1
|
Wang C, Fan X, Shi Y, Tang F. Radiation-Induced Brain Injury with Special Reference to Astrocytes as a Therapeutic Target. J Integr Neurosci 2025; 24:25907. [PMID: 40152565 DOI: 10.31083/jin25907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/22/2024] [Accepted: 11/06/2024] [Indexed: 03/29/2025] Open
Abstract
Radiotherapy is one of the primary modalities for oncologic treatment and has been utilized at least once in over half of newly diagnosed cancer patients. Cranial radiotherapy has significantly enhanced the long-term survival rates of patients with brain tumors. However, radiation-induced brain injury, particularly hippocampal neuronal damage along with impairment of neurogenesis, inflammation, and gliosis, adversely affects the quality of life for these patients. Astrocytes, a type of glial cell that are abundant in the brain, play essential roles in maintaining brain homeostasis and function. Despite their importance, the pathophysiological changes in astrocytes induced by radiation have not been thoroughly investigated, and no systematic or comprehensive review addressing the effects of radiation on astrocytes and related diseases has been conducted. In this paper, we review current studies on the neurophysiological roles of astrocytes following radiation exposure. We describe the pathophysiological changes in astrocytes, including astrogliosis, astrosenescence, and the associated cellular and molecular mechanisms. Additionally, we summarize the roles of astrocytes in radiation-induced impairments of neurogenesis and the blood-brain barrier (BBB). Based on current research, we propose that brain astrocytes may serve as potential therapeutic targets for treating radiation-induced brain injury (RIBI) and subsequent neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Caiping Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001 Nantong, Jiangsu, China
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 138602 Singapore, Singapore
| | - Xingjuan Fan
- Department of Neurology, Affiliated Hospital of Nantong University, 226001 Nantong, Jiangsu, China
| | - Yunwei Shi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001 Nantong, Jiangsu, China
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 138602 Singapore, Singapore
| | - Fengru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 138602 Singapore, Singapore
| |
Collapse
|
2
|
Pérez-Revuelta L, Pérez-Boyero D, Pérez-Martín E, Cabedo VL, Téllez de Meneses PG, Weruaga E, Díaz D, Alonso JR. Neuroprotective Effects of VEGF-B in a Murine Model of Aggressive Neuronal Loss with Childhood Onset. Int J Mol Sci 2025; 26:538. [PMID: 39859255 PMCID: PMC11765331 DOI: 10.3390/ijms26020538] [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: 11/07/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
In recent decades, the scientific community has faced a major challenge in the search for new therapies that can slow down or alleviate the process of neuronal death that accompanies neurodegenerative diseases. This study aimed to identify an effective therapy using neurotrophic factors to delay the rapid and aggressive cerebellar degeneration experienced by the Purkinje Cell Degeneration (PCD) mouse, a model of childhood-onset neurodegeneration with cerebellar atrophy (CONDCA). Initially, we analyzed the changes in the expression of several neurotrophic factors related to the degenerative process itself, identifying changes in insulin-like growth factor 1 (IGF-1) and Vascular Endothelial Growth Factor B (VEGF-B) in the affected animals. Then, we administered pharmacological treatments using human recombinant IGF-1 (rhIGF-1) or VEGF-B (rhVEGF-B) proteins, considering their temporal variations during the degenerative process. The effects of these treatments on motor, cognitive, and social behavior, as well as on cerebellar destructuration were analyzed. Whereas treatment with rhIGF-1 did not demonstrate any neuroprotective effect, rhVEGF-B administration at moderate dosages stopped the process of neuronal death and restored motor, cognitive, and social functions altered in PCD mice (and CONDCA patients). However, increasing the frequency of rhVEGF-B administration had a detrimental effect on Purkinje cell survival, suggesting an inverted U-shaped dose-response curve of this substance. Additionally, we demonstrate that this neuroprotective effect was achieved through a partial inhibition or delay of apoptosis. These findings provide strong evidence supporting the use of rhVEGF-B as a pharmacological agent to limit severe cerebellar neurodegenerative processes.
Collapse
Affiliation(s)
- Laura Pérez-Revuelta
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - David Pérez-Boyero
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Ester Pérez-Martín
- Neuroscience Innovative Technologies, Neurostech, 33428 Llanera, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Intervenciones Traslacionales para la Salud, 33011 Oviedo, Spain
| | - Valeria Lorena Cabedo
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Pablo González Téllez de Meneses
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Eduardo Weruaga
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - David Díaz
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - José Ramón Alonso
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
3
|
Velloso LA, Donato J. Growth Hormone, Hypothalamic Inflammation, and Aging. J Obes Metab Syndr 2024; 33:302-313. [PMID: 39639711 PMCID: PMC11704225 DOI: 10.7570/jomes24032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 09/26/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
While inflammation is a crucial response in injury repair and tissue regeneration, chronic inflammation is a prevalent feature in various chronic, non-communicable diseases such as obesity, diabetes, and cancer and in cardiovascular and neurodegenerative diseases. Long-term inflammation considerably affects disease prevalence, quality of life, and longevity. Our research indicates that the growth hormone/insulin-like growth factor 1 (GH/IGF-1) axis is a pivotal regulator of inflammation in some tissues, including the hypothalamus, which is a key player in systemic metabolism regulation. Moreover, the GH/IGF-1 axis is strongly linked to longevity, as GH- or GH receptor-deficient mice live approximately twice as long as wild-type animals and exhibit protection against aging-induced inflammation. Conversely, GH excess leads to increased neuroinflammation and reduced longevity. Our review studies the associations between the GH/IGF-1 axis, inflammation, and aging, with a particular focus on evidence suggesting that GH receptor signaling directly induces hypothalamic inflammation. This finding underscores the significant impact of changes in the GH axis on metabolism and on the predisposition to chronic, non-communicable diseases.
Collapse
Affiliation(s)
- Licio A. Velloso
- Laboratory of Cell Signalling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation, Campinas, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
4
|
Huerta de la Cruz S, Santiago-Castañeda C, Rodríguez-Palma EJ, Rocha L, Sancho M. Lateral fluid percussion injury: A rat model of experimental traumatic brain injury. Methods Cell Biol 2024; 185:197-224. [PMID: 38556449 DOI: 10.1016/bs.mcb.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Traumatic brain injury (TBI) represents one of the leading causes of disability and death worldwide. The annual economic impact of TBI-including direct and indirect costs-is high, particularly impacting low- and middle-income countries. Despite extensive research, a comprehensive understanding of the primary and secondary TBI pathophysiology, followed by the development of promising therapeutic approaches, remains limited. These fundamental caveats in knowledge have motivated the development of various experimental models to explore the molecular mechanisms underpinning the pathogenesis of TBI. In this context, the Lateral Fluid Percussion Injury (LFPI) model produces a brain injury that mimics most of the neurological and systemic aspects observed in human TBI. Moreover, its high reproducibility makes the LFPI model one of the most widely used rodent-based TBI models. In this chapter, we provide a detailed surgical protocol of the LFPI model used to induce TBI in adult Wistar rats. We further highlight the neuroscore test as a valuable tool for the evaluation of TBI-induced sensorimotor consequences and their severity in rats. Lastly, we briefly summarize the current knowledge on the pathological aspects and functional outcomes observed in the LFPI-induced TBI model in rodents.
Collapse
Affiliation(s)
- Saúl Huerta de la Cruz
- Department of Pharmacology, University of Vermont, Burlington, VT, United States; Departamento de Farmacobiología, Cinvestav Sede Sur, Ciudad de México, México.
| | | | - Erick J Rodríguez-Palma
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, Sede Sur, Mexico City, Mexico
| | - Luisa Rocha
- Departamento de Farmacobiología, Cinvestav Sede Sur, Ciudad de México, México
| | - Maria Sancho
- Department of Pharmacology, University of Vermont, Burlington, VT, United States; Department of Physiology, Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
5
|
Wasinski F, Tavares MR, Gusmao DO, List EO, Kopchick JJ, Alves GA, Frazao R, Donato J. Central growth hormone action regulates neuroglial and proinflammatory markers in the hypothalamus of male mice. Neurosci Lett 2023; 806:137236. [PMID: 37030549 PMCID: PMC10133206 DOI: 10.1016/j.neulet.2023.137236] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/10/2023]
Abstract
Growth hormone (GH) action in specific neuronal populations regulates neuroendocrine responses, metabolism, and behavior. However, the potential role of central GH action on glial function is less understood. The present study aims to determine how the hypothalamic expression of several neuroglial markers is affected by central GH action in male mice. The dwarf GH- and insulin-like growth factor-1 (IGF-1)-deficient Ghrhrlit/lit mice showed decreased mRNA expression of Nes (Nestin), Gfap, Iba1, Adgre1 (F4/80), and Tnf (TNFα) in the hypothalamus, compared to wild-type animals. In contrast, transgenic overexpression of GH led to high serum GH and IGF-1 levels, and increased hypothalamic expression of Nes, Gfap, Adgre1, Iba1, and Rax. Hepatocyte-specific GH receptor (GHR) knockout mice, which are characterized by high serum GH levels, but reduced IGF-1 secretion, showed increased mRNA expression of Gfap, Iba1, Tnf, and Sox10, demonstrating that the increase in GH levels alters the hypothalamic expression of glial markers associated with neuroinflammation, independently of IGF-1. Conversely, brain-specific GHR knockout mice showed reduced expression of Gfap, Adgre1, and Vim (vimentin), indicating that brain GHR signaling is necessary to mediate GH-induced changes in the expression of several neuroglial markers. In conclusion, the hypothalamic mRNA levels of several neuroglial markers associated with inflammation are directly modulated by GHR signaling in male mice.
Collapse
Affiliation(s)
- Frederick Wasinski
- Department of Neurology and Neurosurgery, Universidade Federal de Sao Paulo, Sao Paulo, SP 04039-032, Brazil
| | - Mariana R Tavares
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Daniela O Gusmao
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Guilherme A Alves
- Department of Anatomy, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-900, Brazil
| | - Renata Frazao
- Department of Anatomy, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-900, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil.
| |
Collapse
|
6
|
Klein T, Braunsmann L, Koschate J, Hoffmann U, Foitschik T, Krieger S, Crucian B, Schneider S, Abeln V. Short-term isolation effects on the brain, cognitive performance, and sleep-The role of exercise. Front Physiol 2023; 14:903072. [PMID: 36798941 PMCID: PMC9927017 DOI: 10.3389/fphys.2023.903072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
Isolation is stressful and negatively affects sleep and mood and might also affect the structure and function of the brain. Physical exercise improves brain function. We investigated the influence of physical exercise during isolation on sleep, affect, and neurobehavioral function. N = 16 were isolated for 30 days with daily exercise routines (ISO100) and n = 16 isolated for 45 days with every second day exercise (ISO50). N = 27 were non-isolated controls who either exercised on a daily basis (CTRLEx) or refused exercise (CTRLNonEx) for 30 days. At the beginning and the end of each intervention, intravenous morning cortisol, melatonin, brain-derived neurotrophic factor and IGF-1, positive and negative affect scales, electroencephalography, cognitive function, and sleep patterns (actigraphy) were assessed. High levels of cortisol were observed for the isolated groups (p < .05) without negative effects on the brain, cognitive function, sleep, and mood after 4 to 6 weeks of isolation, where physical exercise was performed regularly. An increase in cortisol and impairments of sleep quality, mood, cognitive function, and neurotrophic factors (p < .05) were observed after 4 weeks of absence of physical exercise in the CTRLNonEx group. These findings raise the assumption that regular physical exercise routines are a key component during isolation to maintain brain health and function.
Collapse
Affiliation(s)
- Timo Klein
- Institute of Movement and Neuroscience, German Sport University Cologne, Cologne, Germany,University of Rostock, Institute of Sport Science, Rostock, Germany,Centre for Health and Integrative Physiology in Space (CHIPS), German Sport University Cologne, Cologne, Germany,VasoActive Research Group, School of Health and Sport Sciences, University of the Sunshine Coast, Maroochydore, QLD, Australia,*Correspondence: Timo Klein, ; Vera Abeln,
| | - Leonard Braunsmann
- Institute of Movement and Neuroscience, German Sport University Cologne, Cologne, Germany
| | - Jessica Koschate
- Geriatric Medicine, Department for Health Services Research, School of Medicine and Health Sciences, University of Oldenburg, Cologne, Germany
| | - Uwe Hoffmann
- Centre for Health and Integrative Physiology in Space (CHIPS), German Sport University Cologne, Cologne, Germany
| | - Tina Foitschik
- Institute of Movement and Neuroscience, German Sport University Cologne, Cologne, Germany
| | | | - Brian Crucian
- NASA-Johnson Space Center, Houston, TX, United States
| | - Stefan Schneider
- Institute of Movement and Neurosciences, Center for Health and Integrative Physiology in Space, German Sport University Cologne, Cologne, Germany,School of Maritime Studies, Memorial University of Newfoundland, St. Johns, NL, Canada,Faculty for Science, Health, Education and Engineering, University of the Sunshine Coast, Maroochydore, QLD, Australia
| | - Vera Abeln
- Institute of Movement and Neuroscience, German Sport University Cologne, Cologne, Germany,Centre for Health and Integrative Physiology in Space (CHIPS), German Sport University Cologne, Cologne, Germany,*Correspondence: Timo Klein, ; Vera Abeln,
| |
Collapse
|
7
|
Rai M, Demontis F. Muscle-to-Brain Signaling Via Myokines and Myometabolites. Brain Plast 2022; 8:43-63. [PMID: 36448045 PMCID: PMC9661353 DOI: 10.3233/bpl-210133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Skeletal muscle health and function are important determinants of systemic metabolic homeostasis and organism-wide responses, including disease outcome. While it is well known that exercise protects the central nervous system (CNS) from aging and disease, only recently this has been found to depend on the endocrine capacity of skeletal muscle. Here, we review muscle-secreted growth factors and cytokines (myokines), metabolites (myometabolites), and other unconventional signals (e.g. bioactive lipid species, enzymes, and exosomes) that mediate muscle-brain and muscle-retina communication and neuroprotection in response to exercise and associated processes, such as the muscle unfolded protein response and metabolic stress. In addition to impacting proteostasis, neurogenesis, and cognitive functions, muscle-brain signaling influences complex brain-dependent behaviors, such as depression, sleeping patterns, and biosynthesis of neurotransmitters. Moreover, myokine signaling adapts feeding behavior to meet the energy demands of skeletal muscle. Contrary to protective myokines induced by exercise and associated signaling pathways, inactivity and muscle wasting may derange myokine expression and secretion and in turn compromise CNS function. We propose that tailoring muscle-to-CNS signaling by modulating myokines and myometabolites may combat age-related neurodegeneration and brain diseases that are influenced by systemic signals.
Collapse
Affiliation(s)
- Mamta Rai
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
8
|
Rajala A, Teel K, Bhat MA, Batushansky A, Griffin TM, Purcell L, Rajala RVS. Insulin-like growth factor 1 receptor mediates photoreceptor neuroprotection. Cell Death Dis 2022; 13:613. [PMID: 35840554 PMCID: PMC9287313 DOI: 10.1038/s41419-022-05074-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 01/21/2023]
Abstract
Insulin-like growth factor I (IGF-1) is a neurotrophic factor and is the ligand for insulin-like growth factor 1 receptor (IGF-1R). Reduced expression of IGF-1 has been reported to cause deafness, mental retardation, postnatal growth failure, and microcephaly. IGF-1R is expressed in the retina and photoreceptor neurons; however, its functional role is not known. Global IGF-1 KO mice have age-related vision loss. We determined that conditional deletion of IGF-1R in photoreceptors and pan-retinal cells produces age-related visual function loss and retinal degeneration. Retinal pigment epithelial cell-secreted IGF-1 may be a source for IGF-1R activation in the retina. Altered retinal, fatty acid, and phosphoinositide metabolism are observed in photoreceptor and retinal cells lacking IGF-1R. Our results suggest that the IGF-1R pathway is indispensable for photoreceptor survival, and activation of IGF-1R may be an essential element of photoreceptor and retinal neuroprotection.
Collapse
Affiliation(s)
- Ammaji Rajala
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Dean McGee Eye Institute, Oklahoma City, OK, 73104, USA
| | - Kenneth Teel
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Dean McGee Eye Institute, Oklahoma City, OK, 73104, USA
| | - Mohd A Bhat
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Dean McGee Eye Institute, Oklahoma City, OK, 73104, USA
| | | | | | - Lindsey Purcell
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Dean McGee Eye Institute, Oklahoma City, OK, 73104, USA
| | - Raju V S Rajala
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- Dean McGee Eye Institute, Oklahoma City, OK, 73104, USA.
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
9
|
ZeinElAbdeen YA, AbdAlSeed A, Youness RA. Decoding Insulin-Like Growth Factor Signaling Pathway From a Non-coding RNAs Perspective: A Step Towards Precision Oncology in Breast Cancer. J Mammary Gland Biol Neoplasia 2022; 27:79-99. [PMID: 35146629 DOI: 10.1007/s10911-022-09511-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/24/2022] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BC) is a highly complex and heterogenous disease. Several oncogenic signaling pathways drive BC oncogenic activity, thus hindering scientists to unravel the exact molecular pathogenesis of such multifaceted disease. This highlights the urgent need to find a key regulator that tunes up such intertwined oncogenic drivers to trim the malignant transformation process within the breast tissue. The Insulin-like growth factor (IGF) signaling pathway is a tenacious axis that is heavily intertwined with BC where it modulates the amplitude and activity of vital downstream oncogenic signaling pathways. Yet, the complexity of the pathway and the interactions driven by its different members seem to aggravate its oncogenicity and hinder its target-ability. In this review, the authors shed the light on the stubbornness of the IGF signaling pathway and its potential regulation by non-coding RNAs in different BC subtypes. Nonetheless, this review also spots light on the possible transport systems available for efficient delivery of non-coding RNAs to their respective targets to reach a personalized treatment code for BC patients.
Collapse
Affiliation(s)
- Yousra Ahmed ZeinElAbdeen
- The Molecular Genetics Research Team, Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University, Main Entrance Al Tagamoa Al Khames, New Cairo CityCairo, 11835, Egypt
| | - Amna AbdAlSeed
- The Molecular Genetics Research Team, Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University, Main Entrance Al Tagamoa Al Khames, New Cairo CityCairo, 11835, Egypt
- University of Khartoum, Al-Gama a Avenue, 11115, Khartoum, Sudan
| | - Rana A Youness
- The Molecular Genetics Research Team, Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University, Main Entrance Al Tagamoa Al Khames, New Cairo CityCairo, 11835, Egypt.
- School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, New Administrative Capital, Cairo, 11586, Egypt.
| |
Collapse
|
10
|
Research Progress on Neuroprotection of Insulin-like Growth Factor-1 towards Glutamate-Induced Neurotoxicity. Cells 2022; 11:cells11040666. [PMID: 35203315 PMCID: PMC8870287 DOI: 10.3390/cells11040666] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/05/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
Insulin-like growth factor-1 (IGF-1) and its binding proteins and receptors are widely expressed in the central nervous system (CNS), proposing IGF-1-induced neurotrophic actions in normal growth, development, and maintenance. However, while there is convincing evidence that the IGF-1 system has specific endocrine roles in the CNS, the concept is emerging that IGF-I might be also important in disorders such as ischemic stroke, brain trauma, Alzheimer’s disease, epilepsy, etc., by inducing neuroprotective effects towards glutamate-mediated excitotoxic signaling pathways. Research in rodent models has demonstrated rescue of pathophysiological and behavioral abnormalities when IGF-1 was administered by different routes, and several clinical studies have shown safety and promise of efficacy in neurological disorders of the CNS. Focusing on the relationship between IGF-1-induced neuroprotection and glutamate-induced excitatory neurotoxicity, this review addresses the research progress in the field, intending to provide a rationale for using IGF-I clinically to confer neuroprotective therapy towards neurological diseases with glutamate excitotoxicity as a common pathological pathway.
Collapse
|
11
|
Williams HC, Carlson SW, Saatman KE. A role for insulin-like growth factor-1 in hippocampal plasticity following traumatic brain injury. VITAMINS AND HORMONES 2022; 118:423-455. [PMID: 35180936 DOI: 10.1016/bs.vh.2021.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Traumatic brain injury (TBI) initiates a constellation of secondary injury cascades, leading to neuronal damage and dysfunction that is often beyond the scope of endogenous repair mechanisms. Cognitive deficits are among the most persistent morbidities resulting from TBI, necessitating a greater understanding of mechanisms of posttraumatic hippocampal damage and neuroplasticity and identification of therapies that improve recovery by enhancing repair pathways. Focusing here on hippocampal neuropathology associated with contusion-type TBIs, the impact of brain trauma on synaptic structure and function and the process of adult neurogenesis is discussed, reviewing initial patterns of damage as well as evidence for spontaneous recovery. A case is made that insulin-like growth factor-1 (IGF-1), a growth-promoting peptide synthesized in both the brain and the periphery, is well suited to augment neuroplasticity in the injured brain. Essential during brain development, multiple lines of evidence delineate roles in the adult brain for IGF-1 in the maintenance of synapses, regulation of neurotransmission, and modulation of forms of synaptic plasticity such as long-term potentiation. Further, IGF-1 enhances adult hippocampal neurogenesis though effects on proliferation and neuronal differentiation of neural progenitor cells and on dendritic growth of newly born neurons. Post-injury administration of IGF-1 has been effective in rodent models of TBI in improving learning and memory, attenuating death of mature hippocampal neurons and promoting neurogenesis, providing critical proof-of-concept data. More studies are needed to explore the effects of IGF-1-based therapies on synaptogenesis and synaptic plasticity following TBI and to optimize strategies in order to stimulate only appropriate, functional neuroplasticity.
Collapse
Affiliation(s)
- Hannah C Williams
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY, United States
| | - Shaun W Carlson
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kathryn E Saatman
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
12
|
Fronczak KM, Li Y, Henchir J, Dixon CE, Carlson SW. Reductions in Synaptic Vesicle Glycoprotein 2 Isoforms in the Cortex and Hippocampus in a Rat Model of Traumatic Brain Injury. Mol Neurobiol 2021; 58:6006-6019. [PMID: 34435329 PMCID: PMC8602666 DOI: 10.1007/s12035-021-02534-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/15/2021] [Indexed: 11/25/2022]
Abstract
Traumatic brain injury (TBI) can produce lasting cognitive, emotional, and somatic difficulties that can impact quality of life for patients living with an injury. Impaired hippocampal function and synaptic alterations have been implicated in contributing to cognitive difficulties in experimental TBI models. In the synapse, neuronal communication is facilitated by the regulated release of neurotransmitters from docking presynaptic vesicles. The synaptic vesicle glycoprotein 2 (SV2) isoforms SV2A and SV2B play central roles in the maintenance of the readily releasable pool of vesicles and the coupling of calcium to the N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex responsible for vesicle docking. Recently, we reported the findings of TBI-induced reductions in presynaptic vesicle density and SNARE complex formation; however, the effect of TBI on SV2 is unknown. To investigate this, rats were subjected to controlled cortical impact (CCI) or sham control surgery. Abundance of SV2A and SV2B were assessed at 1, 3, 7, and 14 days post-injury by immunoblot. SV2A and SV2B were reduced in the cortex at several time points and in the hippocampus at every time point assessed. Immunohistochemical staining and quantitative intensity measurements completed at 14 days post-injury revealed reduced SV2A immunoreactivity in all hippocampal subregions and reduced SV2B immunoreactivity in the molecular layer after CCI. Reductions in SV2A abundance and immunoreactivity occurred concomitantly with motor dysfunction and spatial learning and memory impairments in the 2 weeks post-injury. These findings provide novel evidence for the effect of TBI on SV2 with implications for impaired neurotransmission neurobehavioral dysfunction after TBI.
Collapse
Affiliation(s)
- Katherine M Fronczak
- Neurological Surgery, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Youming Li
- Neurological Surgery, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Jeremy Henchir
- Neurological Surgery, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - C Edward Dixon
- Neurological Surgery, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
- VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Shaun W Carlson
- Neurological Surgery, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
| |
Collapse
|
13
|
Littlejohn EL, DeSana AJ, Williams HC, Chapman RT, Joseph B, Juras JA, Saatman KE. IGF1-Stimulated Posttraumatic Hippocampal Remodeling Is Not Dependent on mTOR. Front Cell Dev Biol 2021; 9:663456. [PMID: 34095131 PMCID: PMC8174097 DOI: 10.3389/fcell.2021.663456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/26/2021] [Indexed: 01/29/2023] Open
Abstract
Adult hippocampal neurogenesis is stimulated acutely following traumatic brain injury (TBI). However, many hippocampal neurons born after injury develop abnormally and the number that survive long-term is debated. In experimental TBI, insulin-like growth factor-1 (IGF1) promotes hippocampal neuronal differentiation, improves immature neuron dendritic arbor morphology, increases long-term survival of neurons born after TBI, and improves cognitive function. One potential downstream mediator of the neurogenic effects of IGF1 is mammalian target of rapamycin (mTOR), which regulates proliferation as well as axonal and dendritic growth in the CNS. Excessive mTOR activation is posited to contribute to aberrant plasticity related to posttraumatic epilepsy, spurring preclinical studies of mTOR inhibitors as therapeutics for TBI. The degree to which pro-neurogenic effects of IGF1 depend upon upregulation of mTOR activity is currently unknown. Using immunostaining for phosphorylated ribosomal protein S6, a commonly used surrogate for mTOR activation, we show that controlled cortical impact TBI triggers mTOR activation in the dentate gyrus in a time-, region-, and injury severity-dependent manner. Posttraumatic mTOR activation in the granule cell layer (GCL) and dentate hilus was amplified in mice with conditional overexpression of IGF1. In contrast, delayed astrocytic activation of mTOR signaling within the dentate gyrus molecular layer, closely associated with proliferation, was not affected by IGF1 overexpression. To determine whether mTOR activation is necessary for IGF1-mediated stimulation of posttraumatic hippocampal neurogenesis, wildtype and IGF1 transgenic mice received the mTOR inhibitor rapamycin daily beginning at 3 days after TBI, following pulse labeling with bromodeoxyuridine. Compared to wildtype mice, IGF1 overexpressing mice exhibited increased posttraumatic neurogenesis, with a higher density of posttrauma-born GCL neurons at 10 days after injury. Inhibition of mTOR did not abrogate IGF1-stimulated enhancement of posttraumatic neurogenesis. Rather, rapamycin treatment in IGF1 transgenic mice, but not in WT mice, increased numbers of cells labeled with BrdU at 3 days after injury that survived to 10 days, and enhanced the proportion of posttrauma-born cells that differentiated into neurons. Because beneficial effects of IGF1 on hippocampal neurogenesis were maintained or even enhanced with delayed inhibition of mTOR, combination therapy approaches may hold promise for TBI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kathryn E. Saatman
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
14
|
Knockdown of miR-130a-3p alleviates spinal cord injury induced neuropathic pain by activating IGF-1/IGF-1R pathway. J Neuroimmunol 2020; 351:577458. [PMID: 33360969 DOI: 10.1016/j.jneuroim.2020.577458] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/29/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recent studies have elucidated the instrumental role of microRNAs (miRNAs) in neuropathic pain (NP) progression. As one member of miRNAs, miR-130a-3p has been proved as a mediator in inflammation and neuronal maturation. The present study attempted to elucidate what effect miR-130a-3p exerts on NP. MATERIALS AND METHODS The miR-130a-3p expression in the spinal cord tissues of rat with spinal cord compression injury (SCI) and LPS-induced BV2 microglia was determined with RT-PCR, which was further applied to analyze the clinical relevance between miR-130a-3p and neuropathic pain. Besides, the expression of IGF-1, IL-1β, IL-6, and TNF-α in the spinal cord tissues of rats was measured using RT-PCR and ELISA after intrathecal injection of miR-130a-3p inhibitors and tail vein injection of IGF-1 low-expression lentivirus (Lv-shIGF-1). Further, neuronal apoptosis (labeled by Caspase3) and microglial activation (labeled by Iba1) were examined by immunohistochemistry (IHC), and the levels of IGF-1, IGF-1R, NF-κB were determined by western blot. Additionally, bioinformatic was employed to analyze the potential target genes of miR-130a-3p. Furthermore, the dual luciferase activity assay and RNA immunoprecipitation assay were conducted to further substantiate whether miR-130a-3p targets IGF-1. RESULTS In comparison with the sham group, the miR-130a-3p expression was remarkably up-regulated in the spinal cord lesions of SCI rats. The ELISA results showed that inhibiting the miR-130a-3p significantly reduced NP symptoms of SCI rats, mitigated neuronal apoptosis, microglial activation, repressed NF-κB phosphorylation and the IL-1β, IL-6 and TNF-α expressions in SCI rats. Contrarily, downregulation of miR-130a-3p increased IGF-1 and IGF-1R expression. What's more, we observed the same effects in BV2 microglia. In addition, the bioinformatics analysis showed that miR-130-3p targeted at the 3'-untranslated region of IGF-1 and inhibiting its expression. However, abolishing IGF-1 not only promoted the inflammatory responses in the SCI lesions, but also aggravated NP of SCI rats, while those effects were attenuated by the downregulation of miR-130a-3p. CONCLUSION The inhibition of miR-130a-3p expression up-regulates the IGF-1/IGF-1R signaling pathway, thus reducing neuropathic pain caused by spinal cord injury.
Collapse
|
15
|
Wasinski F, Klein MO, Bittencourt JC, Metzger M, Donato J. Distribution of growth hormone-responsive cells in the brain of rats and mice. Brain Res 2020; 1751:147189. [PMID: 33152340 DOI: 10.1016/j.brainres.2020.147189] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/25/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023]
Abstract
A growth hormone (GH) injection is able to induce the phosphorylated form of the signal transducer and activator of transcription 5 (pSTAT5) in a large number of cells throughout the mouse brain. The present study had the objective to map the distribution of GH-responsive cells in the brain of rats that received an intracerebroventricular injection of GH and compare it to the pattern found in mice. We observed that rats and mice exhibited a similar distribution of GH-induced pSTAT5 in the majority of areas of the telencephalon, hypothalamus and brainstem. However, rats exhibited a higher density of GH-responsive cells than mice in the horizontal limb of the diagonal band of Broca (HDB), supraoptic and suprachiasmatic nuclei, whereas mice displayed more GH-responsive cells than rats in the hippocampus, lateral hypothalamic area and dorsal motor nucleus of the vagus (DMX). Since both HDB and DMX contain acetylcholine-producing neurons, pSTAT5 was co-localized with choline acetyltransferase in GH-injected animals. We found that 50.0 ± 4.5% of cholinergic neurons in the rat HDB coexpressed GH-induced pSTAT5, whereas very few co-localizations were observed in the mouse HDB. In contrast, rats displayed fewer cholinergic neurons responsive to GH in the DMX at the level of the area postrema. In summary, pSTAT5 can be used as a marker of GH-responsive cells in the rat brain. Although rats and mice exhibit a relatively similar distribution of GH-responsive neurons, some species-specific differences exist, as exemplified for the responsiveness to GH in distinct populations of cholinergic neurons.
Collapse
Affiliation(s)
- Frederick Wasinski
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil
| | - Marianne O Klein
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil
| | - Jackson C Bittencourt
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil
| | - Martin Metzger
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil
| | - Jose Donato
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil.
| |
Collapse
|
16
|
Adamis D, Coada I, Eikelenboom P, Chu CS, Finn K, Melvin V, Williams J, Meagher DJ, McCarthy G. Delirium, insulin-like growth factor I, growth hormone in older inpatients. World J Psychiatry 2020; 10:212-222. [PMID: 33014722 PMCID: PMC7515747 DOI: 10.5498/wjp.v10.i9.212] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 06/16/2020] [Accepted: 08/25/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Delirium is a common disorder in elderly medical inpatients with serious adverse outcomes and is characterized by sudden onset, disturbance in attention, awareness, consciousness and cognition, and often with behavioural disturbances. Central to understanding delirium, is understanding mechanisms by which body and brain wellbeing are linked and in particular how brain responses to bodily homeostatic stress is mediated. A number of studies have investigated the relationship between insulin-like growth factor I (IGF-I) and delirium in medically ill hospitalised patients with conflicting results. However, none have investigated growth hormone (GH) which is related to IGF-I via negative feedback.
AIM To investigate the relationship between serum levels of IGF-I and GH, and the occurrence of delirium.
METHODS Prospective, longitudinal, observational study. Consecutive elderly inpatients (aged 70+), were assessed twice weekly with Montreal cognitive assessment (MoCA), Confusion assessment method (CAM), Acute Physiology and Chronic Health Evaluation II. Delirium was defined using CAM. Previous history of dementia was evaluated with the Informant Questionnaire on Cognitive Decline in the Elderly. IGF-I and GH levels were estimated with the ELISA method. Generalized estimating equations (GEE) model was applied for the first five assessments to analyze those longitudinal data.
RESULTS The sample consisted of 198 participants (mean age 80.63 ± 6.81; range 70-97). Of these 92 (46.5%) were females. Eighty six (43.4%) were identified with a history of dementia. Incident or prevalent delirium during hospitalisation was identified with CAM in 40 participants (20.2%). Evaluation of missing values with Little's MCAR test indicated that they were missing completely at random (MCAR χ2 = 12.24, u: 9, P = 0.20). Using GEE for the analysis we found that low MoCA scores, low levels of IGF-I and high levels of GH were significantly associated with any delirium (prevalence, incident, or fluctuating , during the study period (Wald χ2 = 12.231; u: 1, P < 0.001, Wald χ2 = 7.196, u: 1, P = 0.007, Wald χ2 = 6.210; : u: 1, P = 0.013 respectively).
CONCLUSION The results show that low levels of IGF-I, high levels of GH and low scores in cognition are independently associated with the occurrence of any delirium during the hospitalisation of medically ill older people. The results of the study supports the hypothesis that deficits in the immunoreactivity of the brain (low cerebral reserve) may be associated with delirium.
Collapse
Affiliation(s)
- Dimitrios Adamis
- Department of Psychiatry, Sligo/Leitrim Mental Health Services, Sligo F91 CD34, Ireland
- Department of Psychiatry, Research and Academic Institute of Athens, Athens 11742, Greece
- Department of Psychiatry, University Hospital Limerick, Limerick V94 F858, Ireland
| | - Iulian Coada
- Department of Psychiatry, Sligo/Leitrim Mental Health Services, Sligo F91 CD34, Ireland
| | - Piet Eikelenboom
- Department of Psychiatry, GGZinGeest and VuMC, Amsterdam 1081 HV, the Netherlands
| | - Che-Sheng Chu
- Department of Psychiatry and Center for Geriatric and Gerontology, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
| | - Karen Finn
- Department of Biopharmaceutical and Medical Science, School of Science and Computing, Galway-Mayo Institute of Technology, Galway H91 T8NW, Ireland
| | - Vincent Melvin
- Department of Psychiatry, Sligo/Leitrim Mental Health Services, Sligo F91 CD34, Ireland
| | - John Williams
- Department of Pathology, Sligo University Hospital, Sligo F91 H684, Ireland
| | - David James Meagher
- Department of Psychiatry, University Hospital Limerick, Limerick V94 F858, Ireland
| | - Geraldine McCarthy
- Department of Psychiatry, Sligo/Leitrim Mental Health Services, Sligo F91 CD34, Ireland
| |
Collapse
|
17
|
Nishida F, Zanuzzi CN, Sisti MS, Falomir Lockhart E, Camiña AE, Hereñú CB, Bellini MJ, Portiansky EL. Intracisternal IGF-1 gene therapy abrogates kainic acid-induced excitotoxic damage of the rat spinal cord. Eur J Neurosci 2020; 52:3339-3352. [PMID: 32573850 DOI: 10.1111/ejn.14876] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 12/28/2022]
Abstract
Development of alternative therapies for treating functional deficits after different neurological damages is a challenge in neuroscience. Insulin-like growth factor-1 (IGF-1) is a potent neurotrophic factor exerting neuroprotective actions in brain and spinal cord. It is used to prevent or treat injuries of the central nervous system using different administration routes in different animal models. In this study, we evaluated whether intracisternal (IC) route for IGF-1 gene therapy may abrogate or at least reduce the structural and behavioral damages induced by the intraparenchymal injection of kainic acid (KA) into the rat spinal cord. Experimental (Rad-IGF-1) and control (Rad-DsRed-KA) rats were evaluated using a battery of motor and sensory tests before the injection of the recombinant adenovector (day -3), before KA injection (day 0) and at every post-injection (pi) time point (days 1, 2, 3 and 7 pi). Histopathological changes and neuronal and glial counting were assessed. Pretreatment using IC delivery of RAd-IGF-1 improved animal's general condition and motor and sensory functions as compared to Rad-DsRed-KA-injected rats. Besides, IC Rad-IGF-1 therapy abrogated later spinal cord damage and reduced the glial response induced by KA as observed in Rad-DsRed-KA rats. We conclude that the IC route for delivering RAd-IGF-1 prevents KA-induced excitotoxicity in the spinal cord.
Collapse
Affiliation(s)
- Fabián Nishida
- Image Analysis Laboratory, School of Veterinary Sciences, National University of La Plata (UNLP), La Plata, Argentina.,National Research Council of Science and Technology (CONICET), Buenos Aires, Argentina
| | - Carolina N Zanuzzi
- Image Analysis Laboratory, School of Veterinary Sciences, National University of La Plata (UNLP), La Plata, Argentina.,National Research Council of Science and Technology (CONICET), Buenos Aires, Argentina.,Department of Histology and Embryology, School of Veterinary Sciences, National University of La Plata (UNLP), Buenos Aires, Argentina
| | - María S Sisti
- Image Analysis Laboratory, School of Veterinary Sciences, National University of La Plata (UNLP), La Plata, Argentina.,National Research Council of Science and Technology (CONICET), Buenos Aires, Argentina
| | - Eugenia Falomir Lockhart
- National Research Council of Science and Technology (CONICET), Buenos Aires, Argentina.,INIBIOLP-Histology B, School of Medicine, National University of La Plata (UNLP), La Plata, Argentina
| | - Agustina E Camiña
- Image Analysis Laboratory, School of Veterinary Sciences, National University of La Plata (UNLP), La Plata, Argentina
| | - Claudia B Hereñú
- Department of Pharmacology, School of Chemistry, National University of Córdoba (UNC), Córdoba, Argentina.,Institute for Experimental Pharmacology, Córdoba, Argentina
| | - María J Bellini
- National Research Council of Science and Technology (CONICET), Buenos Aires, Argentina.,INIBIOLP-Histology B, School of Medicine, National University of La Plata (UNLP), La Plata, Argentina
| | - Enrique L Portiansky
- Image Analysis Laboratory, School of Veterinary Sciences, National University of La Plata (UNLP), La Plata, Argentina.,National Research Council of Science and Technology (CONICET), Buenos Aires, Argentina
| |
Collapse
|
18
|
Kim HN, Langley MR, Simon WL, Yoon H, Kleppe L, Lanza IR, LeBrasseur NK, Matveyenko A, Scarisbrick IA. A Western diet impairs CNS energy homeostasis and recovery after spinal cord injury: Link to astrocyte metabolism. Neurobiol Dis 2020; 141:104934. [PMID: 32376475 PMCID: PMC7982964 DOI: 10.1016/j.nbd.2020.104934] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/28/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
A diet high in fat and sucrose (HFHS), the so-called Western diet promotes metabolic syndrome, a significant co-morbidity for individuals with spinal cord injury (SCI). Here we demonstrate that the spinal cord of mice consuming HFHS expresses reduced insulin-like growth factor 1 (IGF-1) and its receptor and shows impaired tricarboxylic acid cycle function, reductions in PLP and increases in astrogliosis, all prior to SCI. After SCI, Western diet impaired sensorimotor and bladder recovery, increased microgliosis, exacerbated oligodendrocyte loss and reduced axon sprouting. Direct and indirect neural injury mechanisms are suggested since HFHS culture conditions drove parallel injury responses directly and indirectly after culture with conditioned media from HFHS-treated astrocytes. In each case, injury mechanisms included reductions in IGF-1R, SIRT1 and PGC-1α and were prevented by metformin. Results highlight the potential for a Western diet to evoke signs of neural insulin resistance and injury and metformin as a strategy to improve mechanisms of neural neuroprotection and repair.
Collapse
Affiliation(s)
- Ha Neui Kim
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America; Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Monica R Langley
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America; Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Whitney L Simon
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America; Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Laurel Kleppe
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Ian R Lanza
- Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Nathan K LeBrasseur
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America; Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Aleksey Matveyenko
- Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Isobel A Scarisbrick
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America; Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America; Neurosciuence Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America.
| |
Collapse
|
19
|
Turnquist C, Beck JA, Horikawa I, Obiorah IE, Von Muhlinen N, Vojtesek B, Lane DP, Grunseich C, Chahine JJ, Ames HM, Smart DD, Harris BT, Harris CC. Radiation-induced astrocyte senescence is rescued by Δ133p53. Neuro Oncol 2020; 21:474-485. [PMID: 30615147 DOI: 10.1093/neuonc/noz001] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Cellular senescence and the senescence-associated secretory phenotype (SASP) may contribute to the development of radiation therapy-associated side effects in the lung and blood vessels by promoting chronic inflammation. In the brain, inflammation contributes to the development of neurologic disease, including Alzheimer's disease. In this study, we investigated the roles of cellular senescence and Δ133p53, an inhibitory isoform of p53, in radiation-induced brain injury. METHODS Senescent cell types in irradiated human brain were identified with immunohistochemical labeling of senescence-associated proteins p16INK4A and heterochromatin protein Hp1γ in 13 patient cases, including 7 irradiated samples. To investigate the impact of radiation on astrocytes specifically, primary human astrocytes were irradiated and examined for expression of Δ133p53 and induction of SASP. Lentiviral expression of ∆133p53 was performed to investigate its role in regulating radiation-induced cellular senescence and astrocyte-mediated neuroinflammation. RESULTS Astrocytes expressing p16INK4A and Hp1γ were identified in all irradiated tissues, were increased in number in irradiated compared with untreated cancer patient tissues, and had higher labeling intensity in irradiated tissues compared with age-matched controls. Human astrocytes irradiated in vitro also experience induction of cellular senescence, have diminished Δ133p53, and adopt a neurotoxic phenotype as demonstrated by increased senescence-associated beta-galactosidase activity, p16INK4A, and interleukin (IL)-6. In human astrocytes, Δ133p53 inhibits radiation-induced senescence, promotes DNA double-strand break repair, and prevents astrocyte-mediated neuroinflammation and neurotoxicity. CONCLUSIONS Restoring expression of the endogenous p53 isoform, ∆133p53, protects astrocytes from radiation-induced senescence, promotes DNA repair, and inhibits astrocyte-mediated neuroinflammation.
Collapse
Affiliation(s)
- Casmir Turnquist
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jessica A Beck
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Izumi Horikawa
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ifeyinwa E Obiorah
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
| | - Natalia Von Muhlinen
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Borivoj Vojtesek
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - David P Lane
- p53 Laboratory, Biomedical Sciences Institutes (A*STAR), Singapore
| | - Christopher Grunseich
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Joeffrey J Chahine
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
| | - Heather M Ames
- Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland, USA.,Department of Pathology, University of Maryland, Baltimore, Maryland, USA
| | - Dee Dee Smart
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Brent T Harris
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA.,Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
20
|
Littlejohn EL, Scott D, Saatman KE. Insulin-like growth factor-1 overexpression increases long-term survival of posttrauma-born hippocampal neurons while inhibiting ectopic migration following traumatic brain injury. Acta Neuropathol Commun 2020; 8:46. [PMID: 32276671 PMCID: PMC7147070 DOI: 10.1186/s40478-020-00925-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 03/29/2020] [Indexed: 01/29/2023] Open
Abstract
Cellular damage associated with traumatic brain injury (TBI) manifests in motor and cognitive dysfunction following injury. Experimental models of TBI reveal cell death in the granule cell layer (GCL) of the hippocampal dentate gyrus acutely after injury. Adult-born neurons residing in the neurogenic niche of the GCL, the subgranular zone, are particularly vulnerable. Injury-induced proliferation of neural progenitors in the subgranular zone supports recovery of the immature neuron population, but their development and localization may be altered, potentially affecting long-term survival. Here we show that increasing hippocampal levels of insulin-like growth factor-1 (IGF1) is sufficient to promote end-stage maturity of posttrauma-born neurons and improve cognition following TBI. Mice with conditional overexpression of astrocyte-specific IGF1 and wild-type mice received controlled cortical impact or sham injury and bromo-2'-deoxyuridine injections for 7d after injury to label proliferating cells. IGF1 overexpression increased the number of GCL neurons born acutely after trauma that survived 6 weeks to maturity (NeuN+BrdU+), and enhanced their outward migration into the GCL while significantly reducing the proportion localized ectopically to the hilus and molecular layer. IGF1 selectively affected neurons, without increasing the persistence of posttrauma-proliferated glia in the dentate gyrus. IGF1 overexpressing animals performed better during radial arm water maze reversal testing, a neurogenesis-dependent cognitive test. These findings demonstrate the ability of IGF1 to promote the long-term survival and appropriate localization of granule neurons born acutely after a TBI, and suggest these new neurons contribute to improved cognitive function.
Collapse
Affiliation(s)
- Erica L. Littlejohn
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, B473 Biomedical & Biological Sciences Research Building (BBSRB), 741 South Limestone St, Lexington, KY 40536-0509 USA
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3901 USA
| | - Danielle Scott
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, B473 Biomedical & Biological Sciences Research Building (BBSRB), 741 South Limestone St, Lexington, KY 40536-0509 USA
| | - Kathryn E. Saatman
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, B473 Biomedical & Biological Sciences Research Building (BBSRB), 741 South Limestone St, Lexington, KY 40536-0509 USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536 USA
| |
Collapse
|
21
|
Wasinski F, Frazão R, Donato J. Effects of growth hormone in the central nervous system. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2019; 63:549-556. [PMID: 31939479 PMCID: PMC10522235 DOI: 10.20945/2359-3997000000184] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 08/29/2019] [Indexed: 11/23/2022]
Abstract
Growth hormone (GH) is best known for its effect stimulating tissue and somatic growth through the regulation of cell division, regeneration and proliferation. However, GH-responsive neurons are spread over the entire central nervous system, suggesting that they have important roles in the brain. The objective of the present review is to summarize and discuss the potential physiological importance of GH action in the central nervous system. We provide evidence that GH signaling in the brain regulates the physiology of numerous functions such as cognition, behavior, neuroendocrine changes and metabolism. Data obtained from experimental animal models have shown that disruptions in GH signaling in specific neuronal populations can affect the reproductive axis and impair food intake during glucoprivic conditions, neuroendocrine adaptions during food restriction, and counter-regulatory responses to hypoglycemia, and they can modify gestational metabolic adaptions. Therefore, the brain is an important target tissue of GH, and changes in GH action in the central nervous system can explain some dysfunctions presented by individuals with excessive or deficient GH secretion. Furthermore, GH acts in specific neuronal populations during situations of metabolic stress to promote appropriate physiological adjustments that restore homeostasis. Arch Endocrinol Metab. 2019;63(6):549-56.
Collapse
Affiliation(s)
- Frederick Wasinski
- Departamento de Fisiologia e BiofísicaInstituto de Ciências BiomédicasUniversidade de São PauloSão PauloSPBrasilDepartamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, SP, Brasil
| | - Renata Frazão
- Departamento de AnatomiaInstituto de Ciências BiomédicasUniversidade de São PauloSão PauloSPBrasilDepartamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, SP, Brasil
| | - Jose Donato
- Departamento de Fisiologia e BiofísicaInstituto de Ciências BiomédicasUniversidade de São PauloSão PauloSPBrasilDepartamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, SP, Brasil
| |
Collapse
|
22
|
Santi A, Genis L, Torres Aleman I. A Coordinated Action of Blood-Borne and Brain Insulin-Like Growth Factor I in the Response to Traumatic Brain Injury. Cereb Cortex 2019; 28:2007-2014. [PMID: 28449086 DOI: 10.1093/cercor/bhx106] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/11/2017] [Indexed: 01/20/2023] Open
Abstract
In response to injury, the brain produces different neuroprotective molecules, such as insulin-like growth factor I (IGF-I). However, IGF-I is also taken up by the brain from the circulation in response to physiological stimuli. Herein, we analyzed in mice the relative contribution of circulating and locally produced IGF-I to increased brain IGF-I levels after insult. Traumatic brain injury (TBI) induced by a controlled impact resulted in increased IGF-I levels in the vicinity of the lesion, but mice with low serum IGF-I showed significantly lower increases. Indeed, in normal mice, peripheral IGF-I accumulated at the lesion site after injury, and at the same time serum IGF-I levels decreased. Collectively, these data suggest that serum IGF-I enter into the brain after TBI and contributes to increased brain IGF-I levels at the injury site. This connection between central and circulating IGF-I provides an amenable route for treatment, as subcutaneous administration of IGF-I to TBI mice led to functional recovery. These latter results add further support to the use of systemic IGF-I or its mimetics for treatment of brain injuries.
Collapse
Affiliation(s)
- A Santi
- Cajal Institute, CSIC, Avda Dr Arce 37, 28002 Madrid, Spain.,Ciberned, C/ Valderrebollo 5, 28031 Madrid, Spain
| | - L Genis
- Cajal Institute, CSIC, Avda Dr Arce 37, 28002 Madrid, Spain.,Ciberned, C/ Valderrebollo 5, 28031 Madrid, Spain
| | - I Torres Aleman
- Cajal Institute, CSIC, Avda Dr Arce 37, 28002 Madrid, Spain.,Ciberned, C/ Valderrebollo 5, 28031 Madrid, Spain
| |
Collapse
|
23
|
Neurophysiological, neuropsychological, and cognitive effects of 30 days of isolation. Exp Brain Res 2019; 237:1563-1573. [DOI: 10.1007/s00221-019-05531-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 03/25/2019] [Indexed: 01/22/2023]
|
24
|
Yang Y, Liang S, Li Y, Gao F, Cao Y, Zhao X, Gao G, Li L. Effects of early administration of insulin-like growth factor-1 on cognitive function in septic encephalopathy. Neuropsychiatr Dis Treat 2019; 15:323-337. [PMID: 30774344 PMCID: PMC6353230 DOI: 10.2147/ndt.s190845] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Both protective and therapeutic functions of insulin-like growth factor-1 (IGF-1) in brain injury have been reported, but its effects on cognitive sequelae after septic encephalopathy (SE) remain unclear. MATERIALS AND METHODS This study was divided into three parts, and a septic model was built by cecal ligation and puncture (CLP). First, survival analysis was performed, and IGF-1's effects on long-term cognition and depressive emotion were assessed. Second, the characteristics of IGF-1 function in cognition were evaluated. Finally, cytochrome C, caspase-9, tumor necrosis factor receptor (TNFR), and caspase-8 levels as well as cell apoptosis in the hippocampus were evaluated. RESULTS IGF-1 did not reduce mortality or alleviate depressive symptoms in septic rats, but improved the memory of noxious stimulation and spatial learning and memory. These effects were observed only when IGF-1 was administered within 0-6 hours after CLP. Moreover, cytochrome C and caspase-9 expression levels were increased at 6 hours after CLP in the hippocampus, while TNFR and caspase-8 amounts were not increased until 12 hours after CLP. Cell apoptosis increased at 12 hours after CLP, but was inhibited by IGF-1. CONCLUSION Cognitive impairment in rats recovering from SE is associated with cell apoptosis in the hippocampus. Supplementation of IGF-1 reduces cell apoptosis by preventing the over-expression of cytochrome C and TNFR, and results in improved cognitive function. However, improvement only occurs when IGF-1 is administered at the early stage (within 6 hours) of sepsis. As cytochrome C activation occurs earlier than that of TNFR in this study, cytochrome C may be the main factor inducing apoptosis in early SE.
Collapse
Affiliation(s)
- Yang Yang
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China, ;
| | - Shengru Liang
- Department of Endocrinology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710032, China
| | - Yuqian Li
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China, ;
| | - Fei Gao
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China, ;
| | - Yuan Cao
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710032, China
| | - Xiaoyu Zhao
- Department of Neurosurgery, The 986th Hospital of Chinese People's Libertation Army, Xi'an, Shaanxi Province 710054, China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China, ;
| | - Lihong Li
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China, ;
| |
Collapse
|
25
|
Skop NB, Singh S, Antikainen H, Saqcena C, Calderon F, Rothbard DE, Cho CH, Gandhi CD, Levison SW, Dobrowolski R. Subacute Transplantation of Native and Genetically Engineered Neural Progenitors Seeded on Microsphere Scaffolds Promote Repair and Functional Recovery After Traumatic Brain Injury. ASN Neuro 2019; 11:1759091419830186. [PMID: 30818968 PMCID: PMC6399762 DOI: 10.1177/1759091419830186] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/11/2018] [Accepted: 12/18/2018] [Indexed: 01/09/2023] Open
Abstract
There is intense interest and effort toward regenerating the brain after severe injury. Stem cell transplantation after insult to the central nervous system has been regarded as the most promising approach for repair; however, engrafting cells alone might not be sufficient for effective regeneration. In this study, we have compared neural progenitors (NPs) from the fetal ventricular zone (VZ), the postnatal subventricular zone, and an immortalized radial glia (RG) cell line engineered to conditionally secrete the trophic factor insulin-like growth factor 1 (IGF-1). Upon differentiation in vitro, the VZ cells were able to generate a greater number of neurons than subventricular zone cells. Furthermore, differentiated VZ cells generated pyramidal neurons . In vitro, doxycycline-driven secretion of IGF-1 strongly promoted neuronal differentiation of cells with hippocampal, interneuron and cortical specificity. Accordingly, VZ and engineered RG-IGF-1-hemagglutinin (HA) cells were selected for subsequent in vivo experiments. To increase cell survival, we delivered the NPs attached to a multifunctional chitosan-based scaffold. The microspheres containing adherent NPs were injected subacutely into the lesion cavity of adult rat brains that had sustained controlled cortical impact injury. At 2 weeks posttransplantation, the exogenously introduced cells showed a reduction in stem cell or progenitor markers and acquired mature neuronal and glial markers. In beam walking tests assessing sensorimotor recovery, transplanted RG cells secreting IGF-1 contributed significantly to functional improvement while native VZ or RG cells did not promote significant recovery. Altogether, these results support the therapeutic potential of chitosan-based multifunctional microsphere scaffolds seeded with genetically modified NPs expressing IGF-1 to promote repair and functional recovery after traumatic brain injuries.
Collapse
Affiliation(s)
- Nolan B. Skop
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, NJ, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Sweta Singh
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
- Stem Cell and Gene Therapy Research Group, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Henri Antikainen
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Chaitali Saqcena
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Frances Calderon
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, NJ, USA
| | - Deborah E. Rothbard
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, NJ, USA
| | - Cheul H. Cho
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Chirag D. Gandhi
- Department of Neurosurgery, Westchester Medical Center at NY Medical College, Valhalla, NY, USA
| | - Steven W. Levison
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, NJ, USA
| | - Radek Dobrowolski
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, TX, USA
| |
Collapse
|
26
|
Sama DM, Carlson SW, Joseph B, Saenger S, Metzger F, Saatman KE. Assessment of systemic administration of PEGylated IGF-1 in a mouse model of traumatic brain injury. Restor Neurol Neurosci 2018; 36:559-569. [PMID: 29889090 DOI: 10.3233/rnn-180831] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Traumatic brain injury can result in lasting cognitive dysfunction due to degeneration of mature hippocampal neurons as well as the loss of immature neurons within the dentate gyrus. While endogenous neurogenesis affords a partial recovery of the immature neuron population, hippocampal neurogenesis may be enhanced through therapeutic intervention. Insulin-like growth factor-1 (IGF-1) has the potential to improve cognitive function and promote neurogenesis after TBI, but its short half-life in the systemic circulation makes it difficult to maintain a therapeutic concentration. IGF-1 modified with a polyethylene glycol moiety (PEG-IGF-1) exhibits improved stability and half-life while retaining its ability to enter the brain from the periphery, increasing its viability as a translational approach. OBJECTIVE The goal of this study was to evaluate the ability of systemic PEG-IGF-1 administration to attenuate acute neuronal loss and stimulate the recovery of hippocampal immature neurons in brain-injured mice. METHODS In a series of studies utilizing a well-established contusion brain injury model, PEG-IGF-1 was administered subcutaneously after injury. Serum levels of PEG were verified using ELISA and histological staining was used to investigate numbers of degenerating neurons and cortical contusion size at 24 h after injury. Immunofluorescent staining was used to evaluate numbers of immature neurons at 10 d after injury. RESULTS Although subcutaneous injections of PEG-IGF-1 increased serum IGF-1 levels in a dose-dependent manner, no effects were observed on cortical contusion size, neurodegeneration within the dentate gyrus, or recovery of hippocampal immature neuron numbers. CONCLUSIONS In contrast to its efficacy in rodent models of neurodegenerative diseases, PEG- IGF-1 was not effective in ameliorating early neuronal loss after contusion brain trauma.
Collapse
Affiliation(s)
- Diana M Sama
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Shaun W Carlson
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA.,Present address: Department of Neurological Surgery, Safar Center for Resuscitation Research, University of Pittsburgh, PA, USA
| | - Binoy Joseph
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Stefanie Saenger
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, Grenzacherstrasse, Basel, Switzerland
| | - Friedrich Metzger
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, Grenzacherstrasse, Basel, Switzerland.,Versameb AG, Peter Merian-Strasse, Basel, Switzerland
| | - Kathryn E Saatman
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
27
|
Dubiel R, Callender L, Dunklin C, Harper C, Bennett M, Kreber L, Auchus R, Diaz-Arrastia R. Phase 2 Randomized, Placebo-Controlled Clinical Trial of Recombinant Human Growth Hormone (rhGH) During Rehabilitation From Traumatic Brain Injury. Front Endocrinol (Lausanne) 2018; 9:520. [PMID: 30250451 PMCID: PMC6139310 DOI: 10.3389/fendo.2018.00520] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 08/20/2018] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability, but there are currently no therapies with proven efficacy for optimizing regeneration of repair during rehabilitation. Using standard stimulation tests, as many as 40-50% of survivors of severe TBI have deficiency of one or more pituitary hormones. Of these, the somatotropic axis is the most commonly affected, with Growth Hormone (GH) deficiency affecting ~20% of persons with severe TBI. Treatment with recombinant human Growth Hormone (rhGH) is generally effective in reversing the effects of acquired GH deficiency, but there is no evidence documenting functional or neurocognitive improvement after GH replacement in TBI patients. As a consequence, screening for GH deficiency and GH replacement when deficiency is found is not routinely performed as part of the rehabilitation of TBI survivors. Given that most of the recovery after TBI occurs within the first 6-12 months after injury and IGF-1 and GH are part of a coordinated restorative neurotrophic system, we hypothesized that patients will optimally benefit from GH therapy during the window of maximal neuroregenerative activity. We performed a Phase IIa, randomized, double-blind, placebo-controlled feasibility trial of recombinant human Growth Hormone (rhGH), starting at discharge from an inpatient rehabilitation unit, with follow up at 6 and 12 months. Our primary hypothesis was that treatment with rhGH in the subacute period would result in improved functional outcomes 6 months after injury. Our secondary hypothesis proposed that treatment with rhGH would increase IGF-1 levels and be well tolerated. Sixty-three subjects were randomized, and 40 completed the trial. At baseline, there was no correlation between IGF-1 levels and peak GH levels after L-arginine stimulation. IGF-1 levels increased after rhGH treatment, but it took longer than 1 month for levels to be higher than for placebo-treated patients. rhGH therapy was well-tolerated. The rhGH group was no different from placebo in the Disability Rating Scale, Glasgow Outcome Scale-Extended, or neuropsychological function. However, a trend toward greater improvement from baseline in Functional Independence Measure (FIM) was noted in the rhGH treated group. Future studies should include longer treatment periods, faster titration of rhGH, and larger sample sizes.
Collapse
Affiliation(s)
- Rosemary Dubiel
- Department of Physical Medicine and Rehabilitation, Baylor Institute for Rehabilitation, Dallas, TX, United States
| | - Librada Callender
- Department of Physical Medicine and Rehabilitation, Baylor Institute for Rehabilitation, Dallas, TX, United States
| | - Cynthia Dunklin
- Department of Physical Medicine and Rehabilitation, Baylor Institute for Rehabilitation, Dallas, TX, United States
| | - Caryn Harper
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Monica Bennett
- Department of Physical Medicine and Rehabilitation, Baylor Institute for Rehabilitation, Dallas, TX, United States
| | - Lisa Kreber
- Center for Neuro Skills, Bakersfield, CA, United States
| | - Richard Auchus
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
28
|
Neurite Growth and Polarization on Vitronectin Substrate after in Vitro Trauma is not Enhanced after IGF Treatment. Brain Sci 2018; 8:brainsci8080151. [PMID: 30103517 PMCID: PMC6119911 DOI: 10.3390/brainsci8080151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/31/2018] [Accepted: 08/08/2018] [Indexed: 11/23/2022] Open
Abstract
Following traumatic brain injuries (TBI), insulin-like growth factor (IGF) is cortically widely upregulated. This upregulation has a potential role in the recovery of neuronal tissue, plasticity, and neurotrophic activity, though the molecular mechanisms involved in IGF regulation and the exact role of IGF after TBI remain unclear. Vitronectin (VN), an extracellular matrix (ECM) molecule, has recently been shown to be of importance for IGF-mediated cellular growth and migration. Since VN is downregulated after TBI, we hypothesized that insufficient VN levels after TBI impairs the potential beneficial activity of IGF. To test if vitronectin and IGF-1/IGFBP-2 could contribute to neurite growth, we cultured hippocampal neurons on ± vitronectin-coated coverslips and them treated with ± IGF-1/IGF binding protein 2 (IGFBP-2). Under same conditions, cell cultures were also subjected to in vitro trauma to investigate differences in the posttraumatic regenerative capacity with ± vitronectin-coated coverslips and with ± IGF-1/IGFBP-2 treatment. In both the control and trauma situations, hippocampal neurons showed a stronger growth pattern on vitronectin than on the control substrate. Surprisingly, the addition of IGF-1/IGFBP-2 showed a decrease in neurite growth. Since neurite growth was measured as the number of neurites per area, we hypothesized that IGF-1/IGFBP-2 contributes to the polarization of neurons and thus induced a less dense neurite network after IGF-1/IGFBP-2 treatment. This hypothesis could not be confirmed and we therefore conclude that vitronectin has a positive effect on neurite growth in vitro both under normal conditions and after trauma, but that addition of IGF-1/IGFBP-2 does not have a positive additive effect.
Collapse
|
29
|
Carlson SW, Saatman KE. Central Infusion of Insulin-Like Growth Factor-1 Increases Hippocampal Neurogenesis and Improves Neurobehavioral Function after Traumatic Brain Injury. J Neurotrauma 2018; 35:1467-1480. [PMID: 29455576 PMCID: PMC5998830 DOI: 10.1089/neu.2017.5374] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Traumatic brain injury (TBI) produces neuronal dysfunction and cellular loss that can culminate in lasting impairments in cognitive and motor abilities. Therapeutic agents that promote repair and replenish neurons post-TBI hold promise in improving recovery of function. Insulin-like growth factor-1 (IGF-1) is a neurotrophic factor capable of mediating neuroprotective and neuroplasticity mechanisms. Targeted overexpression of IGF-1 enhances the generation of hippocampal newborn neurons in brain-injured mice; however, the translational neurogenic potential of exogenously administered IGF-1 post-TBI remains unknown. In a mouse model of controlled cortical impact, continuous intracerebroventricular infusion of recombinant human IGF-1 (hIGF) for 7 days, beginning 15 min post-injury, resulted in a dose-dependent increase in the number of immature neurons in the hippocampus. Infusion of 10 μg/day of IGF-1 produced detectable levels of hIGF-1 in the cortex and hippocampus and a concomitant increase in protein kinase B activation in the hippocampus. Both motor function and cognition were improved over 7 days post-injury in IGF-1-treated cohorts. Vehicle-treated brain-injured mice showed reduced hippocampal immature neuron density relative to sham controls at 7 days post-injury. In contrast, the density of hippocampal immature neurons in brain-injured mice receiving acute onset IGF-1 infusion was significantly higher than in injured mice receiving vehicle and equivalent to that in sham-injured control mice. Importantly, the neurogenic effect of IGF-1 was maintained with as much as a 6-h delay in the initiation of infusion. These data suggest that central infusion of IGF-1 enhances the generation of immature neurons in the hippocampus, with a therapeutic window of at least 6 h post-injury, and promotes neurobehavioral recovery post-TBI.
Collapse
Affiliation(s)
- Shaun W. Carlson
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Kathryn E. Saatman
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
30
|
Amtul Z, Hill DJ, Arany EJ, Cechetto DF. Altered Insulin/Insulin-Like Growth Factor Signaling in a Comorbid Rat model of Ischemia and β-Amyloid Toxicity. Sci Rep 2018; 8:5136. [PMID: 29572520 PMCID: PMC5865153 DOI: 10.1038/s41598-018-22985-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 02/06/2018] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke and diabetes are vascular risk factors for the development of impaired memory such as dementia and/or Alzheimer's disease. Clinical studies have demonstrated that minor striatal ischemic lesions in combination with β-amyloid (Aβ) load are critical in generating cognitive deficits. These cognitive deficits are likely to be associated with impaired insulin signaling. In this study, we examined the histological presence of insulin-like growth factor-I (IGF-1) and insulin receptor substrate (IRS-1) in anatomically distinct brain circuits compared with morphological brain damage in a co-morbid rat model of striatal ischemia (ET1) and Aβ toxicity. The results demonstrated a rapid increase in the presence of IGF-1 and IRS-1 immunoreactive cells in Aβ + ET1 rats, mainly in the ipsilateral striatum and distant regions with synaptic links to the striatal lesion. These regions included subcortical white matter, motor cortex, thalamus, dentate gyrus, septohippocampal nucleus, periventricular region and horizontal diagonal band of Broca in the basal forebrain. The alteration in IGF-1 and IRS-1 presence induced by ET1 or Aβ rats alone was not severe enough to affect the entire brain circuit. Understanding the causal or etiologic interaction between insulin and IGF signaling and co-morbidity after ischemia and Aβ toxicity will help design more effective therapeutics.
Collapse
Affiliation(s)
- Zareen Amtul
- Department of Anatomy and Cell Biology, University of Western Ontario, London, N6A 5C1, Canada.
| | - David J Hill
- Departments of Medicine, Physiology and Pharmacology, and Pediatrics, University of Western Ontario, London, N6A 5C1, Canada
- Lawson Health Research Institute, London, Ontario, N6A 4V2, Canada
| | - Edith J Arany
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, N6A 5C1, Canada
| | - David F Cechetto
- Department of Anatomy and Cell Biology, University of Western Ontario, London, N6A 5C1, Canada
| |
Collapse
|
31
|
PIP3-binding proteins promote age-dependent protein aggregation and limit survival in C. elegans. Oncotarget 2018; 7:48870-48886. [PMID: 27429199 PMCID: PMC5226477 DOI: 10.18632/oncotarget.10549] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 06/20/2016] [Indexed: 12/17/2022] Open
Abstract
Class-I phosphatidylinositol 3-kinase (PI3KI) converts phosphatidylinositol 4,5-bisphosphate (PIP2) to phosphatidylinositol 3,4,5-triphosphate (PIP3). PIP3 comprises two fatty-acid chains that embed in lipid-bilayer membranes, joined by glycerol to inositol triphosphate. Proteins with domains that specifically bind that head-group (e.g. pleckstrin-homology [PH] domains) are thus tethered to the inner plasma-membrane surface where they have an enhanced likelihood of interaction with other PIP3-bound proteins, in particular other components of their signaling pathways. Null alleles of the C. elegans age-1 gene, encoding the catalytic subunit of PI3KI, lack any detectable class-I PI3K activity and so cannot form PIP3. These mutant worms survive almost 10-fold longer than the longest-lived normal control, and are highly resistant to a variety of stresses including oxidative and electrophilic challenges. Traits associated with age-1 mutation are widely believed to be mediated through AKT-1, which requires PIP3 for both tethering and activation. Active AKT complex phosphorylates and thereby inactivates the DAF-16/FOXO transcription factor. However, extensive evidence indicates that pleiotropic effects of age-1-null mutations, including extreme longevity, cannot be explained by insulin like-receptor/AKT/FOXO signaling alone, suggesting involvement of other PIP3-binding proteins. We used ligand-affinity capture to identify membrane-bound proteins downstream of PI3KI that preferentially bind PIP3. Computer modeling supports a subset of candidate proteins predicted to directly bind PIP3 in preference to PIP2, and functional testing by RNAi knockdown confirmed candidates that partially mediate the stress-survival, aggregation-reducing and longevity benefits of PI3KI disruption. PIP3-specific candidate sets are highly enriched for proteins previously reported to affect translation, stress responses, lifespan, proteostasis, and lipid transport.
Collapse
|
32
|
Alterations in brain-derived neurotrophic factor and insulin-like growth factor-1 protein levels after penetrating ballistic-like brain injury in rats. J Trauma Acute Care Surg 2017. [PMID: 28628600 DOI: 10.1097/ta.0000000000001471] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) and insulin-like growth factor-1 (IGF-1) are essential for neuroplasticity and neuronal survival. Despite the importance of these endogenous factors in mediating posttraumatic recovery, little is known about their response after penetrating type traumatic brain injury. The objective of this study was to quantify the expression levels BDNF and IGF-1, two well-known neuroplasticity mediators, after penetrating ballistic-like brain injury (PBBI). METHODS Rats were randomly assigned to receive unilateral sham or PBBI injuries. Using enzyme-linked immunosorbent assay and immunohistochemistry, we performed a comprehensive evaluation of BDNF and IGF-1 expression at acute (1 hour, 6 hours, 1 day) and subacute (2, 3, 7, and 14 days) timepoints after injury. RESULTS BDNF and IGF-1 expression was transiently upregulated in both cortex and hippocampus after PBBI. Although BDNF levels increased at acute timepoints, IGF-1 expression peaked at 3 days in cortical homogenates. Although there was loss of staining in cells bordering the cavity, increased BDNF and IGF-1 immunoreactivity was observed in scattered neurons away from the contusion site. Glial upregulation of both growth factors was observed at early timepoints in the hippocampus. CONCLUSION Our findings demonstrate that PBBI results in a brief upregulation of BDNF and IGF-1 during early posttraumatic period, providing critical information for interventions aiming to enhance neuronal survival and brain plasticity.
Collapse
|
33
|
The Effects of Voluntary Physical Exercise-Activated Neurotrophic Signaling in Rat Hippocampus on mRNA Levels of Downstream Signaling Molecules. J Mol Neurosci 2017; 62:142-153. [DOI: 10.1007/s12031-017-0918-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/05/2017] [Indexed: 12/13/2022]
|
34
|
Mychasiuk R, Hehar H, Ma I, Candy S, Esser MJ. Reducing the time interval between concussion and voluntary exercise restores motor impairment, short-term memory, and alterations to gene expression. Eur J Neurosci 2016; 44:2407-2417. [DOI: 10.1111/ejn.13360] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 07/26/2016] [Accepted: 07/27/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Richelle Mychasiuk
- Alberta Children's Hospital Research Institute; Department of Psychology; University of Calgary; Heritage Medical Research Building, 3330 Hospital Drive NW Calgary AB T2N 1N4 Canada
| | - Harleen Hehar
- Alberta Children's Hospital Research Institute; Department of Psychology; University of Calgary; Heritage Medical Research Building, 3330 Hospital Drive NW Calgary AB T2N 1N4 Canada
| | - Irene Ma
- Alberta Children's Hospital Research Institute; Department of Psychology; University of Calgary; Heritage Medical Research Building, 3330 Hospital Drive NW Calgary AB T2N 1N4 Canada
| | - Sydney Candy
- Alberta Children's Hospital Research Institute; Faculty of Medicine; University of Calgary; Calgary AB Canada
| | - Michael J. Esser
- Alberta Children's Hospital Research Institute; Faculty of Medicine; University of Calgary; Calgary AB Canada
| |
Collapse
|
35
|
Song Y, Pimentel C, Walters K, Boller L, Ghiasvand S, Liu J, Staley KJ, Berdichevsky Y. Neuroprotective levels of IGF-1 exacerbate epileptogenesis after brain injury. Sci Rep 2016; 6:32095. [PMID: 27561791 PMCID: PMC4999804 DOI: 10.1038/srep32095] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 08/02/2016] [Indexed: 02/06/2023] Open
Abstract
Exogenous Insulin-Like Growth Factor-1 (IGF-1) is neuroprotective in animal models of brain injury, and has been considered as a potential therapeutic. Akt-mTOR and MAPK are downstream targets of IGF-1 signaling that are activated after brain injury. However, both brain injury and mTOR are linked to epilepsy, raising the possibility that IGF-1 may be epileptogenic. Here, we considered the role of IGF-1 in development of epilepsy after brain injury, using the organotypic hippocampal culture model of post-traumatic epileptogenesis. We found that IGF-1 was neuroprotective within a few days of injury but that long-term IGF-1 treatment was pro-epileptic. Pro-epileptic effects of IGF-1 were mediated by Akt-mTOR signaling. We also found that IGF-1 - mediated increase in epileptic activity led to neurotoxicity. The dualistic nature of effects of IGF-1 treatment demonstrates that anabolic enhancement through IGF-1 activation of mTOR cascade can be beneficial or harmful depending on the stage of the disease. Our findings suggest that epilepsy risk may need to be considered in the design of neuroprotective treatments for brain injury.
Collapse
Affiliation(s)
- Yu Song
- Bioengineering Program, Lehigh University, Bethlehem, PA 18015, USA
| | - Corrin Pimentel
- Bioengineering Program, Lehigh University, Bethlehem, PA 18015, USA
| | - Katherine Walters
- Integrated Degree in Engineering, Arts, and Sciences (IDEAS) Program, Lehigh University, PA 18015, USA
| | - Lauren Boller
- Bioengineering Program, Lehigh University, Bethlehem, PA 18015, USA
| | | | - Jing Liu
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, PA 18015, USA
| | - Kevin J Staley
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA.,Harvard Medical School, Boston, MA 02129, USA
| | - Yevgeny Berdichevsky
- Bioengineering Program, Lehigh University, Bethlehem, PA 18015, USA.,Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, PA 18015, USA
| |
Collapse
|
36
|
Wong VS, Langley B. Epigenetic changes following traumatic brain injury and their implications for outcome, recovery and therapy. Neurosci Lett 2016; 625:26-33. [PMID: 27155457 PMCID: PMC4915732 DOI: 10.1016/j.neulet.2016.04.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/03/2016] [Accepted: 04/05/2016] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) contributes to nearly a third of all injury-related deaths in the United States. For survivors of TBI, depending on severity, patients can be left with devastating neurological disabilities that include impaired cognition or memory, movement, sensation, or emotional function. Despite the efforts to identify novel therapeutics, the only strategy to combat TBI is risk reduction (helmets, seatbelts, removal of fall hazards, etc.). Enormous heterogeneity exists within TBI, and it depends on the severity, the location, and whether the injury was focal or diffuse. Evidence from recent studies support the involvement of epigenetic mechanisms such as DNA methylation, chromatin post-translational modification, and miRNA regulation of gene expression in the post-injured brain. In this review, we discuss studies that have assessed epigenetic changes and mechanisms following TBI, how epigenetic changes might not only be limited to the nucleus but also impact the mitochondria, and the implications of these changes with regard to TBI recovery.
Collapse
Affiliation(s)
- Victor S Wong
- Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, United States
| | - Brett Langley
- Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, United States; Department of Neurology and Neuroscience, Weill Medical College of Cornell University, 525 E. 68th Street, New York, NY 10065, United States.
| |
Collapse
|
37
|
Prediction of Post-Concussive Behavioral Changes in a Rodent Model Based on Head Rotational Acceleration Characteristics. Ann Biomed Eng 2016; 44:3252-3265. [PMID: 27188340 PMCID: PMC5093216 DOI: 10.1007/s10439-016-1647-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/11/2016] [Indexed: 02/07/2023]
Abstract
Quantifying injury tolerance for concussion is complicated by variability in the type, severity, and time course of post-injury physiological and behavioral changes. The current study outlined acute and chronic changes in behavioral metrics following rotational acceleration-induced concussion in rats. The Medical College of Wisconsin (MCW) rotational injury model independently controlled magnitude and duration of the rotational acceleration pulse. Increasing rotational acceleration magnitude produced longer recovery times, which were used in this study and our prior work as an assessment of acute injury severity. However, longer duration rotational accelerations produced changes in emotionality as measured using the elevated plus maze. Cognitive deficits were for the most part not apparent in the Morris water maze assessment, possibly due to the lower severity of rotational acceleration pulses incorporated in this study. Changes in emotionality evolved between acute and chronic assessments, in some cases increasing in severity and in others reversing polarity. These findings highlight the complexity of quantifying injury tolerance for concussion and demonstrate a need to incorporate rotational acceleration magnitude and duration in proposed injury tolerance metrics. Rotational velocity on its own was not a strong predictor of the magnitude or type of acute behavioral changes following concussion, although its combination with rotational acceleration magnitude using multivariate analysis was the strongest predictor for acute recovery time and some chronic emotional-type behavioral changes.
Collapse
|
38
|
Kline AE, Leary JB, Radabaugh HL, Cheng JP, Bondi CO. Combination therapies for neurobehavioral and cognitive recovery after experimental traumatic brain injury: Is more better? Prog Neurobiol 2016; 142:45-67. [PMID: 27166858 DOI: 10.1016/j.pneurobio.2016.05.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 04/26/2016] [Accepted: 05/01/2016] [Indexed: 12/18/2022]
Abstract
Traumatic brain injury (TBI) is a significant health care crisis that affects two million individuals in the United Sates alone and over ten million worldwide each year. While numerous monotherapies have been evaluated and shown to be beneficial at the bench, similar results have not translated to the clinic. One reason for the lack of successful translation may be due to the fact that TBI is a heterogeneous disease that affects multiple mechanisms, thus requiring a therapeutic approach that can act on complementary, rather than single, targets. Hence, the use of combination therapies (i.e., polytherapy) has emerged as a viable approach. Stringent criteria, such as verification of each individual treatment plus the combination, a focus on behavioral outcome, and post-injury vs. pre-injury treatments, were employed to determine which studies were appropriate for review. The selection process resulted in 37 papers that fit the specifications. The review, which is the first to comprehensively assess the effects of combination therapies on behavioral outcomes after TBI, encompasses five broad categories (inflammation, oxidative stress, neurotransmitter dysregulation, neurotrophins, and stem cells, with and without rehabilitative therapies). Overall, the findings suggest that combination therapies can be more beneficial than monotherapies as indicated by 46% of the studies exhibiting an additive or synergistic positive effect versus on 19% reporting a negative interaction. These encouraging findings serve as an impetus for continued combination studies after TBI and ultimately for the development of successful clinically relevant therapies.
Collapse
Affiliation(s)
- Anthony E Kline
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, United States; Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213, United States, United States; Psychology, University of Pittsburgh, Pittsburgh, PA 15213, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, United States; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15213, United States.
| | - Jacob B Leary
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Hannah L Radabaugh
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Jeffrey P Cheng
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Corina O Bondi
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, United States; Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, United States
| |
Collapse
|
39
|
Stemper BD, Shah AS, Budde MD, Olsen CM, Glavaski-Joksimovic A, Kurpad SN, McCrea M, Pintar FA. Behavioral Outcomes Differ between Rotational Acceleration and Blast Mechanisms of Mild Traumatic Brain Injury. Front Neurol 2016; 7:31. [PMID: 27014184 PMCID: PMC4789366 DOI: 10.3389/fneur.2016.00031] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 02/29/2016] [Indexed: 11/20/2022] Open
Abstract
Mild traumatic brain injury (mTBI) can result from a number of mechanisms, including blunt impact, head rotational acceleration, exposure to blast, and penetration of projectiles. Mechanism is likely to influence the type, severity, and chronicity of outcomes. The objective of this study was to determine differences in the severity and time course of behavioral outcomes following blast and rotational mTBI. The Medical College of Wisconsin (MCW) Rotational Injury model and a shock tube model of primary blast injury were used to induce mTBI in rats and behavioral assessments were conducted within the first week, as well as 30 and 60 days following injury. Acute recovery time demonstrated similar increases over protocol-matched shams, indicating acute injury severity equivalence between the two mechanisms. Post-injury behavior in the elevated plus maze demonstrated differing trends, with rotationally injured rats acutely demonstrating greater activity, whereas blast-injured rats had decreased activity that developed at chronic time points. Similarly, blast-injured rats demonstrated trends associated with cognitive deficits that were not apparent following rotational injuries. These findings demonstrate that rotational and blast injury result in behavioral changes with different qualitative and temporal manifestations. Whereas rotational injury was characterized by a rapidly emerging phenotype consistent with behavioral disinhibition, blast injury was associated with emotional and cognitive differences that were not evident acutely, but developed later, with an anxiety-like phenotype still present in injured animals at our most chronic measurements.
Collapse
Affiliation(s)
- Brian D. Stemper
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alok S. Shah
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Matthew D. Budde
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Christopher M. Olsen
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Shekar N. Kurpad
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael McCrea
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Frank A. Pintar
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
| |
Collapse
|
40
|
Karelina K, Sarac B, Freeman LM, Gaier KR, Weil ZM. Traumatic brain injury and obesity induce persistent central insulin resistance. Eur J Neurosci 2016; 43:1034-43. [DOI: 10.1111/ejn.13194] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/30/2015] [Accepted: 01/21/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Kate Karelina
- Department of Neuroscience; Center for Brain and Spinal Cord Repair and Group in Behavioral Neuroendocrinology; The Ohio State University Wexner Medical Center; Biomedical Research Tower #618 460 West 12th Avenue Columbus OH 43210 USA
| | - Benjamin Sarac
- Department of Neuroscience; Center for Brain and Spinal Cord Repair and Group in Behavioral Neuroendocrinology; The Ohio State University Wexner Medical Center; Biomedical Research Tower #618 460 West 12th Avenue Columbus OH 43210 USA
| | - Lindsey M. Freeman
- Department of Neuroscience; Center for Brain and Spinal Cord Repair and Group in Behavioral Neuroendocrinology; The Ohio State University Wexner Medical Center; Biomedical Research Tower #618 460 West 12th Avenue Columbus OH 43210 USA
| | - Kristopher R. Gaier
- Department of Neuroscience; Center for Brain and Spinal Cord Repair and Group in Behavioral Neuroendocrinology; The Ohio State University Wexner Medical Center; Biomedical Research Tower #618 460 West 12th Avenue Columbus OH 43210 USA
| | - Zachary M. Weil
- Department of Neuroscience; Center for Brain and Spinal Cord Repair and Group in Behavioral Neuroendocrinology; The Ohio State University Wexner Medical Center; Biomedical Research Tower #618 460 West 12th Avenue Columbus OH 43210 USA
| |
Collapse
|
41
|
Inácio AR, Liu Y, Clausen BH, Svensson M, Kucharz K, Yang Y, Stankovich T, Khorooshi R, Lambertsen KL, Issazadeh-Navikas S, Deierborg T. Endogenous IFN-β signaling exerts anti-inflammatory actions in experimentally induced focal cerebral ischemia. J Neuroinflammation 2015; 12:211. [PMID: 26581581 PMCID: PMC4652356 DOI: 10.1186/s12974-015-0427-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 11/05/2015] [Indexed: 12/21/2022] Open
Abstract
Background Interferon (IFN)-β exerts anti-inflammatory effects, coupled to remarkable neurological improvements in multiple sclerosis, a neuroinflammatory condition of the central nervous system. Analogously, it has been hypothesized that IFN-β, by limiting inflammation, decreases neuronal death and promotes functional recovery after stroke. However, the core actions of endogenous IFN-β signaling in stroke are unclear. Methods To address this question, we used two clinically relevant models of focal cerebral ischemia, transient and permanent middle cerebral artery occlusion, and two genetically modified mouse lines, lacking either IFN-β or its receptor, the IFN-α/β receptor. Subsets of inflammatory and immune cells isolated from the brain, blood, and spleen were studied using flow cytometry. Sensorimotor deficits were assessed by a modified composite neuroscore, the rotating pole and grip strength tests, and cerebral infarct volumes were given by lack of neuronal nuclei immunoreactivity. Results Here, we report alterations in local and systemic inflammation in IFN-β knockout (IFN-βKO) mice over 8 days after induction of focal cerebral ischemia. Notably, IFN-βKO mice showed a higher number of infiltrating leukocytes in the brain 2 days after stroke. Concomitantly, in the blood of IFN-βKO mice, we found a higher percentage of total B cells but a similar percentage of mature and activated B cells, collectively indicating a higher proliferation rate. The additional differential regulation of circulating cytokines and splenic immune cell populations in wild-type and IFN-βKO mice further supports an important immunoregulatory function of IFN-β in stroke. Moreover, we observed a significant weight loss 2–3 days and a reduction in grip strength 2 days after stroke in the IFN-βKO group, while endogenous IFN-β signaling did not affect the infarct volume. Conclusions We conclude that endogenous IFN-β signaling attenuates local inflammation, regulates peripheral immune cells, and, thereby, may contribute positively to stroke outcome. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0427-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ana R Inácio
- Laboratory for Experimental Brain Research, Department of Clinical Sciences, Lund University, BMC A13, Sölvegatan 17, 22184, Lund, Sweden. .,Present Address: INMED, INSERM U901, Parc Scientifique de Luminy, 163 route de Luminy, BP13, 13273, Marseille cedex 09, France. .,Present Address: Aix-Marseille Université, UMR S901, 13009, Marseille, France.
| | - Yawei Liu
- Neuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
| | - Bettina H Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, JB Winsloewsvej 21, st + 25, 2, 5000, Odense C, Denmark
| | - Martina Svensson
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Lund University, BMC B11, Sölvegatan 19, 22184, Lund, Sweden
| | - Krzysztof Kucharz
- Laboratory for Experimental Brain Research, Department of Clinical Sciences, Lund University, BMC A13, Sölvegatan 17, 22184, Lund, Sweden.,7Present Address: Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, 2200, Denmark
| | - Yiyi Yang
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Lund University, BMC B11, Sölvegatan 19, 22184, Lund, Sweden
| | - Totte Stankovich
- Laboratory for Experimental Brain Research, Department of Clinical Sciences, Lund University, BMC A13, Sölvegatan 17, 22184, Lund, Sweden
| | - Reza Khorooshi
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, JB Winsloewsvej 21, st + 25, 2, 5000, Odense C, Denmark
| | - Kate L Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, JB Winsloewsvej 21, st + 25, 2, 5000, Odense C, Denmark
| | - Shohreh Issazadeh-Navikas
- Neuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark.
| | - Tomas Deierborg
- Laboratory for Experimental Brain Research, Department of Clinical Sciences, Lund University, BMC A13, Sölvegatan 17, 22184, Lund, Sweden.,Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Lund University, BMC B11, Sölvegatan 19, 22184, Lund, Sweden
| |
Collapse
|
42
|
Effects of Intermittent Fasting, Caloric Restriction, and Ramadan Intermittent Fasting on Cognitive Performance at Rest and During Exercise in Adults. Sports Med 2015; 46:35-47. [DOI: 10.1007/s40279-015-0408-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
43
|
Karelina K, Weil ZM. Neuroenergetics of traumatic brain injury. ACTA ACUST UNITED AC 2015; 1:CNC9. [PMID: 30202553 PMCID: PMC6114023 DOI: 10.2217/cnc.15.9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/13/2015] [Indexed: 01/21/2023]
Abstract
A subset of traumatic brain injury (TBI) patients exhibit cognitive deficits later in life which may be due to the underlying pathology associated with Alzheimer's disease (AD) or chronic traumatic encephalopathy. The similarities between chronic traumatic encephalopathy and AD merit investigation of potentially similar mechanisms underlying the two diseases. Experimental and clinical studies of AD brains have revealed that insulin resistance links metabolic dysfunction to the neurodegeneration and cognitive deficits associated with AD. Recent work in experimental TBI has established that recovery is dependent on the return of normal brain metabolism and mounting evidence for a role of brain insulin in regulating central metabolism suggests that TBI, like AD, results in central insulin resistance. Here, we review the converging evidence from AD, TBI and diabetes research linking insulin insensitivity to neurodegeneration.
Collapse
Affiliation(s)
- Kate Karelina
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus OH, USA
| | - Zachary M Weil
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus OH, USA
| |
Collapse
|
44
|
Role and Importance of IGF-1 in Traumatic Brain Injuries. BIOMED RESEARCH INTERNATIONAL 2015; 2015:736104. [PMID: 26417600 PMCID: PMC4568328 DOI: 10.1155/2015/736104] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 12/24/2014] [Indexed: 01/02/2023]
Abstract
It is increasingly affirmed that most of the long-term consequences of TBI are due to molecular and cellular changes occurring during the acute phase of the injury and which may, afterwards, persist or progress. Understanding how to prevent secondary damage and improve outcome in trauma patients, has been always a target of scientific interest. Plans of studies focused their attention on the posttraumatic neuroendocrine dysfunction in order to achieve a correlation between hormone blood level and TBI outcomes. The somatotropic axis (GH and IGF-1) seems to be the most affected, with different alterations between the acute and late phases. IGF-1 plays an important role in brain growth and development, and it is related to repair responses to damage for both the central and peripheral nervous system. The IGF-1 blood levels result prone to decrease during both the early and late phases after TBI. Despite this, experimental studies on animals have shown that the CNS responds to the injury upregulating the expression of IGF-1; thus it appears to be related to the secondary mechanisms of response to posttraumatic damage. We review the mechanisms involving IGF-1 in TBI, analyzing how its expression and metabolism may affect prognosis and outcome in head trauma patients.
Collapse
|
45
|
Guan J, Harris P, Brimble M, Lei Y, Lu J, Yang Y, Gunn AJ. The role for IGF-1-derived small neuropeptides as a therapeutic target for neurological disorders. Expert Opin Ther Targets 2015; 19:785-93. [PMID: 25652713 DOI: 10.1517/14728222.2015.1010514] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Exogenous IGF-1 protects the brain from ischemic injury and improves function. However, its clinical application to neurological disorders is limited by its large molecular size, poor central uptake and mitogenic potential. AREAS COVERED In this review, the authors have discussed the efficacy, pharmacokinetics and mechanisms of IGF-1 derivatives on protecting acute brain injury, preventing memory impairment and improving recovery from neurological degenerative conditions evaluated in various animal models. We have included natural metabolites of IGF-1, glycine-proline-glutamate (GPE), cleaved from N-terminal IGF-1 and cyclic glycine-proline (cGP) as well as the structural analogues of GPE and cGP, glycine-2-methyl-proline-glutamate and cyclo-l-glycyl-l-2-allylproline, respectively. In addition, the regulatory role for cGP in bioavailability of IGF-1 has also been discussed. EXPERT OPINION These small neuropeptides provide effective neuroprotection by offering an improved pharmacokinetic profile and more practical route of administration compared with IGF-1 administration. Developing modified neuropeptides to overcome the limitations of their endogenous counterparts represents a novel strategy of pharmaceutical discovery for neurological disorders. The mechanism of action may involve a regulation of IGF-1 bioavailability.
Collapse
Affiliation(s)
- Jian Guan
- University of Auckland, Liggins Institute , Private Bag 92019, Auckland , New Zealand +64 93 737 599 ext. 86134 ; +64 93 082 385 ;
| | | | | | | | | | | | | |
Collapse
|
46
|
Rubovitch V, Barak S, Rachmany L, Goldstein RB, Zilberstein Y, Pick CG. The Neuroprotective Effect of Salubrinal in a Mouse Model of Traumatic Brain Injury. Neuromolecular Med 2015; 17:58-70. [DOI: 10.1007/s12017-015-8340-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 01/06/2015] [Indexed: 12/15/2022]
|
47
|
Stemper BD, Shah AS, Pintar FA, McCrea M, Kurpad SN, Glavaski-Joksimovic A, Olsen C, Budde MD. Head rotational acceleration characteristics influence behavioral and diffusion tensor imaging outcomes following concussion. Ann Biomed Eng 2014; 43:1071-88. [PMID: 25344352 DOI: 10.1007/s10439-014-1171-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 10/17/2014] [Indexed: 11/28/2022]
Abstract
A majority of traumatic brain injuries (TBI) in motor vehicle crashes and sporting environments are mild and caused by high-rate acceleration of the head. For injuries caused by rotational acceleration, both magnitude and duration of the acceleration pulse were shown to influence injury outcomes. This study incorporated a unique rodent model of rotational acceleration-induced mild TBI (mTBI) to quantify independent effects of magnitude and duration on behavioral and neuroimaging outcomes. Ninety-two Sprague-Dawley rats were exposed to head rotational acceleration at peak magnitudes of 214 or 350 krad/s(2) and acceleration pulse durations of 1.6 or 3.4 ms in a full factorial design. Rats underwent a series of behavioral tests including the Composite Neuroscore (CN), Elevated Plus Maze (EPM), and Morris Water Maze (MWM). Ex vivo diffusion tensor imaging (DTI) of the fixed brains was conducted to assess the effects of rotational injury on brain microstructure as revealed by the parameter fractional anisotropy (FA). While the injury did not cause significant locomotor or cognitive deficits measured with the CN and MWM, respectively, a main effect of duration was consistently observed for the EPM. Increased duration caused significantly greater activity and exploratory behaviors measured as open arm time and number of arm changes. DTI demonstrated significant effects of both magnitude and duration, with the FA of the amygdala related to both the magnitude and duration. Increased duration also caused FA changes at the interface of gray and white matter. Collectively, the findings demonstrate that the consequences of rotational acceleration mTBI were more closely associated with duration of the rotational acceleration impulse, which is often neglected as an independent factor, and highlight the need for animal models of TBI with strong biomechanical foundations to associate behavioral outcomes with brain microstructure.
Collapse
Affiliation(s)
- Brian D Stemper
- Department of Neurosurgery, Medical College of Wisconsin, Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA,
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Diaz-Arrastia R, Kochanek PM, Bergold P, Kenney K, Marx CE, Grimes CJB, Loh LTCY, Adam LTCGE, Oskvig D, Curley KC, Salzer W. Pharmacotherapy of traumatic brain injury: state of the science and the road forward: report of the Department of Defense Neurotrauma Pharmacology Workgroup. J Neurotrauma 2014; 31:135-58. [PMID: 23968241 DOI: 10.1089/neu.2013.3019] [Citation(s) in RCA: 192] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Despite substantial investments by government, philanthropic, and commercial sources over the past several decades, traumatic brain injury (TBI) remains an unmet medical need and a major source of disability and mortality in both developed and developing societies. The U.S. Department of Defense neurotrauma research portfolio contains more than 500 research projects funded at more than $700 million and is aimed at developing interventions that mitigate the effects of trauma to the nervous system and lead to improved quality of life outcomes. A key area of this portfolio focuses on the need for effective pharmacological approaches for treating patients with TBI and its associated symptoms. The Neurotrauma Pharmacology Workgroup was established by the U.S. Army Medical Research and Materiel Command (USAMRMC) with the overarching goal of providing a strategic research plan for developing pharmacological treatments that improve clinical outcomes after TBI. To inform this plan, the Workgroup (a) assessed the current state of the science and ongoing research and (b) identified research gaps to inform future development of research priorities for the neurotrauma research portfolio. The Workgroup identified the six most critical research priority areas in the field of pharmacological treatment for persons with TBI. The priority areas represent parallel efforts needed to advance clinical care; each requires independent effort and sufficient investment. These priority areas will help the USAMRMC and other funding agencies strategically guide their research portfolios to ensure the development of effective pharmacological approaches for treating patients with TBI.
Collapse
Affiliation(s)
- Ramon Diaz-Arrastia
- 1 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hertze J, Nägga K, Minthon L, Hansson O. Changes in cerebrospinal fluid and blood plasma levels of IGF-II and its binding proteins in Alzheimer's disease: an observational study. BMC Neurol 2014; 14:64. [PMID: 24685003 PMCID: PMC3973836 DOI: 10.1186/1471-2377-14-64] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 03/28/2014] [Indexed: 12/17/2022] Open
Abstract
Background The Insulin-like Growth Factor (IGF)-related system is implicated in neuroregeneration and cell repair, as well as regulating lifespan. IGF-II, one component of this system, has also been found to affect memory functions in a rat model. In this study we explored changes in the IGF-related system in patients with Alzheimer’s disease (AD), including changes in IGF-II levels. Methods We measured blood plasma and cerebrospinal fluid (CSF) levels of IGF-I, IGF-II, IGFBP-2 and IGFBP-3 in 72 healthy controls and 92 patients with AD. Results We found significantly lower blood plasma levels of IGF-II and IGFBP-3 in patients with AD, compared with controls. The levels of IGF-II and IGFBP-2 were significantly elevated in the CSF from patients with AD. We also found correlations between established CSF biomarkers for AD (tau and P-tau) and components of the IGF system. Conclusions CSF and blood plasma levels of IGF-II and some of its binding proteins are changed in patients with AD. Further investigation into this area may unravel important clues to the nature of this disease.
Collapse
Affiliation(s)
- Joakim Hertze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.
| | | | | | | |
Collapse
|
50
|
Gold EM, Su D, López-Velázquez L, Haus DL, Perez H, Lacuesta GA, Anderson AJ, Cummings BJ. Functional assessment of long-term deficits in rodent models of traumatic brain injury. Regen Med 2014; 8:483-516. [PMID: 23826701 DOI: 10.2217/rme.13.41] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Traumatic brain injury (TBI) ranks as the leading cause of mortality and disability in the young population worldwide. The annual US incidence of TBI in the general population is estimated at 1.7 million per year, with an estimated financial burden in excess of US$75 billion a year in the USA alone. Despite the prevalence and cost of TBI to individuals and society, no treatments have passed clinical trial to clinical implementation. The rapid expansion of stem cell research and technology offers an alternative to traditional pharmacological approaches targeting acute neuroprotection. However, preclinical testing of these approaches depends on the selection and characterization of appropriate animal models. In this article we consider the underlying pathophysiology for the focal and diffuse TBI subtypes, discuss the existing preclinical TBI models and functional outcome tasks used for assessment of injury and recovery, identify criteria particular to preclinical animal models of TBI in which stem cell therapies can be tested for safety and efficacy, and review these criteria in the context of the existing TBI literature. We suggest that 2 months post-TBI is the minimum period needed to evaluate human cell transplant efficacy and safety. Comprehensive review of the published TBI literature revealed that only 32% of rodent TBI papers evaluated functional outcome ≥1 month post-TBI, and only 10% evaluated functional outcomes ≥2 months post-TBI. Not all published papers that evaluated functional deficits at a minimum of 2 months post-TBI reported deficits; hence, only 8.6% of overall TBI papers captured in this review demonstrated functional deficits at 2 months or more postinjury. A 2-month survival and assessment period would allow sufficient time for differentiation and integration of human neural stem cells with the host. Critically, while trophic effects might be observed at earlier time points, it will also be important to demonstrate the sustainability of such an effect, supporting the importance of an extended period of in vivo observation. Furthermore, regulatory bodies will likely require at least 6 months survival post-transplantation for assessment of toxicology/safety, particularly in the context of assessing cell abnormalities.
Collapse
Affiliation(s)
- Eric M Gold
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine 2030 Gross Hall, CA 92697-1705, USA
| | | | | | | | | | | | | | | |
Collapse
|