlighting the signalling pathways through which they induce these changes. Lastly, we discuss promising avenues for future research investigating the roles of these pathological conditions on retinal metabolism, potentially offering novel therapeutic approaches to combat age-related retinal disease.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
4
|
Guldiken G, Karayagmurlu A, Kucukgergin C, Coskun M. VEGF, IGF-1 and FGF-2 Serum Levels in Children and Adolescents with Autism Spectrum Disorder with and without Bipolar Disorder. J Autism Dev Disord 2024; 54:3854-3862. [PMID: 37668852 DOI: 10.1007/s10803-023-06089-1] [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] [Accepted: 07/30/2023] [Indexed: 09/06/2023]
Abstract
PURPOSE To investigate serum levels of VEGF, IGF-1 and FGF-2, and relationships with several clinical characteristics in children and adolescents with autism spectrum disorder (ASD) with and without bipolar disorder (BD). METHOD 40 subjects with ASD + BD as study group, and 40 subjects with ASD as control group were included. Serum levels of VEGF, IGF-1, and FGF-2 were measured using commercial enzyme-linked immunosorbent assay kits. RESULTS The study group was significantly higher than the control group in terms of ASD severity, self-harming behavior and sleep disturbance. Serum VEGF and FGF-2 levels were significantly higher in the ASD + BD group than in the control group. There was no significant difference in serum IGF-1 levels between the two groups. There was no correlation between VEGF, IGF-1 and FGF-2 serum levels and ASD severity in the study group. However there was a negative correlation between VEGF levels and age at first diagnosis of BD, and a positive correlation between IGF-1 levels and the number of bipolar episodes in the study group. CONCLUSION Growth factors like VEGF and FGF-2 may be potential biomarkers of bipolar disorder in young subjects with ASD. Given the difficulty of clinical management of BD in young subjects with ASD, potential biomarkers would help clinicians in the diagnosis and follow up of BD in this special population. Further research is needed whether VEGF and FGF-2 can be potential biomarkers in the clinical management of young subjects with ASD and BD.
Collapse
Affiliation(s)
- Gokce Guldiken
- Health Ministry of Turkish Republic Reyhanlı State Hospital, Hatay, Turkey.
| | - Ali Karayagmurlu
- Department of Child and Adolescent Psychiatry, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Canan Kucukgergin
- Department of Medical Biochemistry, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Murat Coskun
- Department of Child and Adolescent Psychiatry, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| |
Collapse
|
5
|
Dzik KP, Flis DJ, Kaczor-Keller KB, Bytowska ZK, Karnia MJ, Ziółkowski W, Kaczor JJ. Spinal cord abnormal autophagy and mitochondria energy metabolism are modified by swim training in SOD1-G93A mice. J Mol Med (Berl) 2024; 102:379-390. [PMID: 38197966 PMCID: PMC10879285 DOI: 10.1007/s00109-023-02410-8] [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: 03/07/2023] [Revised: 11/16/2023] [Accepted: 12/12/2023] [Indexed: 01/11/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) may result from the dysfunctions of various mechanisms such as protein accumulation, mitophagy, and biogenesis of mitochondria. The purpose of the study was to evaluate the molecular mechanisms in ALS development and the impact of swim training on these processes. In the present study, an animal model of ALS, SOD1-G93A mice, was used with the wild-type mice as controls. Mice swam five times per week for 30 min. Mice were analyzed before ALS onset (70 days old), at ALS 1 disease onset (116 days old), and at the terminal stage of the disease ALS (130 days old), and compared with the corresponding ALS untrained groups and normalized to the wild-type group. Enzyme activity and protein content were analyzed in the spinal cord homogenates. The results show autophagy disruptions causing accumulation of p62 accompanied by low PGC-1α and IGF-1 content in the spinal cord of SOD1-G93A mice. Swim training triggered a neuroprotective effect, attenuation of NF-l degradation, less accumulated p62, and lower autophagy initiation. The IGF-1 pathway induces pathophysiological adaptation to maintain energy demands through anaerobic metabolism and mitochondrial protection. KEY MESSAGES: The increased protein content of p62 in the spinal cord of SOD1-G93A mice suggests that autophagic clearance and transportation are disrupted. Swim training attenuates neurofilament light destruction in the spinal cord of SOD1-G93A mice. Swim training reducing OGDH provokes suppression of ATP-consuming anabolic pathways. Swim training induces energy metabolic changes and mitochondria protection through the IGF-1 signaling pathway.
Collapse
Affiliation(s)
- Katarzyna Patrycja Dzik
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Bazynskiego 8, 80-309, Gdansk, Poland
| | - Damian Józef Flis
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Katarzyna Barbara Kaczor-Keller
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Magdalenka, Poland
| | - Zofia Kinga Bytowska
- Division of Bioenergetics and Physiology of Exercise, Faculty of Health Sciences With Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Mateusz Jakub Karnia
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Bazynskiego 8, 80-309, Gdansk, Poland
| | - Wiesław Ziółkowski
- Department of Rehabilitation Medicine, Faculty of Health Sciences With Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Jan Jacek Kaczor
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Bazynskiego 8, 80-309, Gdansk, Poland.
| |
Collapse
|
6
|
Zegarra-Valdivia JA, Pignatelli J, Nuñez A, Torres Aleman I. The Role of Insulin-like Growth Factor I in Mechanisms of Resilience and Vulnerability to Sporadic Alzheimer's Disease. Int J Mol Sci 2023; 24:16440. [PMID: 38003628 PMCID: PMC10671249 DOI: 10.3390/ijms242216440] [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: 10/11/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Despite decades of intense research, disease-modifying therapeutic approaches for Alzheimer's disease (AD) are still very much needed. Apart from the extensively analyzed tau and amyloid pathological cascades, two promising avenues of research that may eventually identify new druggable targets for AD are based on a better understanding of the mechanisms of resilience and vulnerability to this condition. We argue that insulin-like growth factor I (IGF-I) activity in the brain provides a common substrate for the mechanisms of resilience and vulnerability to AD. We postulate that preserved brain IGF-I activity contributes to resilience to AD pathology as this growth factor intervenes in all the major pathological cascades considered to be involved in AD, including metabolic impairment, altered proteostasis, and inflammation, to name the three that are considered to be the most important ones. Conversely, disturbed IGF-I activity is found in many AD risk factors, such as old age, type 2 diabetes, imbalanced diet, sedentary life, sociality, stroke, stress, and low education, whereas the Apolipoprotein (Apo) E4 genotype and traumatic brain injury may also be influenced by brain IGF-I activity. Accordingly, IGF-I activity should be taken into consideration when analyzing these processes, while its preservation will predictably help prevent the progress of AD pathology. Thus, we need to define IGF-I activity in all these conditions and develop a means to preserve it. However, defining brain IGF-I activity cannot be solely based on humoral or tissue levels of this neurotrophic factor, and new functionally based assessments need to be developed.
Collapse
Affiliation(s)
- Jonathan A. Zegarra-Valdivia
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain;
- Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain;
- School of Medicine, Universidad Señor de Sipán, Chiclayo 14000, Peru
| | - Jaime Pignatelli
- Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain;
- Cajal Institute (CSIC), 28002 Madrid, Spain
| | - Angel Nuñez
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| | - Ignacio Torres Aleman
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain;
- Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| |
Collapse
|
7
|
Nuñez A, Zegarra-Valdivia J, Fernandez de Sevilla D, Pignatelli J, Torres Aleman I. The neurobiology of insulin-like growth factor I: From neuroprotection to modulation of brain states. Mol Psychiatry 2023; 28:3220-3230. [PMID: 37353586 DOI: 10.1038/s41380-023-02136-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/30/2023] [Accepted: 06/13/2023] [Indexed: 06/25/2023]
Abstract
After decades of research in the neurobiology of IGF-I, its role as a prototypical neurotrophic factor is undisputed. However, many of its actions in the adult brain indicate that this growth factor is not only involved in brain development or in the response to injury. Following a three-layer assessment of its role in the central nervous system, we consider that at the cellular level, IGF-I is indeed a bona fide neurotrophic factor, modulating along ontogeny the generation and function of all the major types of brain cells, contributing to sculpt brain architecture and adaptive responses to damage. At the circuit level, IGF-I modulates neuronal excitability and synaptic plasticity at multiple sites, whereas at the system level, IGF-I intervenes in energy allocation, proteostasis, circadian cycles, mood, and cognition. Local and peripheral sources of brain IGF-I input contribute to a spatially restricted, compartmentalized, and timed modulation of brain activity. To better define these variety of actions, we consider IGF-I a modulator of brain states. This definition aims to reconcile all aspects of IGF-I neurobiology, and may provide a new conceptual framework in the design of future research on the actions of this multitasking neuromodulator in the brain.
Collapse
Affiliation(s)
- A Nuñez
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, Madrid, Spain
| | - J Zegarra-Valdivia
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIBERNED, Madrid, Spain
- Universidad Señor de Sipán, Chiclayo, Perú
| | - D Fernandez de Sevilla
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, Madrid, Spain
| | - J Pignatelli
- CIBERNED, Madrid, Spain
- Cajal Institute (CSIC), Madrid, Spain
| | - I Torres Aleman
- Achucarro Basque Center for Neuroscience, Leioa, Spain.
- CIBERNED, Madrid, Spain.
- Ikerbasque Science Foundation, Bilbao, Spain.
| |
Collapse
|
8
|
Zhang S, Zhang Y, Wen Z, Yang Y, Bu T, Bu X, Ni Q. Cognitive dysfunction in diabetes: abnormal glucose metabolic regulation in the brain. Front Endocrinol (Lausanne) 2023; 14:1192602. [PMID: 37396164 PMCID: PMC10312370 DOI: 10.3389/fendo.2023.1192602] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/25/2023] [Indexed: 07/04/2023] Open
Abstract
Cognitive dysfunction is increasingly recognized as a complication and comorbidity of diabetes, supported by evidence of abnormal brain structure and function. Although few mechanistic metabolic studies have shown clear pathophysiological links between diabetes and cognitive dysfunction, there are several plausible ways in which this connection may occur. Since, brain functions require a constant supply of glucose as an energy source, the brain may be more susceptible to abnormalities in glucose metabolism. Glucose metabolic abnormalities under diabetic conditions may play an important role in cognitive dysfunction by affecting glucose transport and reducing glucose metabolism. These changes, along with oxidative stress, inflammation, mitochondrial dysfunction, and other factors, can affect synaptic transmission, neural plasticity, and ultimately lead to impaired neuronal and cognitive function. Insulin signal triggers intracellular signal transduction that regulates glucose transport and metabolism. Insulin resistance, one hallmark of diabetes, has also been linked with impaired cerebral glucose metabolism in the brain. In this review, we conclude that glucose metabolic abnormalities play a critical role in the pathophysiological alterations underlying diabetic cognitive dysfunction (DCD), which is associated with multiple pathogenic factors such as oxidative stress, mitochondrial dysfunction, inflammation, and others. Brain insulin resistance is highly emphasized and characterized as an important pathogenic mechanism in the DCD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qing Ni
- Department of Endocrinology, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Ronsley R, Lazow M, Henry RK. Growth hormone after CNS tumor diagnosis: the fundamentals, fears, facts, and future directions. Pediatr Hematol Oncol 2023; 40:786-799. [PMID: 36939305 DOI: 10.1080/08880018.2023.2190765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 03/21/2023]
Abstract
Growth hormone deficiency (GHD) may occur in pediatric patients with central nervous system (CNS) tumors at initial tumor presentation or later as treatment-related sequelae. While it is well recognized that growth hormone (GH) has beneficial effects on growth and endocrinopathies, there's often hesitancy by clinicians to initiate GH therapy for GHD after CNS tumor diagnosis due to the perceived increased risk of tumor recurrence. The available data is described here and based on this review, there is no evidence of increased risk of tumor recurrence or secondary malignancy in patients treated with GH after CNS tumor diagnosis. Further understanding of tumor biology and presence of downstream GH targets including insulin-like growth factor-1 (IGF-1) and insulin receptor activity is still needed.
Collapse
Affiliation(s)
- Rebecca Ronsley
- Section of Hematology, Oncology & BMT, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Section of Hematology, Oncology & BMT, Department of Pediatrics, Seattle Children's Hospital, The University of Washington, Seattle, Washington, USA
| | - Margot Lazow
- Section of Hematology, Oncology & BMT, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Rohan K Henry
- Section of Endocrinology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
10
|
Ivan DC, Berve KC, Walthert S, Monaco G, Borst K, Bouillet E, Ferreira F, Lee H, Steudler J, Buch T, Prinz M, Engelhardt B, Locatelli G. Insulin-like growth factor-1 receptor controls the function of CNS-resident macrophages and their contribution to neuroinflammation. Acta Neuropathol Commun 2023; 11:35. [PMID: 36890580 PMCID: PMC9993619 DOI: 10.1186/s40478-023-01535-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 02/20/2023] [Indexed: 03/10/2023] Open
Abstract
Signaling by insulin-like growth factor-1 (IGF-1) is essential for the development of the central nervous system (CNS) and regulates neuronal survival and myelination in the adult CNS. In neuroinflammatory conditions including multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE), IGF-1 can regulate cellular survival and activation in a context-dependent and cell-specific manner. Notwithstanding its importance, the functional outcome of IGF-1 signaling in microglia/macrophages, which maintain CNS homeostasis and regulate neuroinflammation, remains undefined. As a result, contradictory reports on the disease-ameliorating efficacy of IGF-1 are difficult to interpret, together precluding its potential use as a therapeutic agent. To fill this gap, we here investigated the role of IGF-1 signaling in CNS-resident microglia and border associated macrophages (BAMs) by conditional genetic deletion of the receptor Igf1r in these cell types. Using a series of techniques including histology, bulk RNA sequencing, flow cytometry and intravital imaging, we show that absence of IGF-1R significantly impacted the morphology of both BAMs and microglia. RNA analysis revealed minor changes in microglia. In BAMs however, we detected an upregulation of functional pathways associated with cellular activation and a decreased expression of adhesion molecules. Notably, genetic deletion of Igf1r from CNS-resident macrophages led to a significant weight gain in mice, suggesting that absence of IGF-1R from CNS-resident myeloid cells indirectly impacts the somatotropic axis. Lastly, we observed a more severe EAE disease course upon Igf1r genetic ablation, thus highlighting an important immunomodulatory role of this signaling pathway in BAMs/microglia. Taken together, our work shows that IGF-1R signaling in CNS-resident macrophages regulates the morphology and transcriptome of these cells while significantly decreasing the severity of autoimmune CNS inflammation.
Collapse
Affiliation(s)
- Daniela C Ivan
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012, Bern, Switzerland
| | - Kristina Carolin Berve
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012, Bern, Switzerland
| | - Sabrina Walthert
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012, Bern, Switzerland
| | - Gianni Monaco
- Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - Katharina Borst
- Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - Elisa Bouillet
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012, Bern, Switzerland
| | - Filipa Ferreira
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Henry Lee
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012, Bern, Switzerland
| | - Jasmin Steudler
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012, Bern, Switzerland
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Marco Prinz
- Institute of Neuropathology, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012, Bern, Switzerland
| | - Giuseppe Locatelli
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012, Bern, Switzerland.
| |
Collapse
|
11
|
Protective role of IGF-1 and GLP-1 signaling activation in neurological dysfunctions. Neurosci Biobehav Rev 2022; 142:104896. [PMID: 36191807 DOI: 10.1016/j.neubiorev.2022.104896] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 09/09/2022] [Accepted: 09/26/2022] [Indexed: 11/24/2022]
Abstract
Insulin-like growth factor-1 (IGF-1), a pleiotropic polypeptide, plays an essential role in CNS development and maturation. Glucagon-like peptide-1 (GLP-1) is an endogenous incretin hormone that regulates blood glucose levels and fatty acid oxidation in the brain. GLP-1 also exhibits similar functions and growth factor-like properties to IGF-1, which is likely how it exerts its neuroprotective effects. Recent preclinical and clinical evidence indicate that IGF-1 and GLP-1, apart from regulating growth and development, prevent neuronal death mediated by amyloidogenesis, cerebral glucose deprivation, neuroinflammation and apoptosis through modulation of PI3/Akt kinase, mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase (MAPK/ERK). IGF-1 resistance and GLP-1 deficiency impair protective cellular signaling mechanisms, contributing to the progression of neurodegenerative diseases. Over the past decades, IGF-1 and GLP-1 have emerged as an essential component of the neuronal system and as potential therapeutic targets for several neurodegenerative and neuropsychiatric dysfunctions. There is substantial evidence that IGF-1 and GLP-1 analogues penetrate the blood-brain barrier (BBB) and exhibit neuroprotective functions, including synaptic formation, neuronal plasticity, protein synthesis, and autophagy. Conclusively, this review represents the therapeutic potential of IGF-1 and GLP-1 signaling target activators in ameliorating neurological disorders.
Collapse
|
12
|
Gonzalez A, Berg MD, Southey B, Dean M. Effect of estradiol and IGF1 on glycogen synthesis in bovine uterine epithelial cells. Reproduction 2022; 164:97-108. [PMID: 35900330 DOI: 10.1530/rep-22-0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 07/14/2022] [Indexed: 11/08/2022]
Abstract
In brief Glucose is an important nutrient for the endometrium and embryo during pregnancy. This study shows that estradiol (E2)/IGF1 signaling stimulates glycogen synthesis in the uterine epithelium of cows, which could provide glucose when needed. Abstract Glycogen storage in the uterine epithelium peaks near estrus and is a potential source of glucose for the endometrium and embryos. However, the hormonal regulation of glycogen synthesis in the uterine epithelium is poorly understood. Our objective was to evaluate the effect of E2 and insulin-like growth factor 1 (IGF1) on glycogenesis in immortalized bovine uterine epithelial (BUTE) cells. Treatment of BUTE cells with E2 (0.1-10 nM) did not increase glycogen levels. However, treatment of BUTE cells with IGF1 (50 or 100 ng/mL) resulted in a >2-fold increase in glycogen. To determine if the uterine stroma produced IGF1 in response to E2, bovine uterine fibroblasts were treated with E2, which increased IGF1 levels. Immunohistochemistry showed higher levels of IGF1 in the stroma on day 1 than on day 11, which coincides with higher glycogen levels in the uterine epithelium. Western blots revealed that IGF1 treatment increased the levels of phospho-AKT, phospho-GSKβ, hexokinase 1, and glycogen synthase in BUTE cells. Metabolomic (GC-MS) analysis showed that IGF1 increased 3-phosphoglycerate and lactate, potentially indicative of increased flux through glycolysis. We also found higher levels of N-acetyl-glucosamine and protein glycosylation after IGF1 treatment, indicating increased hexosamine biosynthetic pathway activity. In conclusion, IGF1 is produced by uterine fibroblasts due to E2, and IGF1 increases glucose metabolism and glycogenesis in uterine epithelial cells. Glycogen stored in the uterine epithelium due to E2/IGF1 signaling at estrus could provide glucose to the endometrium or be secreted into the uterine lumen as a component of histotroph.
Collapse
Affiliation(s)
- Alexis Gonzalez
- 1Department of Animal Science, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Malia D Berg
- 1Department of Animal Science, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Bruce Southey
- 1Department of Animal Science, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Matthew Dean
- 1Department of Animal Science, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
13
|
Lippert RN, Brüning JC. Maternal Metabolic Programming of the Developing Central Nervous System: Unified Pathways to Metabolic and Psychiatric Disorders. Biol Psychiatry 2022; 91:898-906. [PMID: 34330407 DOI: 10.1016/j.biopsych.2021.06.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/17/2022]
Abstract
The perinatal period presents a critical time in offspring development where environmental insults can have damaging impacts on the future health of the offspring. This can lead to sustained alterations in offspring development, metabolism, and predisposition to both metabolic and psychiatric diseases. The central nervous system is one of the most sensitive targets in response to maternal obesity and/or type 2 diabetes mellitus. While many of the effects of obesity on brain function in adults are known, we are only now beginning to understand the multitude of changes that occur in the brain during development on exposure to maternal overnutrition. Specifically, given recent links between maternal metabolic state and onset of neurodevelopmental diseases, the specific changes that are occurring in the offspring are even more relevant for the study of disease onset. It is therefore critical to understand the developmental effects of maternal obesity and/or type 2 diabetes mellitus and further to define the underlying cellular and molecular changes in the fetal brain. This review focuses on the current advancements in the study of maternal programming of brain development with particular emphasis on brain connectivity, specific regional effects, newly studied peripheral contributors, and key windows of interventions where maternal bodyweight and food intake may drive the most detrimental effects on the brain and associated metabolic and behavioral consequences.
Collapse
Affiliation(s)
- Rachel N Lippert
- German Institute of Human Nutrition Potsdam Rehbrücke, Potsdam, Germany; German Center for Diabetes Research, Neuherberg, Germany; Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Jens C Brüning
- German Center for Diabetes Research, Neuherberg, Germany; Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes and Preventive Medicine, University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
14
|
Holgersen K, Rasmussen MB, Carey G, Burrin DG, Thymann T, Sangild PT. Clinical outcome and gut development after insulin-like growth factor-1 supplementation to preterm pigs. Front Pediatr 2022; 10:868911. [PMID: 35989990 PMCID: PMC9389362 DOI: 10.3389/fped.2022.868911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 07/08/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Elevation of circulating insulin-like growth factor-1 (IGF-1) within normal physiological levels may alleviate several morbidities in preterm infants but safety and efficacy remain unclear. We hypothesized that IGF-1 supplementation during the first 1-2 weeks after preterm birth improves clinical outcomes and gut development, using preterm pigs as a model for infants. METHODS Preterm pigs were given vehicle or recombinant human IGF-1/binding protein-3 (rhIGF-1, 2.25 mg/kg/d) by subcutaneous injections for 8 days (Experiment 1, n = 34), or by systemic infusion for 4 days (Experiment 2, n = 19), before collection of blood and organs for analyses. RESULTS In both experiments, rhIGF-1 treatment increased plasma IGF-1 levels 3-4 fold, reaching the values reported for term suckling piglets. In Experiment 1, rhIGF-1 treatment increased spleen and intestinal weights without affecting clinical outcomes like growth, blood biochemistry (except increased sodium and gamma-glutamyltransferase levels), hematology (e.g., red and white blood cell populations), glucose homeostasis (e.g., basal and glucose-stimulated insulin and glucose levels) or systemic immunity variables (e.g., T cell subsets, neutrophil phagocytosis, LPS stimulation, bacterial translocation to bone marrow). The rhIGF-1 treatment increased gut protein synthesis (+11%, p < 0.05) and reduced the combined incidence of all-cause mortality and severe necrotizing enterocolitis (NEC, p < 0.05), but had limited effects on intestinal morphology, cell proliferation, cell apoptosis, brush-border enzyme activities, permeability and levels of cytokines (IL-1β, IL-6, IL-8). In Experiment 2, rhIGF-1 treated pigs had reduced blood creatine kinase, creatinine, potassium and aspartate aminotransferase levels, with no effects on organ weights (except increased spleen weight), blood chemistry values, clinical variables or NEC. CONCLUSION Physiological elevation of systemic IGF-1 levels for 8 days after preterm birth increased intestinal weight and protein synthesis, spleen weight and potential overall viability of pigs, without any apparent negative effects on recorded clinical parameters. The results add further preclinical support for safety and efficacy of supplemental IGF-1 to hospitalized very preterm infants.
Collapse
Affiliation(s)
- Kristine Holgersen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Martin Bo Rasmussen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Douglas G Burrin
- Department of Pediatrics, United States Department of Agriculture/Agricultural Research Service, Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
| | - Thomas Thymann
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Per Torp Sangild
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.,Department of Neonatology, Rigshospitalet, Copenhagen, Denmark.,Department of Pediatrics, Odense University Hospital, Odense, Denmark
| |
Collapse
|
15
|
Predovan D, Berryman N, Lussier M, Comte F, Vu TTM, Villalpando JM, Bherer L. Assessment of the Relationship Between Executive Function and Cardiorespiratory Fitness in Healthy Older Adults. Front Psychol 2021; 12:742184. [PMID: 34803824 PMCID: PMC8595132 DOI: 10.3389/fpsyg.2021.742184] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Associations between cardiorespiratory fitness and brain health in healthy older adults have been reported using a variety of cardiorespiratory fitness estimates (CRFe). Using commonly used methods to determine CRF, we assessed the relationship between CRFe and executive function performance. Healthy older adults (n = 60, mean age 68 years, 77% women), underwent three CRF tests: a Maximal Graded Exercise Test performed on a cycle ergometer, the Rockport Fitness Walking Test, and a Non-Exercise Prediction Equation. Executive function was assessed by a computerized cognitive assessment using an N-Back task (updating cost) and a Stroop task (interference cost, global and local switch cost). Multiple hierarchical regression analyses were conducted to assess the relationship between different CRFe and executive function performance. Regardless of age and education, cardiorespiratory fitness estimated from the Maximal Graded Exercise Test and the Rockport Fitness Walking Test was significantly associated with the global switch cost. All CRFe were associated with the interference cost. No association was observed between CRFe and local switching costs or the updating costs. In the present study, not all subcomponents of executive function were related to CRFe. Interestingly, the executive functions that were associated with CRFe are those that are known to be the most affected by aging.
Collapse
Affiliation(s)
- David Predovan
- Centre de Recherche, Institut Universitaire de Gériatrie de Montréal, Montréal, QC, Canada.,Département de Psychologie, Université du Québec à Montréal, Montréal, QC, Canada.,Centre de Recherche, Institut de Cardiologie de Montréal, Montréal, QC, Canada
| | - Nicolas Berryman
- Centre de Recherche, Institut Universitaire de Gériatrie de Montréal, Montréal, QC, Canada.,Département des Sciences de l'Activité Physique, Université du Québec à Montréal, Montréal, QC, Canada
| | - Maxime Lussier
- Centre de Recherche, Institut Universitaire de Gériatrie de Montréal, Montréal, QC, Canada.,Département de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Francis Comte
- Centre de Recherche, Institut Universitaire de Gériatrie de Montréal, Montréal, QC, Canada
| | - Thien Tuong Minh Vu
- Centre de Recherche, Institut Universitaire de Gériatrie de Montréal, Montréal, QC, Canada.,Département de Médecine, Centre Hospitalier de l'Université de Montréal, Service de Gériatrie, Montréal, QC, Canada
| | | | - Louis Bherer
- Centre de Recherche, Institut Universitaire de Gériatrie de Montréal, Montréal, QC, Canada.,Centre de Recherche, Institut de Cardiologie de Montréal, Montréal, QC, Canada.,Département de Médecine, Université de Montréal, Montréal, QC, Canada.,PERFORM Centre, Concordia University, Montréal, QC, Canada
| |
Collapse
|
16
|
Gong P, Zou Y, Zhang W, Tian Q, Han S, Xu Z, Chen Q, Wang X, Li M. The neuroprotective effects of Insulin-Like Growth Factor 1 via the Hippo/YAP signaling pathway are mediated by the PI3K/AKT cascade following cerebral ischemia/reperfusion injury. Brain Res Bull 2021; 177:373-387. [PMID: 34717965 DOI: 10.1016/j.brainresbull.2021.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 10/07/2021] [Accepted: 10/26/2021] [Indexed: 10/20/2022]
Abstract
Insulin-like growth factor 1 (IGF-1) has neuroprotective actions, including vasodilatory, anti-inflammatory, and antithrombotic effects, following ischemic stroke. However, the molecular mechanisms underlying the neuroprotective effects of IGF-1 following ischemic stroke remain unknown. Therefore, in the present study, we investigated whether IGF-1 exerted its neuroprotective effects by regulating the Hippo/YAP signaling pathway, potentially via activation of the PI3K/AKT cascade, following ischemic stroke. In the in vitro study, we exposed cultured PC12 and SH-5YSY cells, and cortical primary neurons, to oxygen-glucose deprivation. Cell viability was measured using CCK-8 assay. In the in vivo study, Sprague-Dawley rats were subjected to middle cerebral artery occlusion. Neurological function was assessed using a modified neurologic scoring system and the modified neurological severity score (mNSS) test, brain edema was detected by brain water content measurement, infarct volume was measured using triphenyltetrazolium chloride staining, and neuronal death and apoptosis were evaluated by TUNEL/NeuN double staining, HE and Nissl staining, and immunohistochemistry staining for NeuN. Finally, western blot analysis was used to measure the level of IGF-1 in vivo and levels of YAP/TAZ, PI3K and phosphorylated AKT (p-AKT) both in vitro and in vivo. IGF-1 induced activation of YAP/TAZ, which resulted in improved cell viability in vitro, and reduced neurological deficits, brain water content, neuronal death and apoptosis, and cerebral infarct volume in vivo. Notably, the neuroprotective effects of IGF-1 were blocked by an inhibitor of the PI3K/AKT cascade, LY294002. LY294002 treatment not only downregulated PI3K and p-AKT, but YAP/TAZ as well, leading to aggravation of neurological dysfunction and worsening of brain damage. Our findings indicate that the neuroprotective effects of IGF-1 are, at least in part mediated by upregulation of YAP/TAZ via activation of the PI3K/AKT cascade following cerebral ischemic stroke.
Collapse
Affiliation(s)
- Pian Gong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Yichun Zou
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, PR China
| | - Wei Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Shoumeng Han
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Zhou Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China.
| |
Collapse
|
17
|
Dini S, Zakeri M, Ebrahimpour S, Dehghanian F, Esmaeili A. Quercetin‑conjugated superparamagnetic iron oxide nanoparticles modulate glucose metabolism-related genes and miR-29 family in the hippocampus of diabetic rats. Sci Rep 2021; 11:8618. [PMID: 33883592 PMCID: PMC8060416 DOI: 10.1038/s41598-021-87687-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 03/25/2021] [Indexed: 02/02/2023] Open
Abstract
Quercetin (QC) is a dietary bioflavonoid that can be conjugated with nanoparticles to facilitate its brain bioavailability. We previously showed that quercetin-conjugated superparamagnetic iron oxide nanoparticles (QCSPIONs) reduced the level of blood glucose in diabetic rats. Glucose transporters (GLUTs), insulin-like growth factor-1 (IGF-1), and microRNA-29 (miR-29) play a critical role in brain glucose homeostasis. In the current study, we examined the effects of QCSPION on the expression of glucose metabolism-related genes, and the miR-29 family as a candidate regulator of glucose handling in the hippocampus of diabetic rats. Our in silico analyses introduce the miR-29 family as potential regulators of glucose transporters and IGF-1 genes. The expression level of the miR-29 family, IGF-1, GLUT1, GLUT2, GLUT3, and GLUT4 were measured by qPCR. Our results indicate that diabetes significantly results in upregulation of the miR-29 family and downregulation of the GLUT1, 2, 3, 4, and IGF-1 genes. Interestingly, QCSPIONs reduced miR-29 family expression and subsequently enhanced GLUT1, 2, 3, 4, and IGF-1expression. In conclusion, our findings suggest that QCSPION could regulate the expression of the miR-29 family, which in turn increases the expression of glucose transporters and IGF-1, thereby reducing diabetic complications.
Collapse
Affiliation(s)
- Solmaz Dini
- grid.411750.60000 0001 0454 365XDepartment of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mansoureh Zakeri
- grid.411750.60000 0001 0454 365XDepartment of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Shiva Ebrahimpour
- grid.411750.60000 0001 0454 365XDepartment of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Fariba Dehghanian
- grid.411750.60000 0001 0454 365XDepartment of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Abolghasem Esmaeili
- grid.411750.60000 0001 0454 365XDepartment of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
18
|
IGF1R Deficiency Modulates Brain Signaling Pathways and Disturbs Mitochondria and Redox Homeostasis. Biomedicines 2021; 9:biomedicines9020158. [PMID: 33562061 PMCID: PMC7915200 DOI: 10.3390/biomedicines9020158] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/31/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
Insulin-like growth factor 1 receptor (IGF1R)-mediated signaling pathways modulate important neurophysiological aspects in the central nervous system, including neurogenesis, synaptic plasticity and complex cognitive functions. In the present study, we intended to characterize the impact of IGF1R deficiency in the brain, focusing on PI3K/Akt and MAPK/ERK1/2 signaling pathways and mitochondria-related parameters. For this purpose, we used 13-week-old UBC-CreERT2; Igf1rfl/fl male mice in which Igf1r was conditionally deleted. IGF1R deficiency caused a decrease in brain weight as well as the activation of the IR/PI3K/Akt and inhibition of the MAPK/ERK1/2/CREB signaling pathways. Despite no alterations in the activity of caspases 3 and 9, a significant alteration in phosphorylated GSK3β and an increase in phosphorylated Tau protein levels were observed. In addition, significant disturbances in mitochondrial dynamics and content and altered activity of the mitochondrial respiratory chain complexes were noticed. An increase in oxidative stress, characterized by decreased nuclear factor E2-related factor 2 (NRF2) protein levels and aconitase activity and increased H2O2 levels were also found in the brain of IGF1R-deficient mice. Overall, our observations confirm the complexity of IGF1R in mediating brain signaling responses and suggest that its deficiency negatively impacts brain cells homeostasis and survival by affecting mitochondria and redox homeostasis.
Collapse
|
19
|
Berberine alleviates rotenone-induced cytotoxicity by antioxidation and activation of PI3K/Akt signaling pathway in SH-SY5Y cells. Neuroreport 2021; 31:41-47. [PMID: 31688419 DOI: 10.1097/wnr.0000000000001365] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Berberine, an isoquinoline alkaloid isolated from traditional Chinese medicine, has been widely studied for its efficacy in the treatment of neurodegenerative diseases. However, berberine-mediated neuroprotection in the pathogenesis of Parkinson's disease is still uncertain. In this study, the effects of berberine on rotenone-induced neurotoxicity in SH-SY5Y cells were investigated. The results showed that berberine treatment significantly alleviated rotenone-induced decrease in the cell viability in SH-SY5Y cells. Further studies demonstrated that berberine suppressed the production of intracellular reactive oxygen species, restored the mitochondrial transmembrane potential, increased Bcl-2/Bax ratio, and decreased caspase-3 activation that induced by rotenone. Furthermore, berberine also restored the phosphorylation of Akt, which was downregulated by rotenone in SH-SY5Y cells. These results suggest that berberine protects rotenone-treated SH-SY5Y cells by antioxidation and activation of PI3K/Akt signaling pathway.
Collapse
|
20
|
Guerra-Cantera S, Frago LM, Collado-Pérez R, Canelles S, Ros P, Freire-Regatillo A, Jiménez-Hernaiz M, Barrios V, Argente J, Chowen JA. Sex Differences in Metabolic Recuperation After Weight Loss in High Fat Diet-Induced Obese Mice. Front Endocrinol (Lausanne) 2021; 12:796661. [PMID: 34975768 PMCID: PMC8716724 DOI: 10.3389/fendo.2021.796661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/16/2021] [Indexed: 01/10/2023] Open
Abstract
Dietary intervention is a common tactic employed to curtail the current obesity epidemic. Changes in nutritional status alter metabolic hormones such as insulin or leptin, as well as the insulin-like growth factor (IGF) system, but little is known about restoration of these parameters after weight loss in obese subjects and if this differs between the sexes, especially regarding the IGF system. Here male and female mice received a high fat diet (HFD) or chow for 8 weeks, then half of the HFD mice were changed to chow (HFDCH) for 4 weeks. Both sexes gained weight (p < 0.001) and increased their energy intake (p < 0.001) and basal glycemia (p < 0.5) on the HFD, with these parameters normalizing after switching to chow but at different rates in males and females. In both sexes HFD decreased hypothalamic NPY and AgRP (p < 0.001) and increased POMC (p < 0.001) mRNA levels, with all normalizing in HFDCH mice, whereas the HFD-induced decrease in ObR did not normalize (p < 0.05). All HFD mice had abnormal glucose tolerance tests (p < 0.001), with males clearly more affected, that normalized when returned to chow. HFD increased insulin levels and HOMA index (p < 0.01) in both sexes, but only HFDCH males normalized this parameter. Returning to chow normalized the HFD-induced increase in circulating leptin (p < 0.001), total IGF1 (p < 0.001), IGF2 (p < 0.001, only in females) and IGFBP3 (p < 0.001), whereas free IGF1 levels remained elevated (p < 0.01). In males IGFBP2 decreased with HFD and normalized with chow (p < 0.001), with no changes in females. Although returning to a healthy diet improved of most metabolic parameters analyzed, fIGF1 levels remained elevated and hypothalamic ObR decreased in both sexes. Moreover, there was sex differences in both the response to HFD and the switch to chow including circulating levels of IGF2 and IGFBP2, factors previously reported to be involved in glucose metabolism. Indeed, glucose metabolism was also differentially modified in males and females, suggesting that these observations could be related.
Collapse
Affiliation(s)
- Santiago Guerra-Cantera
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura M. Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto Collado-Pérez
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sandra Canelles
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Purificación Ros
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Pediatrics, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Alejandra Freire-Regatillo
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - María Jiménez-Hernaiz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
- *Correspondence: Julie A. Chowen, ; Jesús Argente,
| | - Julie A. Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
- *Correspondence: Julie A. Chowen, ; Jesús Argente,
| |
Collapse
|
21
|
Peng W, Liu X, Tan C, Zhou W, Jiang J, Zhou X, Du J, Mo L, Chen L. Zinc-α2-glycoprotein relieved seizure-Induced neuronal glucose uptake impairment via insulin-like growth factor 1 receptor-regulated glucose transporter 3 expression. J Neurochem 2020; 157:695-709. [PMID: 33258143 DOI: 10.1111/jnc.15254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/30/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022]
Abstract
Glucose hypometabolism is observed in epilepsy and promotes epileptogenesis. Glucose hypometabolism in epilepsy may be attributed to decreased neuronal glucose uptake, but its molecular mechanism remains unclear. Zinc-α2-glycoprotein (ZAG) is related to glucose metabolism and is reported to suppress seizures. The anti-epileptic effect of ZAG may be attributed to its regulation of neuronal glucose metabolism. This study explored the effect of ZAG on neuronal glucose uptake and its molecular mechanism via insulin-like growth factor 1 receptor (IGF1R)-regulated glucose transporter 3 (GLUT-3) expression. The ZAG level was modulated by lentivirus in primary culture neurons. Neuronal seizure models were induced by Mg2+ -free artificial cerebrospinal fluid. We assessed neuronal glucose uptake by the 2-NBDG method and Glucose Uptake Colorimetric Assay Kit. IGF1R was activated by IGF1 and blocked by AXL1717. The expression and distribution of IGF1R and GLUT-3, together with IGF1R phosphorylation, were measured by western blot. The binding between ZAG and IGF1R was determined by coimmunoprecipitation. Neuronal glucose uptake and GLUT-3 expression were significantly decreased by seizure or ZAG knockdown, whereas ZAG over-expression or IGF1 treatment reversed this decrease. The effect of ZAG on neuronal glucose uptake and GLUT-3 expression was blocked by AXL1717. ZAG increased IGF1R distribution and phosphorylation possibly by binding. Additionally, IGF1R increased GLUT-3 activity by increasing GLUT-3 expression. In epilepsy/seizure, neuronal glucose uptake suppression may be attributed to a decrease in ZAG, which suppresses neuronal GLUT-3 expression by regulating the activity of IGF1R. ZAG, IGF1R, and GLUT-3 may be novel potential therapeutic targets of glucose hypometabolism in epilepsy and seizures.
Collapse
Affiliation(s)
- Wuxue Peng
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xi Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Changhong Tan
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wen Zhou
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Jiang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuan Zhou
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Juncong Du
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lijuan Mo
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lifen Chen
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
Murugan M, Boison D. Ketogenic diet, neuroprotection, and antiepileptogenesis. Epilepsy Res 2020; 167:106444. [PMID: 32854046 PMCID: PMC7655615 DOI: 10.1016/j.eplepsyres.2020.106444] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/30/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022]
Abstract
High fat, low carbohydrate ketogenic diets (KD) have been in use for the treatment of epilepsy for almost a hundred years. Remarkably, seizures that are resistant to conventional anti-seizure drugs can in many cases be controlled by the KD therapy, and it has been shown that many patients with epilepsy become seizure free even after discontinuation of the diet. These findings suggest that KD combine anti-seizure effects with disease modifying effects. In addition to the treatment of epilepsy, KDs are now widely used for the treatment of a wide range of conditions including weight reduction, diabetes, and cancer. The reason for the success of metabolic therapies is based on the synergism of at least a dozen different mechanisms through which KDs provide beneficial activities. Among the newest findings are epigenetic mechanisms (DNA methylation and histone acetylation) through which KD exerts long-lasting disease modifying effects. Here we review mechanisms through which KD can affect neuroprotection in the brain, and how a combination of those mechanisms with epigenetic alterations can attenuate and possibly reverse the development of epilepsy.
Collapse
Affiliation(s)
- Madhuvika Murugan
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, United States
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, United States; Department of Neurosurgery, New Jersey Medical School, Rutgers University, Newark, NJ 07102, United States; Rutgers Neurosurgery H.O.P.E. Center, Department of Neurosurgery, Rutgers University, New Brunswick, NJ 08901, United States.
| |
Collapse
|
23
|
Nakhjiri E, Vafaee MS, Hojjati SMM, Shahabi P, Shahpasand K. Tau Pathology Triggered by Spinal Cord Injury Can Play a Critical Role in the Neurotrauma Development. Mol Neurobiol 2020; 57:4845-4855. [PMID: 32808121 DOI: 10.1007/s12035-020-02061-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
Traumatic spinal cord injury (SCI) can result in substantial neurological impairment along with significant emotional and psychological distress. It is clear that there is profound neurodegeneration upon SCI, gradually spread to other spinal cord regions and brain areas. Despite extensive considerations, it remains uncertain how pathogenicity diffuses in the cord. It has been reported that tau protein abnormal hyperphosphorylation plays a central role in neurodegeneration triggered by traumatic brain injury (TBI). Tau is a microtubule-associated protein, heavily implicated in neurodegenerative diseases. Importantly, tau pathology spreads in a traumatic brain in a timely manner. In particular, we have recently demonstrated that phosphorylated tau at Thr231 exists in two distinct cis and trans conformations, in which that cis P-tau is extremely neurotoxic, has a prion nature, and spreads to various brain areas and cerebrospinal fluid (CSF) upon trauma. On the other hand, tau pathology, in particular hyperphosphorylation at Thr231, has been observed upon SCI. Taken these together, we conclude that cis pT231-tau may accumulate and spread in the spinal cord as well as CSF and diffuse tau pathology in the central nervous system (CNS). Moreover, antibody against cis P-tau can target intracellular cis P-tau and protect pathology spreading. Thus, considering cis P-tau as a driver of tau pathology and neurodegeneration upon SCI would open new windows toward understanding the disease development and early biomarkers. Furthermore, it would help us develop effective therapies for SCI patients.
Collapse
Affiliation(s)
- Elnaz Nakhjiri
- Neurosciences Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manuchehr S Vafaee
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | | | - Parviz Shahabi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
24
|
Pennuto M, Pandey UB, Polanco MJ. Insulin-like growth factor 1 signaling in motor neuron and polyglutamine diseases: From molecular pathogenesis to therapeutic perspectives. Front Neuroendocrinol 2020; 57:100821. [PMID: 32006533 DOI: 10.1016/j.yfrne.2020.100821] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 01/24/2020] [Accepted: 01/24/2020] [Indexed: 11/19/2022]
Abstract
The pleiotropic peptide insulin-like growth factor 1 (IGF-I) regulates human body homeostasis and cell growth. IGF-I activates two major signaling pathways, namely phosphoinositide-3-kinase (PI3K)/protein kinase B (PKB/Akt) and Ras/extracellular signal-regulated kinase (ERK), which contribute to brain development, metabolism and function as well as to neuronal maintenance and survival. In this review, we discuss the general and tissue-specific effects of the IGF-I pathways. In addition, we present a comprehensive overview examining the role of IGF-I in neurodegenerative diseases, such as spinal and muscular atrophy, amyotrophic lateral sclerosis, and polyglutamine diseases. In each disease, we analyze the disturbances of the IGF-I pathway, the modification of the disease protein by IGF-I signaling, and the therapeutic strategies based on the use of IGF-I developed to date. Lastly, we highlight present and future considerations in the use of IGF-I for the treatment of these disorders.
Collapse
Affiliation(s)
- Maria Pennuto
- Department of Biomedical Sciences (DBS), University of Padova, 35131 Padova, Italy; Veneto Institute of Molecular Medicine (VIMM), Via Orus 2, 35129 Padova, Italy; Padova Neuroscience Center (PNC), 35131 Padova, Italy; Myology Center (CIR-Myo), 35131 Padova, Italy.
| | - Udai Bhan Pandey
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261, USA; Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15224, USA; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - María José Polanco
- Department of Pharmaceutic and Health Science, University San Pablo CEU, Campus Montepríncipe, 28925 Alcorcón, Madrid, Spain.
| |
Collapse
|
25
|
Guerra-Cantera S, Frago LM, Díaz F, Ros P, Jiménez-Hernaiz M, Freire-Regatillo A, Barrios V, Argente J, Chowen JA. Short-Term Diet Induced Changes in the Central and Circulating IGF Systems Are Sex Specific. Front Endocrinol (Lausanne) 2020; 11:513. [PMID: 32849298 PMCID: PMC7431666 DOI: 10.3389/fendo.2020.00513] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
Insulin-like growth factor (IGF) 1 exerts a wide range of functions in mammalians participating not only in the control of growth and metabolism, but also in other actions such as neuroprotection. Nutritional status modifies the IGF system, although little is known regarding how diet affects the newest members of this system including pregnancy-associated plasma protein-A (PAPP-A) and PAPP-A2, proteases that liberate IGF from the IGF-binding proteins (IGFBPs), and stanniocalcins (STCs) that inhibit PAPP-A and PAPP-A2 activity. Here we explored if a 1-week dietary change to either a high-fat diet (HFD) or a low-fat diet (LFD) modifies the central and peripheral IGF systems in both male and female Wistar rats. The circulating IGF system showed sex differences in most of its members at baseline. Males had higher levels of both free (p < 0.001) and total IGF1 (p < 0.001), as well as IGFBP3 (p < 0.001), IGFBP5 (p < 0.001), and insulin (p < 0.01). In contrast, females had higher serum levels of PAPP-A2 (p < 0.05) and IGFBP2 (p < 0.001). The responses to a short-term dietary change were both diet and sex specific. Circulating levels of IGF2 increased in response to LFD intake in females (p < 0.001) and decreased in response to HFD intake in males (p < 0.001). In females, LFD intake also decreased circulating IGFBP2 levels (p < 0.001). In the hypothalamus LFD intake increased IGF2 (p < 0.01) and IGFBP2 mRNA (p < 0.001) levels, as well as the expression of NPY (p < 0.001) and AgRP (p < 0.01), but only in males. In conclusion, short-term LFD intake induced more changes in the peripheral and central IGF system than did short-term HFD intake. Moreover, these changes were sex-specific, with IGF2 and IGFBP2 being more highly affected than the other members of the IGF system. One of the main differences between the commercial LFD employed and the HFD or normal rodent chow is that the LFD has a significantly higher sucrose content, suggesting that this nutrient could be involved in the observed responses.
Collapse
Affiliation(s)
- Santiago Guerra-Cantera
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura M. Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisca Díaz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Purificacion Ros
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Maria Jiménez-Hernaiz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandra Freire-Regatillo
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
- *Correspondence: Jesús Argente
| | - Julie A. Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
- Julie A. Chowen
| |
Collapse
|
26
|
Carmichael O, Stuchlik P, Pillai S, Biessels GJ, Dhullipudi R, Madden-Rusnak A, Martin S, Hsia DS, Fonseca V, Bazzano L. High-Normal Adolescent Fasting Plasma Glucose Is Associated With Poorer Midlife Brain Health: Bogalusa Heart Study. J Clin Endocrinol Metab 2019; 104:4492-4500. [PMID: 31058974 PMCID: PMC6736207 DOI: 10.1210/jc.2018-02750] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/30/2019] [Indexed: 12/24/2022]
Abstract
CONTEXT It is unclear how adolescent glycemic status relates to brain health in adulthood. OBJECTIVE To assess the association between adolescent fasting plasma glucose (FPG) and MRI-based brain measures in midlife. DESIGN Between 1973 and 1992, the Bogalusa Heart Study (BHS) collected FPG from children, 3 to 18 years old, and followed up between 1992 and 2018. Cognitive tests and brain MRI were collected in 2013 to 2016 and 2018. SETTING Observational longitudinal cohort study. PARTICIPANTS Of 1298 contacted BHS participants, 74 completed screening, and 50 completed MRI. MAIN OUTCOME MEASURES Mean FPG per participant at ages <20, 20 to 40, and over 40 years old; brain white matter hyperintensity (WMH) volume, gray matter volume, and functional MRI (fMRI) activation to a Stroop task; tests of logical and working memory, executive function, and semantic fluency. RESULTS At MRI, participants were middle aged (51.3 ± 4.4 years) and predominantly female (74%) and white (74%). Mean FPG was impaired for zero, two, and nine participants in pre-20, 20 to 40, and over-40 periods. The pre-20 mean FPG above the pre-20 median value (i.e., above 83.5 mg/dL) was associated with greater WMH volume [mean difference: 0.029% of total cranial volume, CI: (0.0059, 0.052), P = 0.015] and less fMRI activation [-1.41 units (-2.78, -0.05), P = 0.043] on midlife MRI compared with below-median mean FPG. In controlling for over-40 mean FPG status did not substantially modify the associations. Cognitive scores did not differ by pre-20 mean FPG. CONCLUSIONS High-normal adolescent FPG may be associated with preclinical brain changes in midlife.
Collapse
Affiliation(s)
- Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
- Correspondence and Reprint Requests: Owen Carmichael, PhD, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, Louisiana 70808. E-mail:
| | | | | | - Geert-Jan Biessels
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands
| | - Ram Dhullipudi
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | | | - Shane Martin
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Daniel S Hsia
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Vivian Fonseca
- Section of Endocrinology, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Lydia Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| |
Collapse
|
27
|
The Effect of Endurance Training with Crocin Consumption on IGF-1 and Glycogen Expression in Rat Hippocampus Tissue of Trimethyltin-Treated Model of Alzheimer’s Disease. Asian J Sports Med 2019. [DOI: 10.5812/asjsm.92246] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
28
|
Piri R, Naghavi-Behzad M, Gerke O, Høilund-Carlsen PF, Vafaee MS. Investigations of possible links between Alzheimer’s disease and type 2 diabetes mellitus by positron emission tomography: a systematic review. Clin Transl Imaging 2019. [DOI: 10.1007/s40336-019-00339-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
29
|
Jin J, Ravindran P, Di Meo D, Püschel AW. Igf1R/InsR function is required for axon extension and corpus callosum formation. PLoS One 2019; 14:e0219362. [PMID: 31318893 PMCID: PMC6638864 DOI: 10.1371/journal.pone.0219362] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 06/21/2019] [Indexed: 12/02/2022] Open
Abstract
One of the earliest steps during the development of the nervous system is the establishment of neuronal polarity and the formation of an axon. The intrinsic mechanisms that promote axon formation have been extensively analyzed. However, much less is known about the extrinsic signals that initiate axon formation. One of the candidates for these signals is Insulin-like growth factor 1 (Igf1) that acts through the Igf1 (Igf1R) and insulin receptors (InsR). Since Igf1R and InsR may act redundantly we analyzed conditional cortex-specific knockout mice that are deficient for both Igf1r and Insr to determine if they regulate the development of the cortex and the formation of axons in vivo. Our results show that Igf1R/InsR function is required for the normal development of the embryonic hippocampus and cingulate cortex while the lateral cortex does not show apparent defects in the Igf1r;Insr knockout. In the cingulate cortex, the number of intermediate progenitors and deep layer neurons is reduced and the corpus callosum is absent at E17. However, cortical organization and axon formation are not impaired in knockout embryos. In culture, cortical and hippocampal neurons from Igf1r;Insr knockout embryos extend an axon but the length of this axon is severely reduced. Our results indicate that Igf1R/InsR function is required for brain development in a region-specific manner and promotes axon growth but is not essential for neuronal polarization and migration in the developing brain.
Collapse
Affiliation(s)
- Jing Jin
- Institut für Molekulare Zellbiologie, University of Münster, Münster, Germany
- Cells-in-Motion Cluster of Excellence, University of Münster, Münster, Germany
| | | | - Danila Di Meo
- Institut für Molekulare Zellbiologie, University of Münster, Münster, Germany
- Cells-in-Motion Cluster of Excellence, University of Münster, Münster, Germany
| | - Andreas W. Püschel
- Institut für Molekulare Zellbiologie, University of Münster, Münster, Germany
- Cells-in-Motion Cluster of Excellence, University of Münster, Münster, Germany
- * E-mail:
| |
Collapse
|
30
|
Lewitt MS, Boyd GW. The Role of Insulin-Like Growth Factors and Insulin-Like Growth Factor-Binding Proteins in the Nervous System. BIOCHEMISTRY INSIGHTS 2019; 12:1178626419842176. [PMID: 31024217 PMCID: PMC6472167 DOI: 10.1177/1178626419842176] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 01/23/2023]
Abstract
The insulin-like growth factors (IGF-I and IGF-II) and their receptors are widely expressed in nervous tissue from early embryonic life. They also cross the blood brain barriers by active transport, and their regulation as endocrine factors therefore differs from other tissues. In brain, IGFs have paracrine and autocrine actions that are modulated by IGF-binding proteins and interact with other growth factor signalling pathways. The IGF system has roles in nervous system development and maintenance. There is substantial evidence for a specific role for this system in some neurodegenerative diseases, and neuroprotective actions make this system an attractive target for new therapeutic approaches. In developing new therapies, interaction with IGF-binding proteins and other growth factor signalling pathways should be considered. This evidence is reviewed, gaps in knowledge are highlighted, and recommendations are made for future research.
Collapse
Affiliation(s)
- Moira S Lewitt
- School of Health & Life Sciences, University of the West of Scotland, Paisley, UK
| | - Gary W Boyd
- School of Health & Life Sciences, University of the West of Scotland, Paisley, UK
| |
Collapse
|
31
|
Specific activation of mGlu2 induced IGF-1R transactivation in vitro through FAK phosphorylation. Acta Pharmacol Sin 2019; 40:460-467. [PMID: 29946167 DOI: 10.1038/s41401-018-0033-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/20/2018] [Indexed: 01/17/2023]
Abstract
Metabotropic glutamate receptor 2 (mGlu2) belongs to the group-II metabotropic glutamate (mGlu) receptors and is a neurotransmitter G protein-coupled receptor. The group-II mGlu receptors are promising antipsychotic targets, but the specific role of mGlu2 signaling remains unclear. Receptor tyrosine kinases (RTKs) are also believed to participate in brain pathogenesis. To investigate whether there is any communication between mGlu2 and RTKs, we generated a CHO-mGlu2 cell line that stably expresses mGlu2 and showed that activation of mGlu2 by LY379268, a group II mGlu agonist, was able to transactivate insulin-like growth factor 1 receptor (IGF-1R). We further determined that the Gi/o protein, Gβγ subunits, phospholipase C, and focal adhesion kinase (FAK) were involved in the IGF-1R transactivation signaling axis, which further induced the phosphorylation of extracellular signal-regulated kinase1/2 (ERK1/2) and cAMP response element-binding protein. In primary mouse cortical neurons, similar signaling pathways were observed when mGlu2 were stimulated by LY487379, an mGlu2 positive allosteric modulator. Transactivation of IGF-1R through FAK in response to mGlu2 should provide a better understanding of the association of mGlu2 with brain disease.
Collapse
|
32
|
Shang Y, Zhi D, Feng G, Wang Z, Mao D, Guo S, Liu R, Liu L, Zhang S, Sun S, Wang K, Kong D, Gao J, Yang Z. Supramolecular Nanofibers with Superior Bioactivity to Insulin-Like Growth Factor-I. NANO LETTERS 2019; 19:1560-1569. [PMID: 30789273 DOI: 10.1021/acs.nanolett.8b04406] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Bioactive peptides derived from proteins generally need to be folded into secondary structures to activate downstream signaling pathways. However, synthetic peptides typically form random-coils, thus losing their bioactivities. Here, we show that by introducing a self-assembling peptide motif and using different preparation pathways, a peptide from insulin-like growth factor-I (IGF-1) can be folded into an α-helix and β-sheet. The β-sheet one exhibits a low dissociation constant to the IGF-1 receptor (IGF-1R, 11.5 nM), which is only about 3 times higher than that of IGF-1 (4.3 nM). However, the α-helical one and the peptide without self-assembling motif show weak affinities to IGF-1R ( KD = 179.1 and 321.6 nM, respectively). At 10 nM, the β-sheet one efficiently activates the IGF-1 downstream pathway, significantly enhancing HUVEC proliferation and preventing cell apoptosis. The β-sheet peptide shows superior performance to IGF-1 in vivo, and it improves ischemic hind-limb salvage by significantly reducing muscle degradation and enhancing limb vascularization. Our study provides a useful strategy to constrain peptides into different conformations, which may lead to the development of supramolecular nanomaterials mimicking biofunctional proteins.
Collapse
Affiliation(s)
- Yuna Shang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials , Nankai University , Tianjin 300071 , P. R. China
| | - Dengke Zhi
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials , Nankai University , Tianjin 300071 , P. R. China
| | - Guowei Feng
- Department of Genitourinary Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy , Tianjin's Clinical Research Center for Cancer , Tianjin 300060 , P. R. China
| | - Zhongyan Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials , Nankai University , Tianjin 300071 , P. R. China
| | - Duo Mao
- Department of Chemical and Biomolecular Engineering , National University of Singapore , Engineering Drive 4 , Singapore , 117585
| | - Shuang Guo
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials , Nankai University , Tianjin 300071 , P. R. China
| | - Ruihua Liu
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials , Nankai University , Tianjin 300071 , P. R. China
| | - Lulu Liu
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials , Nankai University , Tianjin 300071 , P. R. China
| | - Shuhao Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials , Nankai University , Tianjin 300071 , P. R. China
| | - Shenghuan Sun
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials , Nankai University , Tianjin 300071 , P. R. China
| | - Kai Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials , Nankai University , Tianjin 300071 , P. R. China
| | - Deling Kong
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials , Nankai University , Tianjin 300071 , P. R. China
| | - Jie Gao
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials , Nankai University , Tianjin 300071 , P. R. China
| | - Zhimou Yang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials , Nankai University , Tianjin 300071 , P. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute , Xuzhou Medical University , Xuzhou , Jiangsu P. R. China
| |
Collapse
|
33
|
Abstract
Brain development is a highly regulated process that involves the precise spatio-temporal activation of cell signaling cues. Transcription factors play an integral role in this process by relaying information from external signaling cues to the genome. The transcription factor Forkhead box G1 (FOXG1) is expressed in the developing nervous system with a critical role in forebrain development. Altered dosage of FOXG1 due to deletions, duplications, or functional gain- or loss-of-function mutations, leads to a complex array of cellular effects with important consequences for human disease including neurodevelopmental disorders. Here, we review studies in multiple species and cell models where FOXG1 dose is altered. We argue against a linear, symmetrical relationship between FOXG1 dosage states, although FOXG1 levels at the right time and place need to be carefully regulated. Neurodevelopmental disease states caused by mutations in FOXG1 may therefore be regulated through different mechanisms.
Collapse
Affiliation(s)
- Nuwan C Hettige
- Department of Human Genetics, McGill University, Montreal, QC, Canada.,Psychiatric Genetics Group, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Carl Ernst
- Department of Human Genetics, McGill University, Montreal, QC, Canada.,Psychiatric Genetics Group, Douglas Mental Health University Institute, Montreal, QC, Canada.,Department of Psychiatry, McGill University, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| |
Collapse
|
34
|
Abstract
Over 50 years after its first description, Bronchopulmonary Dysplasia (BPD) remains a devastating pulmonary complication in preterm infants with respiratory failure and develops in 30-50% of infants less than 1000-gram birth weight. It is thought to involve ventilator- and oxygen-induced damage to an immature lung that results in an inflammatory response and ends in aberrant lung development with dysregulated angiogenesis and alveolarization. Significant morbidity and mortality are associated with this most common chronic lung disease of childhood. Thus, any therapies that decrease the incidence or severity of this condition would have significant impact on morbidity, mortality, human costs, and healthcare expenditure. It is clear that an inflammatory response and the elaboration of growth factors and cytokines are associated with the development of BPD. Numerous approaches to control the inflammatory process leading to the development of BPD have been attempted. This review will examine the anti-inflammatory approaches that are established or hold promise for the prevention or treatment of BPD.
Collapse
Affiliation(s)
- Rashmin C Savani
- Center for Pulmonary & Vascular Biology, Division of Neonatal-Perinatal Medicine, The Department of Pediatrics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9063, USA.
| |
Collapse
|
35
|
Vassilakos G, Lei H, Yang Y, Puglise J, Matheny M, Durzynska J, Ozery M, Bennett K, Spradlin R, Bonanno H, Park S, Ahima RS, Barton ER. Deletion of muscle IGF-I transiently impairs growth and progressively disrupts glucose homeostasis in male mice. FASEB J 2018; 33:181-194. [PMID: 29932867 DOI: 10.1096/fj.201800459r] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Insulin-like growth factors (IGFs) are essential for local skeletal muscle growth and organismal physiology, but these actions are entwined with glucose homeostasis through convergence with insulin signaling. The objective of this work was to determine whether the effects of IGF-I on growth and metabolism could be separated. We generated muscle-specific IGF-I-deficient (MID) mice that afford inducible deletion of Igf1 at any age. After Igf1 deletion at birth or in young adult mice, evaluations of muscle physiology and glucose homeostasis were performed up to 16 wk of age. MID mice generated at birth had lower muscle and circulating IGF-I, decreased muscle and body mass, and impaired muscle force production. Eight-wk-old male MID had heightened insulin levels with trends of elevated fasting glucose. This phenotype progressed to impaired glucose handling and increased fat deposition without significant muscle mass loss at 16 wk of age. The same phenotype emerged in 16-wk-old MID mice induced at 12 wk of age, compounded with heightened muscle fatigability and exercise intolerance. We assert that muscle IGF-I independently modulates anabolism and metabolism in an age-dependent manner, thus positioning muscle IGF-I maintenance to be critical for both muscle growth and metabolic homeostasis.-Vassilakos, G., Lei, H., Yang, Y., Puglise, J., Matheny, M., Durzynska, J., Ozery, M., Bennett, K., Spradlin, R., Bonanno, H., Park, S., Ahima, R. S., Barton, E. R. Deletion of muscle IGF-I transiently impairs growth and progressively disrupts glucose homeostasis in male mice.
Collapse
Affiliation(s)
- Georgios Vassilakos
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, USA
| | - Hanqin Lei
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, USA
| | - Yun Yang
- Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, Florida, USA.,Gastrointestinal Surgery, West China School of Medicine, Sichuan University-West China Hospital, Chengdu, China
| | - Jason Puglise
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, USA
| | - Michael Matheny
- Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Julia Durzynska
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, USA.,Molecular Virology, Institute of Experimental Biology, A. Mickiewicz University, Poznań, Poland
| | - Matan Ozery
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, USA
| | - Katherine Bennett
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, USA
| | - Ray Spradlin
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, USA
| | - Heather Bonanno
- Animal Care Services, Cancer and Genetics Research Complex, University of Florida, Gainesville, Florida, USA
| | - Soohyun Park
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Rexford S Ahima
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA
| | - Elisabeth R Barton
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, USA.,Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
36
|
Dean M, Rose J. Activation of the IGF1 receptor stimulates glycogen synthesis by mink uterine epithelial cells. Mol Reprod Dev 2018; 85:449-458. [DOI: 10.1002/mrd.22981] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 02/16/2018] [Accepted: 03/19/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Matthew Dean
- Department of Medicinal Chemistry and Pharmacognosy, Center for Biomolecular Sciences, College of Pharmacy; University of Illinois at Chicago; Chicago Illinois
- Department of Biological Sciences, College of Science and Engineering; Idaho State University; Idaho Pocatello
| | - Jack Rose
- Department of Biological Sciences, College of Science and Engineering; Idaho State University; Idaho Pocatello
| |
Collapse
|
37
|
Yan BC, Wang J, Cao J, Won MH. Less hippocampal neuronal death in young gerbils following transient global cerebral ischemia is associated with long‑term maintenance of insulin‑like growth factor 1 and its receptors in the hippocampal CA1 region. Mol Med Rep 2017; 17:3055-3061. [PMID: 29257289 PMCID: PMC5783526 DOI: 10.3892/mmr.2017.8243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/13/2017] [Indexed: 01/03/2023] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a well-known growth factor with well-defined neuroprotective effects against cerebral ischemia. However, the age‑dependent differences in the expression of IGF‑1 and its receptor (IGF‑1R) in the brain following transient cerebral ischemia (TCI) have not been elucidated. In the present study, the differences in IGF‑1 and IGF‑1R in the gerbil hippocampal CA1 region of young and adult gerbils 5 min following TCI were determined. Seven days following TCI, the neuronal death in the hippocampal CA1 region of young gerbils was significantly less than that observed in adult gerbils. In addition, the immunoreactivity, and levels of IGF‑1 and IGF‑1R in the CA1 region of the normal young were higher than those in the normal adult. Four days following TCI, the immunoreactivity, and protein levels of IGF‑1 and IGF‑1R were markedly decreased in the adult group. By contrast, in the young group, the immunoreactivity and expression levels were much greater than those in the adult group. However, 7 days following TCI, all immunoreactivity and expression levels were markedly decreased when compared with those in the normal adult and young groups. In addition, the immunoreactivity and expression levels in the young groups were significantly higher than those of the adult groups. In conclusion, the present study demonstrated that the higher and sustained expression of IGF‑1 and IGF‑1R in the young gerbil hippocampal CA1 region following TCI may be associated with the reduced neuronal death compared to that in the adults.
Collapse
Affiliation(s)
- Bing Chun Yan
- Department of Traditional Chinese and Western Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225001, P.R. China
| | - Jie Wang
- Department of Traditional Chinese and Western Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225001, P.R. China
| | - Jianwen Cao
- Department of Traditional Chinese and Western Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225001, P.R. China
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| |
Collapse
|
38
|
Sural-Fehr T, Bongarzone ER. How membrane dysfunction influences neuronal survival pathways in sphingolipid storage disorders. J Neurosci Res 2017; 94:1042-8. [PMID: 27638590 DOI: 10.1002/jnr.23763] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 04/22/2016] [Accepted: 04/25/2016] [Indexed: 12/21/2022]
Abstract
Sphingolipidoses are a class of inherited diseases that result from the toxic accumulation of undigested sphingolipids in lysosomes and other cellular membranes. Sphingolipids are particularly enriched in cells of the nervous system, and their excessive accumulation during disease has a significant impact on the nervous system. Neuronal dysfunction followed by neurological compromise is a common feature in many of these diseases; however, the underlying mechanisms that cause vulnerability of neurons are not fully understood. The plasma membrane plays a critical role in regulating cellular survival pathways, and its dysfunction has been implicated in neuronal failure in various adult-onset neuropathies. In the context of sphingolipidoses, we hypothesize that gradual accumulation of undigested lipids in plasma membranes causes local disruptions in lipid raft domains, leading to deregulation of multiple signaling pathways important for neuronal survival and function. We propose that defects in downstream signaling as a result of membrane dysfunction are common mechanisms underlying neuronal vulnerability in sphingolipid storage disorders with neurological compromise. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tuba Sural-Fehr
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois.
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
39
|
Glass TJA, Chau V, Gardiner J, Foong J, Vinall J, Zwicker JG, Grunau RE, Synnes A, Poskitt KJ, Miller SP. Severe retinopathy of prematurity predicts delayed white matter maturation and poorer neurodevelopment. Arch Dis Child Fetal Neonatal Ed 2017; 102:F532-F537. [PMID: 28536205 DOI: 10.1136/archdischild-2016-312533] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/04/2017] [Accepted: 04/06/2017] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To determine whether severe retinopathy of prematurity (ROP) is associated with (1) abnormal white matter maturation and (2) neurodevelopmental outcomes at 18 months' corrected age (CA) compared with neonates without severe ROP. DESIGN We conducted a prospective longitudinal cohort of extremely preterm neonates born 24-28 weeks' gestational age recruited between 2006 and 2013 with brain MRIs obtained both early in life and at term-equivalent age. Severe ROP was defined as ROP treated with retinal laser photocoagulation. Using diffusion tensor imaging and tract-based spatial statistics (TBSS), white matter maturation was assessed by mean fractional anisotropy (FA) in seven predefined regions of interest. Neurodevelopmental outcomes were assessed with Bayley Scales of Infant and Toddler Development-III (Bayley-III) composite scores at 18 months' CA. Subjects were compared using Fisher's exact, Kruskal-Wallis and generalised estimating equations. SETTING Families were recruited from the neonatal intensive care unit at BC Women's Hospital. PATIENTS Of 98 extremely preterm neonates (median: 26.0 weeks) assessed locally for ROP, 19 (19%) had severe ROP and 83 (85%) were assessed at 18 months' CA. RESULTS Severe ROP was associated with lower FA in the posterior white matter, and with decreased measures of brain maturation in the optic radiations, posterior limb of the internal capsule (PLIC) and external capsule on TBSS. Bayley-III cognitive and motor scores were lower in infants with severe ROP. CONCLUSIONS Severe ROP is associated with maturational delay in the optic radiations, PLIC, external capsule and posterior white matter, housing the primary visual and motor pathways, and is associated with poorer cognitive and motor outcomes at 18 months' CA.
Collapse
Affiliation(s)
- Torin J A Glass
- Department of Pediatrics (Neurology), University of Toronto and the Hospital for Sick Children, Toronto, Canada.,Neurosciences & Mental Health, SickKids Research Institute, Toronto, Canada
| | - Vann Chau
- Department of Pediatrics (Neurology), University of Toronto and the Hospital for Sick Children, Toronto, Canada.,Neurosciences & Mental Health, SickKids Research Institute, Toronto, Canada.,BC Children's Hospital Research Institute, Vancouver, Canada
| | - Jane Gardiner
- BC Children's Hospital Research Institute, Vancouver, Canada.,Department of Ophthalmology and Vision Science, University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Justin Foong
- Neurosciences & Mental Health, SickKids Research Institute, Toronto, Canada
| | - Jillian Vinall
- Department of Anesthesiology, University of Calgary, Calgary, Canada
| | - Jill G Zwicker
- BC Children's Hospital Research Institute, Vancouver, Canada.,Department of Pediatrics (Developmental Pediatrics), University of British Columbia and BC Children's and Women's Hospitals, Vancouver, Canada.,Department of Occupational Science and Occupational Therapy, Vancouver, Canada.,Sunny Hill Health Centre for Children, Vancouver, Canada
| | - Ruth E Grunau
- BC Children's Hospital Research Institute, Vancouver, Canada.,Department of Pediatrics (Neonatology), University of British Columbia and BC Children's and Women's Hospitals, Vancouver, Canada
| | - Anne Synnes
- BC Children's Hospital Research Institute, Vancouver, Canada.,Department of Pediatrics (Neonatology), University of British Columbia and BC Children's and Women's Hospitals, Vancouver, Canada
| | - Kenneth J Poskitt
- BC Children's Hospital Research Institute, Vancouver, Canada.,Department of Radiology, University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Steven P Miller
- Department of Pediatrics (Neurology), University of Toronto and the Hospital for Sick Children, Toronto, Canada.,Neurosciences & Mental Health, SickKids Research Institute, Toronto, Canada.,BC Children's Hospital Research Institute, Vancouver, Canada
| |
Collapse
|
40
|
Chan-Ling T, Gole GA, Quinn GE, Adamson SJ, Darlow BA. Pathophysiology, screening and treatment of ROP: A multi-disciplinary perspective. Prog Retin Eye Res 2017; 62:77-119. [PMID: 28958885 DOI: 10.1016/j.preteyeres.2017.09.002] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 09/18/2017] [Accepted: 09/20/2017] [Indexed: 12/24/2022]
Abstract
The population of infants at risk for retinopathy of prematurity (ROP) varies by world region; in countries with well developed neonatal intensive care services, the highest risk infants are those born at less than 28 weeks gestational age (GA) and less than 1 kg at birth, while, in regions where many aspects of neonatal intensive and ophthalmological care are not routinely available, more mature infants up to 2000 g at birth and 37 weeks GA are also at risk for severe ROP. Treatment options for both groups of patients include standard retinal laser photocoagulation or, more recently, intravitreal anti-VEGF drugs. In addition to detection and treatment of ROP, this review highlights new opportunities created by telemedicine, where screening and diagnosis of ROP in remote locations can be undertaken by non-ophthalmologists using digital fundus cameras. The ophthalmological care of the ROP infant is undertaken in the wider context of neonatal care and general wellbeing of the infant. Because of this context, this review takes a multi-disciplinary perspective with contributions from retinal vascular biologists, pediatric ophthalmologists, an epidemiologist and a neonatologist. This review highlights the latest insights regarding cellular and molecular mechanisms in the formation of the retinal vasculature in the human infant, pathogenesis of ROP, detection and treatment of severe ROP, the risks and benefits of anti-VEGF therapy, the identification of new therapies over the horizon, and the optimal neonatal care regimen for best ROP outcomes, and the benefits and pitfalls of telemedicine in the remote screening and diagnosis of ROP, all of which have the potential to improve ROP outcomes.
Collapse
Affiliation(s)
- Tailoi Chan-Ling
- Department of Anatomy, School of Medical Sciences and Bosch Institute, University of Sydney, NSW 2006, Australia.
| | - Glen A Gole
- Discipline of Paediatrics and Child Health, University of Queensland, Qld Children's Hospital, Sth Brisbane, Qld 4101, Australia.
| | - Graham E Quinn
- Division of Ophthalmology, The Children's Hospital of Philadelphia and Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Samuel J Adamson
- Department of Anatomy, School of Medical Sciences and Bosch Institute, University of Sydney, NSW 2006, Australia
| | - Brian A Darlow
- Department of Paediatrics, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
41
|
Haque M, Starr LM, Koski KG, Scott ME. Differential expression of genes in fetal brain as a consequence of maternal protein deficiency and nematode infection. Int J Parasitol 2017; 48:51-58. [PMID: 28903026 DOI: 10.1016/j.ijpara.2017.07.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 07/06/2017] [Accepted: 07/10/2017] [Indexed: 11/16/2022]
Abstract
Maternal dietary protein deficiency and gastrointestinal nematode infection during early pregnancy have negative impacts on both maternal placental gene expression and fetal growth in the mouse. Here we used next-generation RNA sequencing to test our hypothesis that maternal protein deficiency and/or nematode infection also alter the expression of genes in the developing fetal brain. Outbred pregnant CD1 mice were used in a 2×2 design with two levels of dietary protein (24% versus 6%) and two levels of infection (repeated sham versus Heligmosomoides bakeri beginning at gestation day 5). Pregnant dams were euthanized on gestation day 18 to harvest the whole fetal brain. Four fetal brains from each treatment group were analyzed using RNA Hi-Seq sequencing and the differential expression of genes was determined by the edgeR package using NetworkAnalyst. In response to maternal H. bakeri infection, 96 genes (88 up-regulated and eight down-regulated) were differentially expressed in the fetal brain. Differentially expressed genes were involved in metabolic processes, developmental processes and the immune system according to the PANTHER classification system. Among the important biological functions identified, several up-regulated genes have known neurological functions including neuro-development (Gdf15, Ing4), neural differentiation (miRNA let-7), synaptic plasticity (via suppression of NF-κβ), neuro-inflammation (S100A8, S100A9) and glucose metabolism (Tnnt1, Atf3). However, in response to maternal protein deficiency, brain-specific serine protease (Prss22) was the only up-regulated gene and only one gene (Dynlt1a) responded to the interaction of maternal nematode infection and protein deficiency. In conclusion, maternal exposure to GI nematode infection from day 5 to 18 of pregnancy may influence developmental programming of the fetal brain.
Collapse
Affiliation(s)
- Manjurul Haque
- Institute of Parasitology, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada; Centre for Host Parasite Interactions, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Lisa M Starr
- Institute of Parasitology, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Kristine G Koski
- School of Human Nutrition, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada; Centre for Host Parasite Interactions, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Marilyn E Scott
- Institute of Parasitology, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada; Centre for Host Parasite Interactions, McGill University (Macdonald Campus), Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada.
| |
Collapse
|
42
|
Reactive oxygen species extend insect life span using components of the insulin-signaling pathway. Proc Natl Acad Sci U S A 2017; 114:E7832-E7840. [PMID: 28847950 DOI: 10.1073/pnas.1711042114] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Reactive oxygen species (ROS) are well-known accelerants of aging, but, paradoxically, we show that physiological levels of ROS extend life span in pupae of the moth Helicoverpa armigera, resulting in the dormant state of diapause. This developmental switch appears to operate through a variant of the conventional insulin-signaling pathway, as evidenced by the facts that Akt, p-Akt, and PRMT1 are elevated by ROS, but not insulin, and that high levels of p-Akt fail to phosphorylate FoxO through PRMT1-mediated methylation. These results suggest a distinct signaling pathway culminating in the elevation of FoxO, which in turn promotes the extension of life span characteristic of diapause.
Collapse
|
43
|
Parvaneh Tafreshi A, Talebi F, Ghorbani S, Bernard C, Noorbakhsh F. Altered expression of IGF-I system in neurons of the inflamed spinal cord during acute experimental autoimmune encephalomyelitis. J Comp Neurol 2017; 525:3072-3082. [PMID: 28617951 DOI: 10.1002/cne.24263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 06/07/2017] [Accepted: 06/09/2017] [Indexed: 11/09/2022]
Abstract
There is growing evidence that the impaired IGF-I system contributes to neurodegeneration. In this study, we examined the spinal cords of the EAE, the animal model of multiple sclerosis, to see if the expression of the IGF-I system is altered. To induce EAE, C57/BL6 mice were immunized with the Hooke lab MOG kit, sacrificed at the peak of the disease and their spinal cords were examined for the immunoreactivities (ir) of the IGF-I, IGF binding protein-1 (IGFBP-1) and glycogen synthase kinase 3β (GSK3β), as one major downstream molecule in the IGF-I signaling. Although neurons in the non EAE spinal cords did not show the IGF-I immunoreactivity, they were numerously positive for the IGFBP-1. In the inflamed EAE spinal cord however, the patterns of expressions were reversed, that is, a significant increased number of IGF-I expressing neurons versus a reduced number of IGFBP-1 positive neurons. Moreover, while nearly all IGF-I-ir neurons expressed GSK3β, some expressed it more intensely. Considering our previous finding where we showed a significant reduced number of the inactive (phosphorylated) but not that of the total GSK3β expressing neurons in the EAE spinal cord, it is conceivable that the intense total GSK3β expression in the IGF-I-ir neurons belongs to the active form of GSK3β known to exert neuroinflammatory effects. We therefore suggest that the altered expression of the IGF-I system including GSK3β in spinal cord neurons might involve in pathophysiological events during the EAE.
Collapse
Affiliation(s)
- Azita Parvaneh Tafreshi
- Department of Molecular Medicine, Faculty of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Farideh Talebi
- Department of Immunology, Faculty of Medicine, Tehran University of Medical, Sciences, Tehran, Iran
| | - Samira Ghorbani
- Department of Immunology, Faculty of Medicine, Tehran University of Medical, Sciences, Tehran, Iran
| | - Claude Bernard
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Farshid Noorbakhsh
- Department of Immunology, Faculty of Medicine, Tehran University of Medical, Sciences, Tehran, Iran
| |
Collapse
|
44
|
Marchetto MC, Belinson H, Tian Y, Freitas BC, Fu C, Vadodaria K, Beltrao-Braga P, Trujillo CA, Mendes AP, Padmanabhan K, Nunez Y, Ou J, Ghosh H, Wright R, Brennand K, Pierce K, Eichenfield L, Pramparo T, Eyler L, Barnes CC, Courchesne E, Geschwind DH, Gage FH, Wynshaw-Boris A, Muotri AR. Altered proliferation and networks in neural cells derived from idiopathic autistic individuals. Mol Psychiatry 2017; 22:820-835. [PMID: 27378147 PMCID: PMC5215991 DOI: 10.1038/mp.2016.95] [Citation(s) in RCA: 292] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/29/2016] [Accepted: 05/05/2016] [Indexed: 02/07/2023]
Abstract
Autism spectrum disorders (ASD) are common, complex and heterogeneous neurodevelopmental disorders. Cellular and molecular mechanisms responsible for ASD pathogenesis have been proposed based on genetic studies, brain pathology and imaging, but a major impediment to testing ASD hypotheses is the lack of human cell models. Here, we reprogrammed fibroblasts to generate induced pluripotent stem cells, neural progenitor cells (NPCs) and neurons from ASD individuals with early brain overgrowth and non-ASD controls with normal brain size. ASD-derived NPCs display increased cell proliferation because of dysregulation of a β-catenin/BRN2 transcriptional cascade. ASD-derived neurons display abnormal neurogenesis and reduced synaptogenesis leading to functional defects in neuronal networks. Interestingly, defects in neuronal networks could be rescued by insulin growth factor 1 (IGF-1), a drug that is currently in clinical trials for ASD. This work demonstrates that selection of ASD subjects based on endophenotypes unraveled biologically relevant pathway disruption and revealed a potential cellular mechanism for the therapeutic effect of IGF-1.
Collapse
Affiliation(s)
| | - Haim Belinson
- University of California San Francisco, Department of Pediatrics, Institute for Human Genetics, CA 94143, USA
| | - Yuan Tian
- University of California Los Angeles, Program in Neurogenetics, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, Los Angeles, CA 90402, USA
| | - Beatriz C. Freitas
- University of California San Diego, Department of Pediatrics/Rady Children’s Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093-0695, USA
| | - Chen Fu
- Case Western Reserve University, Department of Genetics and Genome Sciences, Cleveland, OH 44106, USA
| | | | - Patricia Beltrao-Braga
- University of California San Diego, Department of Pediatrics/Rady Children’s Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093-0695, USA
- University of São Paulo, Department of Obstetrics, Department of Surgery, Center for Cellular and Molecular Therapy, São Paulo, Brazil
| | - Cleber A. Trujillo
- University of California San Diego, Department of Pediatrics/Rady Children’s Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093-0695, USA
| | - Ana P.D. Mendes
- The Salk Institute, Laboratory of Genetics, La Jolla, CA 92037, USA
| | - Krishnan Padmanabhan
- University of Rochester School of Medicine and Dentistry, Department of Neuroscience, 601 Elmwood Avenue, Box 603 Rochester, NY 14642
| | - Yanelli Nunez
- The Salk Institute, Laboratory of Genetics, La Jolla, CA 92037, USA
- University of California San Diego, Department of Pediatrics/Rady Children’s Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093-0695, USA
| | - Jing Ou
- University of California Los Angeles, Program in Neurogenetics, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, Los Angeles, CA 90402, USA
| | - Himanish Ghosh
- The Salk Institute, Laboratory of Genetics, La Jolla, CA 92037, USA
| | - Rebecca Wright
- The Salk Institute, Laboratory of Genetics, La Jolla, CA 92037, USA
| | - Kristen Brennand
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Karen Pierce
- University of California San Diego, Department of Neurosciences, La Jolla, CA 92093, USA
| | - Lawrence Eichenfield
- University of California San Diego, Department of Neurosciences, La Jolla, CA 92093, USA
| | - Tiziano Pramparo
- University of California San Diego, Department of Neurosciences, La Jolla, CA 92093, USA
| | - Lisa Eyler
- University of California San Diego, Department of Neurosciences, La Jolla, CA 92093, USA
| | - Cynthia C. Barnes
- University of California San Diego, Department of Neurosciences, La Jolla, CA 92093, USA
| | - Eric Courchesne
- University of California San Diego, Department of Neurosciences, La Jolla, CA 92093, USA
| | - Daniel H. Geschwind
- University of California Los Angeles, Program in Neurogenetics, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, Los Angeles, CA 90402, USA
| | - Fred H. Gage
- The Salk Institute, Laboratory of Genetics, La Jolla, CA 92037, USA
| | - Anthony Wynshaw-Boris
- University of California San Francisco, Department of Pediatrics, Institute for Human Genetics, CA 94143, USA
- Case Western Reserve University, Department of Genetics and Genome Sciences, Cleveland, OH 44106, USA
| | - Alysson R. Muotri
- University of California San Diego, Department of Pediatrics/Rady Children’s Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093-0695, USA
| |
Collapse
|
45
|
Copani A. The underexplored question of β-amyloid monomers. Eur J Pharmacol 2017; 817:71-75. [PMID: 28577967 DOI: 10.1016/j.ejphar.2017.05.057] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/12/2017] [Accepted: 05/30/2017] [Indexed: 01/03/2023]
Abstract
Conceived more than 25 years ago, the amyloid cascade hypothesis of Alzheimer's disease has evolved to accommodate new findings, namely different forms of β-amyloid aggregates and downstream dysfunctions. Yet, the cascade does not mention its very beginning, the β-amyloid monomer. Here, I will discuss the monomer from a functional evolutionary perspective, highlighting the potential advantages of a native unfolded state that, however, involves an amyloidogenic risk. Finally, I will make a summary of what is known about its functional role in the brain and discuss the implications of its conceivable shortage in the development of Alzheimer's disease.
Collapse
Affiliation(s)
- Agata Copani
- Department of Drug Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy.
| |
Collapse
|
46
|
Neuroprotection by Paeoniflorin in the MPTP mouse model of Parkinson's disease. Neuropharmacology 2017; 116:412-420. [DOI: 10.1016/j.neuropharm.2017.01.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 01/10/2017] [Accepted: 01/11/2017] [Indexed: 12/21/2022]
|
47
|
Wrigley S, Arafa D, Tropea D. Insulin-Like Growth Factor 1: At the Crossroads of Brain Development and Aging. Front Cell Neurosci 2017; 11:14. [PMID: 28203146 PMCID: PMC5285390 DOI: 10.3389/fncel.2017.00014] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/16/2017] [Indexed: 12/15/2022] Open
Abstract
Insulin-like growth factor 1 (IGF1) is a polypeptide hormone structurally similar to insulin. It is central to the somatotropic axis, acting downstream of growth hormone (GH). It activates both the mitogen-activated protein (MAP) kinase and PI3K signaling pathways, acting in almost every tissue in the body to promote tissue growth and maturation through upregulation of anabolic processes. Overall GH and IGF1 signaling falls with age, suggesting that it is this reduced IGF1 activity that leads to age-related changes in organisms. However, mutations that reduce IGF1-signaling activity can dramatically extend the lifespan of organisms. Therefore, the role of IGF1 in the overall aging process is unclear. This review article will focus on the role of IGF1 in brain development and aging. The evidence points towards a role for IGF1 in neurodevelopment both prenatally and in the early post-natal period, and in plasticity and remodeling throughout life. This review article will then discuss the hallmarks of aging and cognitive decline associated with falls in IGF1 levels towards the end of life. Finally, the role of IGF1 will be discussed within the context of both neuropsychiatric disorders caused by impaired development of the nervous system, and neurodegenerative disorders associated with aging. IGF1 and its derivatives are shown to improve the symptoms of certain neuropsychiatric disorders caused by deranged neurodevelopment and these effects have been correlated with changes in the underlying biology in both in vitro and in vivo studies. On the other hand, studies looking at IGF1 in neurodegenerative diseases have been conflicting, supporting both a role for increased and decreased IGF1 signaling in the underlying pathogenesis of these diseases.
Collapse
Affiliation(s)
- Sarah Wrigley
- School of Medicine, Trinity College Dublin Dublin, Ireland
| | - Donia Arafa
- School of Medicine, Trinity College Dublin Dublin, Ireland
| | - Daniela Tropea
- Neuropsychiatric Genetics, Trinity Translational Medicine Institute St. James HospitalDublin, Ireland; Institute of Neuroscience, Trinity College DublinDublin, Ireland
| |
Collapse
|
48
|
Fernandez AM, Hernandez-Garzón E, Perez-Domper P, Perez-Alvarez A, Mederos S, Matsui T, Santi A, Trueba-Saiz A, García-Guerra L, Pose-Utrilla J, Fielitz J, Olson EN, Fernandez de la Rosa R, Garcia Garcia L, Pozo MA, Iglesias T, Araque A, Soya H, Perea G, Martin ED, Torres Aleman I. Insulin Regulates Astrocytic Glucose Handling Through Cooperation With IGF-I. Diabetes 2017; 66:64-74. [PMID: 27999108 DOI: 10.2337/db16-0861] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 09/28/2016] [Indexed: 11/13/2022]
Abstract
Brain activity requires a flux of glucose to active regions to sustain increased metabolic demands. Insulin, the main regulator of glucose handling in the body, has been traditionally considered not to intervene in this process. However, we now report that insulin modulates brain glucose metabolism by acting on astrocytes in concert with IGF-I. The cooperation of insulin and IGF-I is needed to recover neuronal activity after hypoglycemia. Analysis of underlying mechanisms show that the combined action of IGF-I and insulin synergistically stimulates a mitogen-activated protein kinase/protein kinase D pathway resulting in translocation of GLUT1 to the cell membrane through multiple protein-protein interactions involving the scaffolding protein GAIP-interacting protein C terminus and the GTPase RAC1. Our observations identify insulin-like peptides as physiological modulators of brain glucose handling, providing further support to consider the brain as a target organ in diabetes.
Collapse
Affiliation(s)
- Ana M Fernandez
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Edwin Hernandez-Garzón
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Paloma Perez-Domper
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Alberto Perez-Alvarez
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Sara Mederos
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Takashi Matsui
- Laboratory of Exercise Biochemistry and Neuroendocrinology, University of Tsukuba, Tsukuba, Japan
| | - Andrea Santi
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Angel Trueba-Saiz
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Lucía García-Guerra
- CIBERNED, Madrid, Spain
- Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Julia Pose-Utrilla
- CIBERNED, Madrid, Spain
- Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Jens Fielitz
- Experimental and Clinical Research Center, Charité-Universitätsmedizin, Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Brandenburg Heart Center and Medical University of Brandenburg, Brandenburg, Germany
| | - Eric N Olson
- University of Texas Southwestern Medical Center, Dallas, TX
| | | | - Luis Garcia Garcia
- Pluridisciplinary Institute, Complutense University of Madrid, Madrid, Spain
| | - Miguel Angel Pozo
- Pluridisciplinary Institute, Complutense University of Madrid, Madrid, Spain
| | - Teresa Iglesias
- CIBERNED, Madrid, Spain
- Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Alfonso Araque
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Hideaki Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, University of Tsukuba, Tsukuba, Japan
| | - Gertrudis Perea
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Eduardo D Martin
- Science and Technology Park, Institute for Research in Neurological Disabilities, University of Castilla-La Mancha, Albacete, Spain
| | - Ignacio Torres Aleman
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- CIBERNED, Madrid, Spain
| |
Collapse
|
49
|
Abstract
Diabetes mellitus represents a growing international public health issue with a near quadrupling in its worldwide prevalence since 1980. Though it has many known microvascular complications, vision loss from diabetic retinopathy is one of the most devastating for affected individuals. In addition, there is increasing evidence to suggest that diabetic patients have a greater risk for glaucoma as well. Though the pathophysiology of glaucoma is not completely understood, both diabetes and glaucoma appear to share some common risk factors and pathophysiologic similarities with studies also reporting that the presence of diabetes and elevated fasting glucose levels are associated with elevated intraocular pressure-the primary risk factor for glaucomatous optic neuropathy. While no study has completely addressed the possibility of detection bias, most recent epidemiologic evidence suggests that diabetic populations are likely enriched with glaucoma patients. As the association between diabetes and glaucoma becomes better defined, routine evaluation for glaucoma in diabetic patients, particularly in the telemedicine setting, may become a reasonable consideration to reduce the risk of vision loss in these patients.
Collapse
Affiliation(s)
- Brian J Song
- Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 243 Charles Street, Boston, MA, 02114, USA.
| | - Lloyd Paul Aiello
- Beetham Eye Institute, Joslin Diabetes Center, Harvard Medical School, 1 Joslin Place, Boston, MA, 02115, USA
| | - Louis R Pasquale
- Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 243 Charles Street, Boston, MA, 02114, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02215, USA
| |
Collapse
|
50
|
Kamphuis W, Kooijman L, Schetters S, Orre M, Hol EM. Transcriptional profiling of CD11c-positive microglia accumulating around amyloid plaques in a mouse model for Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1847-60. [DOI: 10.1016/j.bbadis.2016.07.007] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/01/2016] [Accepted: 07/13/2016] [Indexed: 12/25/2022]
|