1
|
Willis A, Jeong D, Liu Y, Lithopoulos MA, Yuzwa SA, Frankland PW, Kaplan DR, Miller FD. Single cell approaches define neural stem cell niches and identify microglial ligands that can enhance precursor-mediated oligodendrogenesis. Cell Rep 2025; 44:115194. [PMID: 39823226 DOI: 10.1016/j.celrep.2024.115194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/24/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025] Open
Abstract
Here, we used single cell RNA sequencing and single cell spatial transcriptomics to characterize the forebrain neural stem cell (NSC) niche under homeostatic and injury conditions. We defined the dorsal and lateral ventricular-subventricular zones (V-SVZs) as two distinct neighborhoods and showed that, after white matter injury, NSCs are activated to make oligodendrocytes dorsally for remyelination. This activation is coincident with an increase in transcriptionally distinct microglia in the dorsal V-SVZ niche. We modeled ligand-receptor interactions within this changing niche and identified two remyelination-associated microglial ligands, insulin growth factor 1 and oncostatin M, that promote precursor proliferation and oligodendrogenesis in culture. Infusion of either ligand into the lateral ventricles also enhanced oligodendrogenesis, even in the lateral V-SVZ, where NSCs normally make neuroblasts. These data support a model where gliogenesis versus neurogenesis is determined by the local NSC neighborhood and where injury-induced niche alterations promote NSC activation, local oligodendrogenesis, and likely contribute to myelin repair.
Collapse
Affiliation(s)
- Ashleigh Willis
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Danielle Jeong
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yunlong Liu
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Marissa A Lithopoulos
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Scott A Yuzwa
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Paul W Frankland
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - David R Kaplan
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Freda D Miller
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
2
|
Hayes CA, Wilson D, De Leon MA, Mustapha MJ, Morales S, Odden MC, Ashpole NM. Insulin-like growth factor-1 and cognitive health: Exploring cellular, preclinical, and clinical dimensions. Front Neuroendocrinol 2024; 76:101161. [PMID: 39536910 DOI: 10.1016/j.yfrne.2024.101161] [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: 07/31/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
Age and insulin-like growth factor-1 (IGF-1) have an inverse association with cognitive decline and dementia. IGF-1 is known to have important pleiotropic functions beginning in neurodevelopment and extending into adulthood such as neurogenesis. At the cellular level, IGF-1 has pleiotropic signaling mechanisms through the IGF-1 receptor on neurons and neuroglia to attenuate inflammation, promote myelination, maintain astrocytic functions for homeostatic balances, and neuronal synaptogenesis. In preclinical rodent models of aging and transgenic models of IGF-1, increased IGF-1 improves cognition in a variety of behavioral paradigms along with reducing IGF-1 via knockout models being able to induce cognitive impairment. At the clinical levels, most studies highlight that increased levels of IGF-1 are associated with better cognition. This review provides a comprehensive and up-to-date evaluation of the association between IGF-1 and cognition at the cellular signaling levels, preclinical, and clinical levels.
Collapse
Affiliation(s)
- Cellas A Hayes
- Department of Epidemiology and Population Health, School of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA.
| | - Destiny Wilson
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
| | - Miguel A De Leon
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
| | | | - Sharon Morales
- Department of Biomedical Science, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Michelle C Odden
- Department of Epidemiology and Population Health, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Nicole M Ashpole
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
| |
Collapse
|
3
|
VanRyzin JW, Marquardt AE, McCarthy MM. Feminization of social play behavior depends on microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608675. [PMID: 39229086 PMCID: PMC11370478 DOI: 10.1101/2024.08.19.608675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Many sex differences in brain and behavior are established developmentally by the opposing processes of feminization and masculinization, which manifest following differential steroid hormone exposure in early life. The cellular mechanisms underlying masculinization are well-documented, a result of the fact that it is steroid-mediated and can be easily induced in newborn female rodents via exogenous steroid treatment. However, the study of feminization of particular brain regions has largely been relegated to being "not masculinization" given the absence of an identified initiating trigger. As a result, the mechanisms of this key developmental process remain elusive. Here we describe a novel role for microglia, the brain's innate immune cell, in the feminization of the medial amygdala and a complex social behavior, juvenile play. In the developing amygdala, microglia promote proliferation of astrocytes equally in both sexes, with no apparent effect on rates of cell division, but support cell survival selectively in females through the trophic actions of Tumor Necrosis Factor α (TNFα). We demonstrate that disrupting TNFα signaling, either by depleting microglia or inhibiting the associated signaling pathways, prevents the feminization of astrocyte density and increases juvenile play levels to that seen in males. This data, combined with our previous finding that male-like patterns of astrocyte density are sculpted by developmental microglial phagocytosis, reveals that sexual differentiation of the medial amygdala involves opposing tensions between active masculinization and active feminization, both of which require microglia but are achieved via distinct processes.
Collapse
Affiliation(s)
- Jonathan W VanRyzin
- Department of Pharmacology, Physiology and Drug Development and University of Maryland Medicine – Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD 21201
| | - Ashley E Marquardt
- Department of Pharmacology, Physiology and Drug Development and University of Maryland Medicine – Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD 21201
| | - Margaret M McCarthy
- Department of Pharmacology, Physiology and Drug Development and University of Maryland Medicine – Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
4
|
Huang R, Shi J, Wei R, Li J. Challenges of insulin-like growth factor-1 testing. Crit Rev Clin Lab Sci 2024; 61:388-403. [PMID: 38323343 DOI: 10.1080/10408363.2024.2306804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/27/2023] [Accepted: 01/15/2024] [Indexed: 02/08/2024]
Abstract
Insulin-like growth factor 1 (IGF-1), primarily synthesized in the liver, was initially discovered due to its capacity to replicate the metabolic effects of insulin. Subsequently, it emerged as a key regulator of the actions of growth hormone (GH), managing critical processes like cell proliferation, differentiation, and apoptosis. Notably, IGF-1 displays a longer half-life compared to GH, making it less susceptible to factors that may affect GH concentrations. Consequently, the measurement of IGF-1 proves to be more specific and sensitive when diagnosing conditions such as acromegaly or GH deficiency. The recognition of the existence of IGFBPs and their potential to interfere with IGF-1 immunoassays urged the implementation of various techniques to moderate this issue and provide accurate IGF-1 results. Additionally, in response to the limitations associated with IGF-1 immunoassays and the occurrence of discordant IGF-1 results, modern mass spectrometric methods were developed to facilitate the quantification of IGF-1 levels. Taking advantage of their ability to minimize the interference caused by IGF-1 variants, mass spectrometric methods offer the capacity to deliver robust, reliable, and accurate IGF-1 results, relying on the precision of mass measurements. This also enables the potential detection of pathogenic mutations through protein sequence analysis. However, despite the analytical challenges, the discordance in IGF-1 reference intervals can be attributed to a multitude of factors, potentially leading to distinct interpretations of results. The establishment of reference intervals for each assay is a demanding task, and it requires nationwide multicenter collaboration among laboratorians, clinicians, and assay manufacturers to achieve this common goal in a cost-effective and resource-efficient manner. In this comprehensive review, we examine the challenges associated with the standardization of IGF-1 measurement methods, the minimization of pre-analytical factors, and the harmonization of reference intervals. Particular emphasis will be placed on the development of IGF-1 measurement techniques using "top-down" or "bottom-up" mass spectrometric methods.
Collapse
Affiliation(s)
- Rongrong Huang
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology and Laboratory Medicine, Harris Health System Ben Taub Hospital, Houston, TX, USA
| | - Junyan Shi
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver Coastal Health, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ruhan Wei
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Jieli Li
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
5
|
Shen J, Liu L, Yang Y, Zhou M, Xu S, Zhang W, Zhang C. Insulin-Like Growth Factor 1 Has the Potential to Be Used as a Diagnostic Tool and Treatment Target for Autism Spectrum Disorders. Cureus 2024; 16:e65393. [PMID: 39188438 PMCID: PMC11346671 DOI: 10.7759/cureus.65393] [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] [Accepted: 07/16/2024] [Indexed: 08/28/2024] Open
Abstract
Autism spectrum disorder (ASD), a heterogeneous group of neurodevelopmental disorders, is characterized by social impairment and repetitive and stereotypic behaviors. Because of the lack of approved laboratory diagnostic markers and effective therapeutic medications, it is one of the most challenging diseases. Therefore, it is urgent to explore potential diagnosis markers or therapeutic targets. Insulin-like growth factor 1 (IGF-1) is a neurotrophic growth factor that enhances brain development. IGF-1 levels in body fluids are lower in preschool children with ASD than in typically developing children, which may serve as a potential diagnostic marker. In various ASD models associated with genetic or environmental exposure, IGF-1 treatment can improve core symptoms or pathological changes, including neuronal development, neural cell survival, balance of synaptic excitation and inhibition, neuroimmunology, and oxidative stress status. In March 2023 an IGF-1 derivative was approved as the first drug for treating Rett syndrome, an ASD-related neurodevelopmental disorder, to improve fundamental symptoms such as social communication. Thus, in this review, we present accumulating evidence of altered IGF-1 levels in ASD patients and the possible mechanisms, as well as evidence that IGF-1 treatment improves the pathophysiology in various ASD models. IGF-1 has the potential to be an early diagnosis marker and an effective therapeutic for ASD.
Collapse
Affiliation(s)
- Jiamin Shen
- Department of Children Health Care, Jingmen Maternity and Child Health Care Hospital, Jingmen, CHN
| | - Lijuan Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, CHN
| | - Yifan Yang
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| | - Miao Zhou
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| | - Shan Xu
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| | - Wanqing Zhang
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| | - Chuanjie Zhang
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| |
Collapse
|
6
|
Punt LD, van der Kaay DCM, van Setten PA, de Groote K, Kruijsen AR, Bocca G, de Munnik SA, Renes JS, de Bruin C, Losekoot M, van Duyvenvoorde HA, Wit JM, Joustra SD. IGF1 Haploinsufficiency: Phenotype and Response to Growth Hormone Treatment in 9 Patients. Horm Res Paediatr 2024:1-11. [PMID: 38952118 DOI: 10.1159/000540053] [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: 04/12/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
INTRODUCTION The clinical features of bi-allelic IGF1 defects are well established, i.e., severe growth failure and microcephaly, delayed psychomotor development, and sensorineural deafness. However, information on clinical and endocrine consequences of heterozygous IGF1 variants and treatment options is scarce. We aimed at extending the knowledge base of the clinical presentation and growth response to recombinant human growth hormone (rhGH) of patients carrying such variants. METHODS Retrospective case series of patients with pathogenic heterozygous IGF1 variants. RESULTS Nine patients from six families were included, harbouring five whole or partial gene deletions and one frameshift variant resulting in a premature stop codon (three de novo, one unknown inheritance). In the other two families, variants segregated with short stature. Mean (SD) birth length was -1.9 (1.3) SDS (n = 7), height -3.8 (0.6) SDS, head circumference -2.5 (0.6) SDS, serum IGF-I -1.9 (0.7) SDS, serum IGFBP-3 1.1 (0.4) SDS (n = 7), and GH peak range 5-31 μg/L (n = 4). Five patients showed feeding problems in infancy. Average height increased after 1 and 2 years of rhGH treatment by 0.8 SDS (range 0.3-1.3 SDS) and 1.3 SDS (range 0.5-2.0 SDS), respectively. Adult height in 2 patients was -2.8 and -1.3 SDS, which was, respectively, 1.3 and 2.9 SDS taller than predicted before start of treatment. CONCLUSION Haploinsufficiency of IGF1 causes a variable phenotype of prenatal and postnatal growth failure, microcephaly, feeding difficulties, low/low-normal serum IGF-I values in contrast to serum IGFBP-3 in the upper-normal range. Treatment with rhGH increased growth in the first 2 years of treatment, and in 2 patients adult height after treatment was higher than predicted at treatment initiation.
Collapse
Affiliation(s)
- Lauren D Punt
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands
| | - Daniëlle C M van der Kaay
- Division of Paediatric Endocrinology, Department of Paediatrics, Erasmus University Medical Centre, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Petra A van Setten
- Department of Paediatrics, Amalia Childrens Hospital, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Kirsten de Groote
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands
| | - Anne R Kruijsen
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands
| | - Gianni Bocca
- Division of Paediatric Endocrinology, Department of Paediatrics, Beatrix Children's Hospital, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Sonja A de Munnik
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Judith S Renes
- Dutch Growth Research Foundation, Rotterdam, The Netherlands
- Department of Paediatrics, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Christiaan de Bruin
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands
| | - Monique Losekoot
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | | | - Jan M Wit
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands
| | - Sjoerd D Joustra
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
7
|
Emery B, Wood TL. Regulators of Oligodendrocyte Differentiation. Cold Spring Harb Perspect Biol 2024; 16:a041358. [PMID: 38503504 PMCID: PMC11146316 DOI: 10.1101/cshperspect.a041358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Myelination has evolved as a mechanism to ensure fast and efficient propagation of nerve impulses along axons. Within the central nervous system (CNS), myelination is carried out by highly specialized glial cells, oligodendrocytes. The formation of myelin is a prolonged aspect of CNS development that occurs well into adulthood in humans, continuing throughout life in response to injury or as a component of neuroplasticity. The timing of myelination is tightly tied to the generation of oligodendrocytes through the differentiation of their committed progenitors, oligodendrocyte precursor cells (OPCs), which reside throughout the developing and adult CNS. In this article, we summarize our current understanding of some of the signals and pathways that regulate the differentiation of OPCs, and thus the myelination of CNS axons.
Collapse
Affiliation(s)
- Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Teresa L Wood
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103, USA
| |
Collapse
|
8
|
Potenzieri A, Uccella S, Preiti D, Pisoni M, Rosati S, Lavarello C, Bartolucci M, Debellis D, Catalano F, Petretto A, Nobili L, Fellin T, Tucci V, Ramenghi LA, Savardi A, Cancedda L. Early IGF-1 receptor inhibition in mice mimics preterm human brain disorders and reveals a therapeutic target. SCIENCE ADVANCES 2024; 10:eadk8123. [PMID: 38427732 PMCID: PMC10906931 DOI: 10.1126/sciadv.adk8123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/29/2024] [Indexed: 03/03/2024]
Abstract
Besides recent advances in neonatal care, preterm newborns still develop sex-biased behavioral alterations. Preterms fail to receive placental insulin-like growth factor-1 (IGF-1), a major fetal growth hormone in utero, and low IGF-1 serum levels correlate with preterm poor neurodevelopmental outcomes. Here, we mimicked IGF-1 deficiency of preterm newborns in mice by perinatal administration of an IGF-1 receptor antagonist. This resulted in sex-biased brain microstructural, functional, and behavioral alterations, resembling those of ex-preterm children, which we characterized performing parallel mouse/human behavioral tests. Pharmacological enhancement of GABAergic tonic inhibition by the U.S. Food and Drug Administration-approved drug ganaxolone rescued functional/behavioral alterations in mice. Establishing an unprecedented mouse model of prematurity, our work dissects the mechanisms at the core of abnormal behaviors and identifies a readily translatable therapeutic strategy for preterm brain disorders.
Collapse
Affiliation(s)
- Alberto Potenzieri
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
- Università degli Studi di Genova, via Balbi, 5, 16126 Genoa, Italy
| | - Sara Uccella
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Deborah Preiti
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Matteo Pisoni
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Silvia Rosati
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Chiara Lavarello
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Martina Bartolucci
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Doriana Debellis
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Federico Catalano
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Lino Nobili
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Valter Tucci
- Genetics and Epigenetics of Behavior (GEB) Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Luca A. Ramenghi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Annalisa Savardi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| |
Collapse
|
9
|
You Y, Chen Z, Hu WW. The role of microglia heterogeneity in synaptic plasticity and brain disorders: Will sequencing shed light on the discovery of new therapeutic targets? Pharmacol Ther 2024; 255:108606. [PMID: 38346477 DOI: 10.1016/j.pharmthera.2024.108606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/05/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Microglia play a crucial role in interacting with neuronal synapses and modulating synaptic plasticity. This function is particularly significant during postnatal development, as microglia are responsible for removing excessive synapses to prevent neurodevelopmental deficits. Dysregulation of microglial synaptic function has been well-documented in various pathological conditions, notably Alzheimer's disease and multiple sclerosis. The recent application of RNA sequencing has provided a powerful and unbiased means to decipher spatial and temporal microglial heterogeneity. By identifying microglia with varying gene expression profiles, researchers have defined multiple subgroups of microglia associated with specific pathological states, including disease-associated microglia, interferon-responsive microglia, proliferating microglia, and inflamed microglia in multiple sclerosis, among others. However, the functional roles of these distinct subgroups remain inadequately characterized. This review aims to refine our current understanding of the potential roles of heterogeneous microglia in regulating synaptic plasticity and their implications for various brain disorders, drawing from recent sequencing research and functional studies. This knowledge may aid in the identification of pathogenetic biomarkers and potential factors contributing to pathogenesis, shedding new light on the discovery of novel drug targets. The field of sequencing-based data mining is evolving toward a multi-omics approach. With advances in viral tools for precise microglial regulation and the development of brain organoid models, we are poised to elucidate the functional roles of microglial subgroups detected through sequencing analysis, ultimately identifying valuable therapeutic targets.
Collapse
Affiliation(s)
- Yi You
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhong Chen
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wei-Wei Hu
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
10
|
Ng ACH, Choudhary A, Barrett KT, Gavrilovici C, Scantlebury MH. Mechanisms of infantile epileptic spasms syndrome: What have we learned from animal models? Epilepsia 2024; 65:266-280. [PMID: 38036453 DOI: 10.1111/epi.17841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023]
Abstract
The devastating developmental and epileptic encephalopathy of infantile epileptic spasms syndrome (IESS) has numerous causes, including, but not limited to, brain injury, metabolic, and genetic conditions. Given the stereotyped electrophysiologic, age-dependent, and clinical findings, there likely exists one or more final common pathways in the development of IESS. The identity of this final common pathway is unknown, but it may represent a novel therapeutic target for infantile spasms. Previous research on IESS has focused largely on identifying the neuroanatomic substrate using specialized neuroimaging techniques and cerebrospinal fluid analysis in human patients. Over the past three decades, several animal models of IESS were created with an aim to interrogate the underlying pathogenesis of IESS, to identify novel therapeutic targets, and to test various treatments. Each of these models have been successful at recapitulating multiple aspects of the human IESS condition. These animal models have implicated several different molecular pathways in the development of infantile spasms. In this review we outline the progress that has been made thus far using these animal models and discuss future directions to help researchers identify novel treatments for drug-resistant IESS.
Collapse
Affiliation(s)
- Andy Cheuk-Him Ng
- Department of Pediatrics, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Anamika Choudhary
- Department of Pediatrics, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Karlene T Barrett
- Department of Pediatrics, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Cezar Gavrilovici
- Department of Pediatrics, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Morris H Scantlebury
- Department of Pediatrics, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
11
|
Rusin D, Vahl Becirovic L, Lyszczarz G, Krueger M, Benmamar-Badel A, Vad Mathiesen C, Sigurðardóttir Schiöth E, Lykke Lambertsen K, Wlodarczyk A. Microglia-Derived Insulin-like Growth Factor 1 Is Critical for Neurodevelopment. Cells 2024; 13:184. [PMID: 38247874 PMCID: PMC10813844 DOI: 10.3390/cells13020184] [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: 12/09/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a peptide hormone essential for the proper development and growth of the organism, as a complete knockout of Igf1 in mice is lethal, causing microcephaly, growth retardation and the defective development of organs. In the central nervous system, neurons and glia have been reported to express Igf1, but their relative importance for postnatal development has not yet been fully defined. In order to address this, here, we obtained mice with a microglia-specific inducible conditional knockout of Igf1. We show that the deficiency in microglial Igf1, starting in the first postnatal week, leads to body and brain growth retardation, severely impaired myelination, changes in microglia numbers, and behavioral abnormalities. These results emphasize the importance of microglial-derived Igf1 for brain development and function and open new perspectives for the investigation of the role of microglial-Igf1 in neurological diseases.
Collapse
Affiliation(s)
- Dominika Rusin
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Lejla Vahl Becirovic
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Gabriela Lyszczarz
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Martin Krueger
- Institute for Anatomy, University of Leipzig, 04103 Leipzig, Germany
| | - Anouk Benmamar-Badel
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Cecilie Vad Mathiesen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Neuroscience Academy Denmark, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Eydís Sigurðardóttir Schiöth
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Department of Clinical Research, BRIDGE—Brain Research Interdisciplinary Guided Excellence, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Department of Neurology, Odense University Hospital, 5000 Odense C, Denmark
| | - Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Department of Clinical Research, BRIDGE—Brain Research Interdisciplinary Guided Excellence, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| |
Collapse
|
12
|
Horvath A, Quinlan P, Eckerström C, Åberg ND, Wallin A, Svensson J. The Associations Between Serum Insulin-like Growth Factor-I, Brain White Matter Volumes, and Cognition in Mild Cognitive Impairment and Alzheimer's Disease. J Alzheimers Dis 2024; 99:609-622. [PMID: 38701139 PMCID: PMC11191442 DOI: 10.3233/jad-231026] [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] [Accepted: 03/21/2024] [Indexed: 05/05/2024]
Abstract
Background Insulin-like growth factor-I (IGF-I) regulates myelin, but little is known whether IGF-I associates with white matter functions in subjective and objective mild cognitive impairment (SCI/MCI) or Alzheimer's disease (AD). Objective To explore whether serum IGF-I is associated with magnetic resonance imaging - estimated brain white matter volumes or cognitive functions. Methods In a prospective study of SCI/MCI (n = 106) and AD (n = 59), we evaluated the volumes of the total white matter, corpus callosum (CC), and white matter hyperintensities (WMHs) as well as Mini-Mental State Examination (MMSE), Trail Making Test A and B (TMT-A/B), and Stroop tests I-III at baseline, and after 2 years. Results IGF-I was comparable in SCI/MCI and AD (113 versus 118 ng/mL, p = 0.44). In SCI/MCI patients, the correlations between higher baseline IGF-I and greater baseline and 2-year volumes of the total white matter and total CC lost statistical significance after adjustment for intracranial volume and other covariates. However, after adjustment for covariates, higher baseline IGF-I correlated with better baseline scores of MMSE and Stroop test II in SCI/MCI and with better baseline results of TMT-B and Stroop test I in AD. IGF-I did not correlate with WMH volumes or changes in any of the variables. Conclusions Both in SCI/MCI and AD, higher IGF-I was associated with better attention/executive functions at baseline after adjustment for covariates. Furthermore, the baseline associations between IGF-I and neuropsychological test results in AD may argue against significant IGF-I resistance in the AD brain.
Collapse
Affiliation(s)
- Alexandra Horvath
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Patrick Quinlan
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carl Eckerström
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - N. David Åberg
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Acute Medicine and Geriatrics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johan Svensson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Internal Medicine, Skaraborg Central Hospital, Skövde, Sweden
| |
Collapse
|
13
|
Zegarra-Valdivia JA, Pignatelli J, Nuñez A, Torres Aleman I. The Role of Insulin-like Growth Factor I in Mechanisms of Resilience and Vulnerability to Sporadic Alzheimer's Disease. Int J Mol Sci 2023; 24:16440. [PMID: 38003628 PMCID: PMC10671249 DOI: 10.3390/ijms242216440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Despite decades of intense research, disease-modifying therapeutic approaches for Alzheimer's disease (AD) are still very much needed. Apart from the extensively analyzed tau and amyloid pathological cascades, two promising avenues of research that may eventually identify new druggable targets for AD are based on a better understanding of the mechanisms of resilience and vulnerability to this condition. We argue that insulin-like growth factor I (IGF-I) activity in the brain provides a common substrate for the mechanisms of resilience and vulnerability to AD. We postulate that preserved brain IGF-I activity contributes to resilience to AD pathology as this growth factor intervenes in all the major pathological cascades considered to be involved in AD, including metabolic impairment, altered proteostasis, and inflammation, to name the three that are considered to be the most important ones. Conversely, disturbed IGF-I activity is found in many AD risk factors, such as old age, type 2 diabetes, imbalanced diet, sedentary life, sociality, stroke, stress, and low education, whereas the Apolipoprotein (Apo) E4 genotype and traumatic brain injury may also be influenced by brain IGF-I activity. Accordingly, IGF-I activity should be taken into consideration when analyzing these processes, while its preservation will predictably help prevent the progress of AD pathology. Thus, we need to define IGF-I activity in all these conditions and develop a means to preserve it. However, defining brain IGF-I activity cannot be solely based on humoral or tissue levels of this neurotrophic factor, and new functionally based assessments need to be developed.
Collapse
Affiliation(s)
- Jonathan A. Zegarra-Valdivia
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain;
- Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain;
- School of Medicine, Universidad Señor de Sipán, Chiclayo 14000, Peru
| | - Jaime Pignatelli
- Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain;
- Cajal Institute (CSIC), 28002 Madrid, Spain
| | - Angel Nuñez
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| | - Ignacio Torres Aleman
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain;
- Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| |
Collapse
|
14
|
Chen L, Xiong XY, Yao TT, Gui LN, Luo F, Du Y, Cheng Y. Blood exosome sensing via neuronal insulin-like growth factor-1 regulates autism-related phenotypes. Pharmacol Res 2023; 197:106965. [PMID: 37852341 DOI: 10.1016/j.phrs.2023.106965] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/15/2023] [Accepted: 10/16/2023] [Indexed: 10/20/2023]
Abstract
The development and progression of autism spectrum disorder (ASD) is characterized by multiple complex molecular events, highlighting the importance of the prefrontal brain regions in this process. Exosomes are nanovesicles that play a critical role in intercellular communication. Peripheral systems influence brain function under both physiological and pathological conditions. We investigated whether this influence was mediated by the direct sensing of peripheral blood exosomes by brain cells. Administration of serum exosomes from rats with valproic acid-induced ASD resulted in ASD-related phenotypes in mice, whereas exosomes from normal rats did not exhibit such effects. RNA sequencing and bioinformatics analysis suggested that negative regulation of medial prefrontal cortex (mPFC) insulin-like growth factor 1 (IGF-1) by exosome-derived miR-29b-3p may contribute to these ASD-associated effects. Further evidence showed that miR-29b-3p-enriched exosomes crossed the blood-brain barrier to reach the mPFC, subsequently inducing the suppression of IGF-1 expression in neurons. Optogenetic activation of excitatory neurons in the mPFC improved behavioral abnormalities in exosome-treated mice. The addition of exogenous IGF-1 or inhibition of miR-29b-3p expression in the mPFC also rescued the ASD-related phenotypes in mice. Importantly, administration of miR-29b-3p-enriched serum exosomes from human donors with ASD into the mouse medial prefrontal cortex was sufficient to induce hallmark ASD behaviors. Together, our findings indicate that blood-brain cross-talk is crucial for ASD pathophysiology and that the brain may sense peripheral system changes through exosomes, which could serve as the basis for future neurological therapies.
Collapse
Affiliation(s)
- Lei Chen
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Xi-Yue Xiong
- NHC Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Tong-Tong Yao
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Lue-Ning Gui
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Fan Luo
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Yang Du
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China.
| | - Yong Cheng
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China; College of Life and Environmental Sciences, Minzu University of China, Beijing, China; Institute of National Security, Minzu University of China, Beijing, China.
| |
Collapse
|
15
|
Nuñez A, Zegarra-Valdivia J, Fernandez de Sevilla D, Pignatelli J, Torres Aleman I. The neurobiology of insulin-like growth factor I: From neuroprotection to modulation of brain states. Mol Psychiatry 2023; 28:3220-3230. [PMID: 37353586 DOI: 10.1038/s41380-023-02136-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/30/2023] [Accepted: 06/13/2023] [Indexed: 06/25/2023]
Abstract
After decades of research in the neurobiology of IGF-I, its role as a prototypical neurotrophic factor is undisputed. However, many of its actions in the adult brain indicate that this growth factor is not only involved in brain development or in the response to injury. Following a three-layer assessment of its role in the central nervous system, we consider that at the cellular level, IGF-I is indeed a bona fide neurotrophic factor, modulating along ontogeny the generation and function of all the major types of brain cells, contributing to sculpt brain architecture and adaptive responses to damage. At the circuit level, IGF-I modulates neuronal excitability and synaptic plasticity at multiple sites, whereas at the system level, IGF-I intervenes in energy allocation, proteostasis, circadian cycles, mood, and cognition. Local and peripheral sources of brain IGF-I input contribute to a spatially restricted, compartmentalized, and timed modulation of brain activity. To better define these variety of actions, we consider IGF-I a modulator of brain states. This definition aims to reconcile all aspects of IGF-I neurobiology, and may provide a new conceptual framework in the design of future research on the actions of this multitasking neuromodulator in the brain.
Collapse
Affiliation(s)
- A Nuñez
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, Madrid, Spain
| | - J Zegarra-Valdivia
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIBERNED, Madrid, Spain
- Universidad Señor de Sipán, Chiclayo, Perú
| | - D Fernandez de Sevilla
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, Madrid, Spain
| | - J Pignatelli
- CIBERNED, Madrid, Spain
- Cajal Institute (CSIC), Madrid, Spain
| | - I Torres Aleman
- Achucarro Basque Center for Neuroscience, Leioa, Spain.
- CIBERNED, Madrid, Spain.
- Ikerbasque Science Foundation, Bilbao, Spain.
| |
Collapse
|
16
|
Christiansen LI, Ventura GC, Holmqvist B, Aasmul-Olsen K, Lindholm SEH, Lycas MD, Mori Y, Secher JBM, Burrin DG, Thymann T, Sangild PT, Pankratova S. Insulin-like growth factor 1 supplementation supports motor coordination and affects myelination in preterm pigs. Front Neurosci 2023; 17:1205819. [PMID: 37404461 PMCID: PMC10315495 DOI: 10.3389/fnins.2023.1205819] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/23/2023] [Indexed: 07/06/2023] Open
Abstract
Introduction Preterm infants have increased risk of impaired neurodevelopment to which reduced systemic levels of insulin-like growth factor 1 (IGF-1) in the weeks after birth may play a role. Hence, we hypothesized that postnatal IGF-1 supplementation would improve brain development in preterm pigs, used as a model for preterm infants. Methods Preterm pigs delivered by cesarean section received recombinant human IGF-1/IGF binding protein-3 complex (rhIGF-1/rhIGFBP-3, 2.25 mg/kg/day) or vehicle from birth to postnatal day 19. Motor function and cognition were assessed by monitoring of in-cage and open field activities, balance beam test, gait parameters, novel object recognition and operant conditioning tests. Collected brains were subject to magnetic resonance imaging (MRI), immunohistochemistry, gene expression analyses and protein synthesis measurements. Results The IGF-1 treatment increased cerebellar protein synthesis rates (both in vivo and ex vivo). Performance in the balance beam test was improved by IGF-1 but not in other neurofunctional tests. The treatment decreased total and relative caudate nucleus weights, without any effects to total brain weight or grey/white matter volumes. Supplementation with IGF-1 reduced myelination in caudate nucleus, cerebellum, and white matter regions and decreased hilar synapse formation, without effects to oligodendrocyte maturation or neuron differentiation. Gene expression analyses indicated enhanced maturation of the GABAergic system in the caudate nucleus (decreased NKCC1:KCC2 ratio) with limited effects in cerebellum or hippocampus. Conclusion Supplemental IGF-1 during the first three weeks after preterm birth may support motor function by enhancing GABAergic maturation in the caudate nucleus, despite reduced myelination. Supplemental IGF-1 may support postnatal brain development in preterm infants, but more studies are required to identify optimal treatment regimens for subgroups of very or extremely preterm infants.
Collapse
Affiliation(s)
- Line I. Christiansen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Gemma C. Ventura
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Karoline Aasmul-Olsen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Sandy E. H. Lindholm
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Matthew D. Lycas
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yuki Mori
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jan Bojsen-Møller Secher
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Douglas G. Burrin
- United States Department of Agriculture, Agricultural Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Thomas Thymann
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Per T. Sangild
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
- Department of Neonatology, Rigshospitalet, Copenhagen, Denmark
- Department of Pediatrics, Odense University Hospital, Odense, Denmark
- Faculty of Theology, University of Copenhagen, Copenhagen, Denmark
| | - Stanislava Pankratova
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
El-Hage N, Haney MJ, Zhao Y, Rodriguez M, Wu Z, Liu M, Swain CJ, Yuan H, Batrakova EV. Extracellular Vesicles Released by Genetically Modified Macrophages Activate Autophagy and Produce Potent Neuroprotection in Mouse Model of Lysosomal Storage Disorder, Batten Disease. Cells 2023; 12:1497. [PMID: 37296618 PMCID: PMC10252192 DOI: 10.3390/cells12111497] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/11/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
Over the recent decades, the use of extracellular vesicles (EVs) has attracted considerable attention. Herein, we report the development of a novel EV-based drug delivery system for the transport of the lysosomal enzyme tripeptidyl peptidase-1 (TPP1) to treat Batten disease (BD). Endogenous loading of macrophage-derived EVs was achieved through transfection of parent cells with TPP1-encoding pDNA. More than 20% ID/g was detected in the brain following a single intrathecal injection of EVs in a mouse model of BD, ceroid lipofuscinosis neuronal type 2 (CLN2) mice. Furthermore, the cumulative effect of EVs repetitive administrations in the brain was demonstrated. TPP1-loaded EVs (EV-TPP1) produced potent therapeutic effects, resulting in efficient elimination of lipofuscin aggregates in lysosomes, decreased inflammation, and improved neuronal survival in CLN2 mice. In terms of mechanism, EV-TPP1 treatments caused significant activation of the autophagy pathway, including altered expression of the autophagy-related proteins LC3 and P62, in the CLN2 mouse brain. We hypothesized that along with TPP1 delivery to the brain, EV-based formulations can enhance host cellular homeostasis, causing degradation of lipofuscin aggregates through the autophagy-lysosomal pathway. Overall, continued research into new and effective therapies for BD is crucial for improving the lives of those affected by this condition.
Collapse
Affiliation(s)
- Nazira El-Hage
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (N.E.-H.); (M.R.)
| | - Matthew J. Haney
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.J.H.); (Y.Z.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.L.); (C.J.S.)
| | - Yuling Zhao
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.J.H.); (Y.Z.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.L.); (C.J.S.)
| | - Myosotys Rodriguez
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (N.E.-H.); (M.R.)
| | - Zhanhong Wu
- Department of Radiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; (Z.W.); (H.Y.)
| | - Mori Liu
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.L.); (C.J.S.)
| | - Carson J. Swain
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.L.); (C.J.S.)
| | - Hong Yuan
- Department of Radiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; (Z.W.); (H.Y.)
| | - Elena V. Batrakova
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.J.H.); (Y.Z.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.L.); (C.J.S.)
| |
Collapse
|
18
|
Borsellino P, Krider RI, Chea D, Grinnell R, Vida TA. Ketamine and the Disinhibition Hypothesis: Neurotrophic Factor-Mediated Treatment of Depression. Pharmaceuticals (Basel) 2023; 16:ph16050742. [PMID: 37242525 DOI: 10.3390/ph16050742] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Ketamine is a promising alternative to traditional pharmacotherapies for major depressive disorder, treatment-resistant depression, and other psychiatric conditions that heavily contribute to the global disease burden. In contrast to the current standard of care medications for these disorders, ketamine offers rapid onset, enduring clinical efficacy, and unique therapeutic potential for use in acute, psychiatric emergencies. This narrative presents an alternative framework for understanding depression, as mounting evidence supports a neuronal atrophy and synaptic disconnection theory, rather than the prevailing monoamine depletion hypothesis. In this context, we describe ketamine, its enantiomers, and various metabolites in a range of mechanistic actions through multiple converging pathways, including N-methyl-D-aspartate receptor (NMDAR) inhibition and the enhancement of glutamatergic signaling. We describe the disinhibition hypothesis, which posits that ketamine's pharmacological action ultimately results in excitatory cortical disinhibition, causing the release of neurotrophic factors, the most important of which is brain-derived neurotrophic factor (BDNF). BDNF-mediated signaling along with vascular endothelial growth factor (VEGF) and insulin-like growth factor 1 (IGF-1) subsequently give rise to the repair of neuro-structural abnormalities in patients with depressive disorders. Ketamine's efficacious amelioration of treatment-resistant depression is revolutionizing psychiatric treatment and opening up fresh vistas for understanding the underlying causes of mental illness.
Collapse
Affiliation(s)
- Philip Borsellino
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| | - Reese I Krider
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| | - Deanna Chea
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| | - Ryan Grinnell
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| | - Thomas A Vida
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| |
Collapse
|
19
|
Sharma DR, Cheng B, Jaiswal MK, Zhang X, Kumar A, Parikh N, Singh D, Sheth H, Varghese M, Dobrenis K, Zhang X, Hof PR, Stanton PK, Ballabh P. Elevated insulin growth factor-1 in dentate gyrus induces cognitive deficits in pre-term newborns. Cereb Cortex 2023; 33:6449-6464. [PMID: 36646459 PMCID: PMC10183730 DOI: 10.1093/cercor/bhac516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 01/18/2023] Open
Abstract
Prematurely born infants are deprived of maternal hormones and cared for in the stressful environment of Neonatal Intensive Care Units (NICUs). They suffer from long-lasting deficits in learning and memory. Here, we show that prematurity and associated neonatal stress disrupt dentate gyrus (DG) development and induce long-term cognitive deficits and that these effects are mediated by insulin growth factor-1 (IGF1). Nonmaternal care of premature rabbits increased the number of granule cells and interneurons and reduced neurogenesis, suggesting accelerated premature maturation of DG. However, the density of glutamatergic synapses, mature dendritic spines, and synaptic transmission were reduced in preterm kits compared with full-term controls, indicating that premature synaptic maturation was abnormal. These findings were consistent with cognitive deficits observed in premature rabbits and appeared to be driven by transcriptomic changes in the granule cells. Preterm kits displayed reduced weight, elevated serum cortisol and growth hormone, and higher IGF1 expression in the liver and DG relative to full-term controls. Importantly, blocking IGF-1 receptor in premature kits restored cognitive deficits, increased the density of glutamatergic puncta, and rescued NR2B and PSD95 levels in the DG. Hence, IGF1 inhibition alleviates prematurity-induced cognitive dysfunction and synaptic changes in the DG through modulation of NR2B and PSD95. The study identifies a novel strategy to potentially rescue DG maldevelopment and cognitive dysfunction in premature infants under stress in NICUs.
Collapse
Affiliation(s)
- Deep R Sharma
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Bokun Cheng
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Manoj Kumar Jaiswal
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xusheng Zhang
- Computational Genomics Core, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ajeet Kumar
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Nirzar Parikh
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Divya Singh
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Hardik Sheth
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Merina Varghese
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kostantin Dobrenis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Xiaolei Zhang
- Departments of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Patric K Stanton
- Departments of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA
| | - Praveen Ballabh
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
20
|
Sullivan M, Fernandez-Aranda F, Camacho-Barcia L, Harkin A, Macrì S, Mora-Maltas B, Jiménez-Murcia S, O'Leary A, Ottomana AM, Presta M, Slattery D, Scholtz S, Glennon JC. Insulin and Disorders of Behavioural Flexibility. Neurosci Biobehav Rev 2023; 150:105169. [PMID: 37059405 DOI: 10.1016/j.neubiorev.2023.105169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/03/2023] [Accepted: 04/10/2023] [Indexed: 04/16/2023]
Abstract
Behavioural inflexibility is a symptom of neuropsychiatric and neurodegenerative disorders such as Obsessive-Compulsive Disorder, Autism Spectrum Disorder and Alzheimer's Disease, encompassing the maintenance of a behaviour even when no longer appropriate. Recent evidence suggests that insulin signalling has roles apart from its regulation of peripheral metabolism and mediates behaviourally-relevant central nervous system (CNS) functions including behavioural flexibility. Indeed, insulin resistance is reported to generate anxious, perseverative phenotypes in animal models, with the Type 2 diabetes medication metformin proving to be beneficial for disorders including Alzheimer's Disease. Structural and functional neuroimaging studies of Type 2 diabetes patients have highlighted aberrant connectivity in regions governing salience detection, attention, inhibition and memory. As currently available therapeutic strategies feature high rates of resistance, there is an urgent need to better understand the complex aetiology of behaviour and develop improved therapeutics. In this review, we explore the circuitry underlying behavioural flexibility, changes in Type 2 diabetes, the role of insulin in CNS outcomes and mechanisms of insulin involvement across disorders of behavioural inflexibility.
Collapse
Affiliation(s)
- Mairéad Sullivan
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland.
| | - Fernando Fernandez-Aranda
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Lucía Camacho-Barcia
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain
| | - Andrew Harkin
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland
| | - Simone Macrì
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Bernat Mora-Maltas
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Susana Jiménez-Murcia
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Aet O'Leary
- University Hospital Frankfurt, Frankfurt, Germany
| | - Angela Maria Ottomana
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; Neuroscience Unit, Department of Medicine, University of Parma, 43100 Parma, Italy
| | - Martina Presta
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | | | | | - Jeffrey C Glennon
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
21
|
Holloway RK, Zhang L, Molina-Gonzalez I, Ton K, Nicoll JAR, Boardman JP, Liang Y, Williams A, Miron VE. Localized microglia dysregulation impairs central nervous system myelination in development. Acta Neuropathol Commun 2023; 11:49. [PMID: 36949514 PMCID: PMC10035254 DOI: 10.1186/s40478-023-01543-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/03/2023] [Indexed: 03/24/2023] Open
Abstract
Myelination of neuronal axons is a critical aspect of central nervous system development and function. However, the fundamental cellular and molecular mechanisms influencing human developmental myelination and its failure are not fully understood. Here, we used digital spatial transcriptomics of a rare bank of human developing white matter to uncover that a localized dysregulated innate immune response is associated with impeded myelination. We identified that poorly myelinating areas have a distinct signature of Type II interferon signalling in microglia/macrophages, relative to adjacent myelinating areas. This is associated with a surprising increase in mature oligodendrocytes, which fail to form myelin processes appropriately. We functionally link these findings by showing that conditioned media from interferon-stimulated microglia is sufficient to dysregulate myelin process formation by oligodendrocytes in culture. We identify the Type II interferon inducer, Osteopontin (SPP1), as being upregulated in poorly myelinating brains, indicating a potential biomarker. Our results reveal the importance of microglia-mature oligodendrocyte interaction and interferon signaling in regulating myelination of the developing human brain.
Collapse
Affiliation(s)
- Rebecca K Holloway
- Keenan Research Centre for Biomedial Science at St. Michael's Hospital, 209 Victoria Street, Toronto, ON, M5B 1T8, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, Scotland, UK
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Liang Zhang
- Nanostring Technologies, Inc., Seattle, WA, USA
| | - Irene Molina-Gonzalez
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, Scotland, UK
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Kathy Ton
- Nanostring Technologies, Inc., Seattle, WA, USA
| | - James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - James P Boardman
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Yan Liang
- Nanostring Technologies, Inc., Seattle, WA, USA
| | - Anna Williams
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Veronique E Miron
- Keenan Research Centre for Biomedial Science at St. Michael's Hospital, 209 Victoria Street, Toronto, ON, M5B 1T8, Canada.
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, UK.
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, Scotland, UK.
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, UK.
| |
Collapse
|
22
|
Locatelli G, Marques-Ferreira F, Katsoulas A, Kalaitzaki V, Krueger M, Ingold-Heppner B, Walthert S, Sankowski R, Prazeres da Costa O, Dolga A, Huber M, Gold M, Culmsee C, Waisman A, Bechmann I, Milchevskaya V, Prinz M, Tresch A, Becher B, Buch T. IGF1R expression by adult oligodendrocytes is not required in the steady-state but supports neuroinflammation. Glia 2023; 71:616-632. [PMID: 36394300 DOI: 10.1002/glia.24299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022]
Abstract
In the central nervous system (CNS), insulin-like growth factor 1 (IGF-1) regulates myelination by oligodendrocyte (ODC) precursor cells and shows anti-apoptotic properties in neuronal cells in different in vitro and in vivo systems. Previous work also suggests that IGF-1 protects ODCs from cell death and enhances remyelination in models of toxin-induced and autoimmune demyelination. However, since evidence remains controversial, the therapeutic potential of IGF-1 in demyelinating CNS conditions is unclear. To finally shed light on the function of IGF1-signaling for ODCs, we deleted insulin-like growth factor 1 receptor (IGF1R) specifically in mature ODCs of the mouse. We found that ODC survival and myelin status were unaffected by the absence of IGF1R until 15 months of age, indicating that IGF-1 signaling does not play a major role in post-mitotic ODCs during homeostasis. Notably, the absence of IGF1R did neither affect ODC survival nor myelin status upon cuprizone intoxication or induction of experimental autoimmune encephalomyelitis (EAE), models for toxic and autoimmune demyelination, respectively. Surprisingly, however, the absence of IGF1R from ODCs protected against clinical neuroinflammation in the EAE model. Together, our data indicate that IGF-1 signaling is not required for the function and survival of mature ODCs in steady-state and disease.
Collapse
Affiliation(s)
- Giuseppe Locatelli
- Institute of Experimental Immunology, University of Zurich, Zurich.,Theodor Kocher Institute, University Bern, Bern, Switzerland
| | | | - Antonis Katsoulas
- Institute of Laboratory Animal Science, University of Zurich, Zurich
| | | | - Martin Krueger
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Barbara Ingold-Heppner
- Institute of Pathology, Campus Mitte, Charité -Universitätsmedizin Berlin, Berlin, Germany
| | | | - Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Olivia Prazeres da Costa
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| | - Amalia Dolga
- Institute for Pharmacology and Clinical Pharmacy, Philipps-Universität Marburg, Marburg, Germany.,Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Magdalena Huber
- Institute for Medical Microbiology and Hospital Hygiene, Philipps University of Marburg, Marburg, Germany
| | - Maike Gold
- Department of Neurology, Philipps University of Marburg, Marburg, Germany
| | - Carsten Culmsee
- Institute for Pharmacology and Clinical Pharmacy, Philipps-Universität Marburg, Marburg, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Vladislava Milchevskaya
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Achim Tresch
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich
| | - Thorsten Buch
- Institute of Experimental Immunology, University of Zurich, Zurich.,Institute of Laboratory Animal Science, University of Zurich, Zurich.,Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| |
Collapse
|
23
|
Neurotrophic Factors as Regenerative Therapy for Neurodegenerative Diseases: Current Status, Challenges and Future Perspectives. Int J Mol Sci 2023; 24:ijms24043866. [PMID: 36835277 PMCID: PMC9968045 DOI: 10.3390/ijms24043866] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/25/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS), spinal cord injury (SCI), and amyotrophic lateral sclerosis (ALS), are characterized by acute or chronic progressive loss of one or several neuronal subtypes. However, despite their increasing prevalence, little progress has been made in successfully treating these diseases. Research has recently focused on neurotrophic factors (NTFs) as potential regenerative therapy for neurodegenerative diseases. Here, we discuss the current state of knowledge, challenges, and future perspectives of NTFs with a direct regenerative effect in chronic inflammatory and degenerative disorders. Various systems for delivery of NTFs, such as stem and immune cells, viral vectors, and biomaterials, have been applied to deliver exogenous NTFs to the central nervous system, with promising results. The challenges that currently need to be overcome include the amount of NTFs delivered, the invasiveness of the delivery route, the blood-brain barrier permeability, and the occurrence of side effects. Nevertheless, it is important to continue research and develop standards for clinical applications. In addition to the use of single NTFs, the complexity of chronic inflammatory and degenerative diseases may require combination therapies targeting multiple pathways or other possibilities using smaller molecules, such as NTF mimetics, for effective treatment.
Collapse
|
24
|
Riikonen R. Biochemical mechanisms in pathogenesis of infantile epileptic spasm syndrome. Seizure 2023; 105:1-9. [PMID: 36634586 DOI: 10.1016/j.seizure.2023.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/01/2023] [Accepted: 01/05/2023] [Indexed: 01/09/2023] Open
Abstract
The molecular mechanisms leading to infantile epileptic spasm syndrome (IESS) remain obscure. The only common factor seems to be that the spasms are restricted to a limited period of infancy, during a certain maturational state. Here the current literature regarding the biochemical mechanisms of brain maturation in IESS is reviewed, and various hypotheses of the pathophysiology are put together. They include: (1) imbalance of inhibitory (NGF, IGF-1, ACTH, GABA) and excitatory factors (glutamate, nitrites) which distinguishes the different etiological subgroups, (2) abnormality of the hypothalamic pituitary adrenal (HPA) axis linking insults and early life stress, (3) inflammation (4) yet poorly known genetic and epigenetic factors, and (5) glucocorticoid and vigabatrin action on brain development, pinpointing at molecular targets of the pathophysiology from another angle. An altered maturational process may explain why so many, seemingly independent etiological factors lead to the same clinical syndrome and frequently to developmental delay. Understanding these factors can provide ideas for novel therapies.
Collapse
Affiliation(s)
- Raili Riikonen
- Children's Hospital, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland.
| |
Collapse
|
25
|
Stereological Evidence of Non-Selective Hippocampal Neurodegeneration, IGF-1 Depletion, and Behavioral Deficit following Short Term Bilateral Adrenalectomy in Wistar Rats. Biomolecules 2022; 13:biom13010022. [PMID: 36671407 PMCID: PMC9855887 DOI: 10.3390/biom13010022] [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: 10/18/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
The development of animal models to study cell death in the brain is a delicate task. One of the models, that was discovered in the late eighties, is the induction of neurodegeneration through glucocorticoid withdrawal by adrenalectomy in albino rats. Such a model is one of the few noninvasive models for studying neurodegeneration. In the present study, using stereological technique and ultrastructural examination, we aimed to investigate the impact of short-term adrenalectomy (2 weeks) on different hippocampal neuronal populations in Wistar rats. In addition, the underlying mechanism(s) of degeneration in these neurons were investigated by measuring the levels of insulin-like growth factor-1 (IGF-1) and β-nerve growth factor (β-NGF). Moreover, we examined whether the biochemical and histological changes in the hippocampus, after short-term adrenalectomy, have an impact on the cognitive behavior of Wistar rats. Stereological counting in the hippocampus revealed significant neuronal deaths in the dentate gyrus and CA4/CA3, but not in the CA2 and CA1 areas, 7 and 14 days post adrenalectomy. The ultrastructural examinations revealed degenerated and degenerating neurons in the dentate, as well as CA4, and CA3 areas, over the course of 3, 7 and 14 days. The levels of IGF-1 were significantly decreased in the hippocampus of ADX rats 24 h post adrenalectomy, and lasted over the course of two weeks. However, β-NGF was not affected in rats. Using a passive avoidance task, we found a cognitive deficit in the ADX compared to the SHAM operated rats over time (3, 7, and 14 days). In conclusion, both granule and pyramidal cells were degenerated in the hippocampus following short-term adrenalectomy. The early depletion of IGF-1 might play a role in hippocampal neuronal degeneration. Consequently, the loss of the hippocampal neurons after adrenalectomy leads to cognitive deficits.
Collapse
|
26
|
Giacomozzi C, Martin A, Fernández MC, Gutiérrez M, Iascone M, Domené HM, Dominici FP, Bergadá I, Cangiano B, Persani L, Pennisi PA. Novel Insulin-Like Growth Factor 1 Gene Mutation: Broadening of the Phenotype and Implications for Insulin Resistance. J Clin Endocrinol Metab 2022; 108:1355-1369. [PMID: 36546343 DOI: 10.1210/clinem/dgac738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 11/23/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE Insulin-like Growth Factor (IGF)1 gene mutations are extremely rare causes of pre- and post-natal growth retardation. Phenotype can be heterogenous with varying degrees of neurosensory deafness, cognitive defects, glucose metabolism impairment and short stature. This study describes a 12.6-year-old girl presenting severe short stature and insulin resistance, but with normal hearing and neurological development at the lower limit of normal. METHODS DNA was obtained from the proband and both parents for whole exome sequencing (WES). In silico analysis was performed to predict the impact of the IGF1 variant on IGF1 and insulin receptors (IGF1R and IR) signalling. Phosphorylation of the IGF1R at activating Tyr residues and cell proliferation analyses were used to assess the ability of each subject's IGF1 to bind and activate IGF1R. RESULTS The proband had low immunoreactive IGF1 in serum and WES revealed a novel homozygous IGF1 missense variant (c.247A > T), causing a change of serine 83 for cysteine (p.Ser83Cys; p.Ser35Cys in mature peptide). The proband's parents were heterozygous for this mutation. In silico analyses indicated the pathogenic potential of the variant with electrostatic variations with the potential of hampering the interaction with the IGF1R but strengthening the binding to IR. The mutant IGF1 protein had a significantly reduced activity on in vitro bioassays. MAIN CONCLUSIONS We describe a novel IGF1 mutation leading to severe loss of circulating IGF1 immunoreactivity and bioactivity, In silico modelling predicts that the mutant IGF1 could interfere with IR signalling, providing a possible explanation for the severe insulin resistance observed in the patient. The absence of significant hearing and neurodevelopmental involvement in the present case is unusual and broadens the clinical spectrum of IGF1 mutations.
Collapse
Affiliation(s)
- Claudio Giacomozzi
- Unit of Pediatrics, Department of Maternal and Child Health, Carlo Poma Hospital, ASST-Mantova, Mantua, Italy
| | - Ayelen Martin
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
| | - María Celia Fernández
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Mariana Gutiérrez
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Maria Iascone
- Department of Medical Genetics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Horacio M Domené
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Fernando P Dominici
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica (IQUIFIB-CONICET), Buenos Aires, Argentina
| | - Ignacio Bergadá
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Biagio Cangiano
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20100 Milan, Italy
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, 20100 Milan, Italy
| | - Luca Persani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20100 Milan, Italy
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, 20100 Milan, Italy
| | - Patricia A Pennisi
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
| |
Collapse
|
27
|
Protective role of IGF-1 and GLP-1 signaling activation in neurological dysfunctions. Neurosci Biobehav Rev 2022; 142:104896. [PMID: 36191807 DOI: 10.1016/j.neubiorev.2022.104896] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 09/09/2022] [Accepted: 09/26/2022] [Indexed: 11/24/2022]
Abstract
Insulin-like growth factor-1 (IGF-1), a pleiotropic polypeptide, plays an essential role in CNS development and maturation. Glucagon-like peptide-1 (GLP-1) is an endogenous incretin hormone that regulates blood glucose levels and fatty acid oxidation in the brain. GLP-1 also exhibits similar functions and growth factor-like properties to IGF-1, which is likely how it exerts its neuroprotective effects. Recent preclinical and clinical evidence indicate that IGF-1 and GLP-1, apart from regulating growth and development, prevent neuronal death mediated by amyloidogenesis, cerebral glucose deprivation, neuroinflammation and apoptosis through modulation of PI3/Akt kinase, mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase (MAPK/ERK). IGF-1 resistance and GLP-1 deficiency impair protective cellular signaling mechanisms, contributing to the progression of neurodegenerative diseases. Over the past decades, IGF-1 and GLP-1 have emerged as an essential component of the neuronal system and as potential therapeutic targets for several neurodegenerative and neuropsychiatric dysfunctions. There is substantial evidence that IGF-1 and GLP-1 analogues penetrate the blood-brain barrier (BBB) and exhibit neuroprotective functions, including synaptic formation, neuronal plasticity, protein synthesis, and autophagy. Conclusively, this review represents the therapeutic potential of IGF-1 and GLP-1 signaling target activators in ameliorating neurological disorders.
Collapse
|
28
|
Ballester-Rosado CJ, Le JT, Lam TT, Mohila CA, Lam S, Anderson AE, Frost JD, Swann JW. A Role for Insulin-like Growth Factor 1 in the Generation of Epileptic Spasms in a murine model. Ann Neurol 2022; 92:45-60. [PMID: 35467038 PMCID: PMC9233100 DOI: 10.1002/ana.26383] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 04/21/2022] [Accepted: 04/23/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Infantile spasms are associated with a wide variety of clinical conditions, including perinatal brain injuries. We have created a model in which prolonged infusion of tetrodotoxin (TTX) into the neocortex, beginning in infancy, produces a localized lesion and reproduces the behavioral spasms, electroencephalogram (EEG) abnormalities, and drug responsiveness seen clinically. Here, we undertook experiments to explore the possibility that the growth factor IGF-1 plays a role in generating epileptic spasms. METHODS We combined long-term video EEG recordings with quantitative immunohistochemical and biochemical analyses to unravel IGF-1's role in spasm generation. Immunohistochemistry was undertaken in surgically resected tissue from infantile spasms patients. We used viral injections in neonatal conditional IGF-1R knock-out mice to show that an IGF-1-derived tripeptide (1-3)IGF-1, acts through the IGF-1 receptor to abolish spasms. RESULTS Immunohistochemical methods revealed widespread loss of IGF-1 from cortical neurons, but an increase in IGF-1 in the reactive astrocytes in the TTX-induced lesion. Very similar changes were observed in the neocortex from patients with spasms. In animals, we observed reduced signaling through the IGF-1 growth pathways in areas remote from the lesion. To show the reduction in IGF-1 expression plays a role in spasm generation, epileptic rats were treated with (1-3)IGF-1. We provide 3 lines of evidence that (1-3)IGF-1 activates the IGF-1 signaling pathway by acting through the receptor for IGF-1. Treatment with (1-3)IGF-1 abolished spasms and hypsarrhythmia-like activity in the majority of animals. INTERPRETATION Results implicate IGF-1 in the pathogenesis of infantile spasms and IGF-1 analogues as potential novel therapies for this neurodevelopmental disorder. ANN NEUROL 2022;92:45-60.
Collapse
Affiliation(s)
- Carlos J. Ballester-Rosado
- The Cain Foundation Laboratories, the Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - John T. Le
- The Cain Foundation Laboratories, the Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Trang T. Lam
- The Cain Foundation Laboratories, the Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Carrie A. Mohila
- Department of Pathology and Immunology, Baylor College of Medicine
- Department of Pathology, Texas Children’s Hospital, Houston, Texas, USA
| | - Sandi Lam
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
| | - Anne E. Anderson
- The Cain Foundation Laboratories, the Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA
| | - James D. Frost
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA
| | - John W. Swann
- The Cain Foundation Laboratories, the Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
29
|
Horvath A, Quinlan P, Eckerström C, Åberg ND, Wallin A, Svensson J. Low Serum Insulin-like Growth Factor-I Is Associated with Decline in Hippocampal Volume in Stable Mild Cognitive Impairment but not in Alzheimer's Disease. J Alzheimers Dis 2022; 88:1007-1016. [PMID: 35723105 PMCID: PMC9484094 DOI: 10.3233/jad-220292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Serum insulin-like growth factor-I (IGF-I) has shown some association with hippocampal volume in healthy subjects, but this relation has not been investigated in stable mild cognitive impairment (sMCI) or Alzheimer’s disease (AD). Objective: At a single memory clinic, we investigated whether serum IGF-I was associated with baseline magnetic resonance imaging (MRI)-estimated brain volumes and longitudinal alterations, defined as annualized changes, up to 6 years of follow-up. Methods: A prospective study of patients with sMCI (n = 110) and AD (n = 60). Brain regions included the hippocampus and amygdala as well as the temporal, parietal, frontal, and occipital lobes, respectively. Results: Serum IGF-I was statistically similar in sMCI and AD patients (112 versus 123 ng/mL, p = 0.31). In sMCI, serum IGF-I correlated positively with all baseline MRI variables except for the occipital lobe, and there was also a positive correlation between serum IGF-I and the annualized change in hippocampal volume (rs = 0.32, p = 0.02). Furthermore, sMCI patients having serum IGF-I above the median had lower annual loss of hippocampal volume than those with IGF-I below the median (p = 0.02). In contrast, in AD patients, IGF-I did not associate with baseline levels or annualized changes in brain volumes. Conclusion: In sMCI patients, our results suggest that IGF-I exerted neuroprotective effects on the brain, thereby maintaining hippocampal volume. In AD, serum IGF-I did not associate with brain volumes, indicating that IGF-I could not induce neuroprotection in this disease. This supports the notion of IGF-I resistance in AD.
Collapse
Affiliation(s)
- Alexandra Horvath
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Patrick Quinlan
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Carl Eckerström
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Immunology and Transfusion Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - N David Åberg
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Acute Medicine and Geriatrics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johan Svensson
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Internal Medicine, Region Västra Götaland, Skaraborg Central Hospital, Skövde, Sweden
| |
Collapse
|
30
|
Kan EK, Atmaca A, Sarisoy G, Ecemis GC, Gokosmanoglu F. Personality traits in acromegalic patients: Comparison with patients with non-functioning adenomas and healthy controls. Growth Horm IGF Res 2022; 62:101439. [PMID: 34814008 DOI: 10.1016/j.ghir.2021.101439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/24/2021] [Accepted: 11/14/2021] [Indexed: 11/18/2022]
Abstract
OBJECTIVES Pituitary diseases may cause psychiatric and personality alterations. We aimed to compare the personality traits of acromegalic patients with those of patients with non-functioning pituitary adenomas and a healthy control group. DESIGN Fifty-eight acromegalic patients, 45 patients with non-functioning adenoma, and 40 healthy subjects were enrolled in the study. Cloninger's Temperament and Character Inventory (TCI), Beck Depression Inventory, Beck Anxiety Inventory, and Rosenberg Self-Esteem Scale (RSES) were used to assess personality, depression, anxiety, and self-esteem. RESULTS Depression score was higher in acromegaly and non-functioning adenoma groups than healthy controls. RSES scores were similar among the three groups. Regarding the scales of TCI, only novelty-seeking was significantly reduced in acromegaly and non-functioning adenoma than the control group. Pairwise comparisons revealed that the difference was due to the difference between acromegalic patients and controls. Scales of TCI were correlated with depression and anxiety in patients with acromegaly and non-functioning adenoma but not in healthy controls. CONCLUSION This study showed that novelty-seeking was reduced in patients with acromegaly. Both the hormonal lack and excess and structural changes can lead to cognitive and personality changes in acromegaly. More studies are needed to be carried out about personality characteristics in pituitary diseases.
Collapse
Affiliation(s)
- Elif Kilic Kan
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Ondokuz Mayis University Medical of School, Samsun 55200, Turkey.
| | - Aysegul Atmaca
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Ondokuz Mayis University Medical of School, Samsun 55200, Turkey
| | - Gokhan Sarisoy
- Department of Psychiatry, Ondokuz Mayis University Medical of School, Samsun 55200, Turkey
| | - Gulcin Cengiz Ecemis
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Ondokuz Mayis University Medical of School, Samsun 55200, Turkey
| | - Feyzi Gokosmanoglu
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Ondokuz Mayis University Medical of School, Samsun 55200, Turkey
| |
Collapse
|
31
|
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
|
32
|
Danjo Y, Shinozaki Y, Natsubori A, Kubota Y, Kashiwagi K, Tanaka KF, Koizumi S. The Mlc1 Promoter Directs Müller Cell-specific Gene Expression in the Retina. Transl Vis Sci Technol 2022; 11:25. [PMID: 35040915 PMCID: PMC8764212 DOI: 10.1167/tvst.11.1.25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Because the importance of glia in regulating brain functions has been demonstrated, genetic technologies that manipulate glial cell-specific gene expression in the brain have become essential and have made great progress. However, it is unknown whether the same strategy that is used in the brain can be applied to the retina because retinal glia differs from glia in the brain. Here, we aimed to find a method for selective gene expression in Müller cells (characteristic glial cells in the retina) and identified Mlc1 as a specific promoter of Müller cells. Methods Mlc1-tTA::Yellow-Cameleon-NanotetO/tetO (YC-Nano) mice were used as a reporter line. YC-Nano, a fluorescent protein, was ectopically expressed in the cell type controlled by the Mlc1 promotor. Immunofluorescence staining was used to identify the cell type expressing YC-Nano protein. Results YC-Nano-positive (+) signals were observed as vertical stalks in the sliced retina and spanned from the nerve fiber layer through the outer nuclear layer. The density of YC-Nano+ cells was higher around the optic nerve head and lower in the peripheral retina. The YC-Nano+ signals colocalized with vimentin, a marker of Müller cells, but not with the cell markers for blood vessels, microglia, neurons, or astrocytes. Conclusions The Mlc1 promoter allows us to manipulate gene expression in Müller cells without affecting astrocytes in the retina. Translational Relevance Gene manipulation under control of Mlc1 promoter offers novel technique to investigate the role of Müller cells.
Collapse
Affiliation(s)
- Yosuke Danjo
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Akiyo Natsubori
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yuto Kubota
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Kenji Kashiwagi
- Department of Ophthalmology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
33
|
Khan MS, Spann RA, Münzberg H, Yu S, Albaugh VL, He Y, Berthoud HR, Morrison CD. Protein Appetite at the Interface between Nutrient Sensing and Physiological Homeostasis. Nutrients 2021; 13:4103. [PMID: 34836357 PMCID: PMC8620426 DOI: 10.3390/nu13114103] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/04/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
Feeding behavior is guided by multiple competing physiological needs, as animals must sense their internal nutritional state and then identify and consume foods that meet nutritional needs. Dietary protein intake is necessary to provide essential amino acids and represents a specific, distinct nutritional need. Consistent with this importance, there is a relatively strong body of literature indicating that protein intake is defended, such that animals sense the restriction of protein and adaptively alter feeding behavior to increase protein intake. Here, we argue that this matching of food consumption with physiological need requires at least two concurrent mechanisms: the first being the detection of internal nutritional need (a protein need state) and the second being the discrimination between foods with differing nutritional compositions. In this review, we outline various mechanisms that could mediate the sensing of need state and the discrimination between protein-rich and protein-poor foods. Finally, we briefly describe how the interaction of these mechanisms might allow an animal to self-select between a complex array of foods to meet nutritional needs and adaptively respond to changes in either the external environment or internal physiological state.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Christopher D. Morrison
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; (M.S.K.); (R.A.S.); (H.M.); (S.Y.); (V.L.A.); (Y.H.); (H.-R.B.)
| |
Collapse
|
34
|
Luo R, Zhang H, Mukherjee N, Karmaus W, Patil V, Arshad H, Mzayek F. Association of grandmaternal smoking during pregnancy with DNA methylation of grandchildren: the Isle of Wight study. Epigenomics 2021; 13:1473-1483. [PMID: 34596434 DOI: 10.2217/epi-2020-0433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background: To investigate the intergenerational effects of grandmaternal smoking during pregnancy (GMSDP) on the DNA methylation of grandchildren. Methods: Data from the Isle of Wight birth cohort with information regarding GMSDP and DNA methylation profiling at the birth of grandchildren (n = 161) were used. Differentially methylated CpG sites related to GMSDP were identified using testing-training screening, analysis of variance and multivariate analysis of covariance. The association between identified CpG sites and expression levels of neighboring genes was tested by linear regression. Results: Twenty-three CpG sites were differentially methylated in grandchildren because of GMSDP, and eight of these were associated with expression levels of 13 neighboring genes. Conclusion: GMSDP has an intergenerational effect on the DNA methylation profile of grandchildren independent of maternal smoking during pregnancy.
Collapse
Affiliation(s)
- Rui Luo
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Memphis, TN 38152, USA
| | - Hongmei Zhang
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Memphis, TN 38152, USA
| | - Nandini Mukherjee
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Memphis, TN 38152, USA
| | - Wilfried Karmaus
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Memphis, TN 38152, USA
| | - Veeresh Patil
- David Hide Asthma and Allergy Research Centre, Newport, PO30 5TG, UK
| | - Hasan Arshad
- David Hide Asthma and Allergy Research Centre, Newport, PO30 5TG, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Fawaz Mzayek
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Memphis, TN 38152, USA
| |
Collapse
|
35
|
Zuccaro E, Piol D, Basso M, Pennuto M. Motor Neuron Diseases and Neuroprotective Peptides: A Closer Look to Neurons. Front Aging Neurosci 2021; 13:723871. [PMID: 34603008 PMCID: PMC8484953 DOI: 10.3389/fnagi.2021.723871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/26/2021] [Indexed: 12/02/2022] Open
Abstract
Motor neurons (MNs) are specialized neurons responsible for muscle contraction that specifically degenerate in motor neuron diseases (MNDs), such as amyotrophic lateral sclerosis (ALS), spinal and bulbar muscular atrophy (SBMA), and spinal muscular atrophy (SMA). Distinct classes of MNs degenerate at different rates in disease, with a particular class named fast-fatigable MNs (FF-MNs) degenerating first. The etiology behind the selective vulnerability of FF-MNs is still largely under investigation. Among the different strategies to target MNs, the administration of protective neuropeptides is one of the potential therapeutic interventions. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide with beneficial effects in many neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and more recently SBMA. Another neuropeptide that has a neurotrophic effect on MNs is insulin-like growth factor 1 (IGF-1), also known as somatomedin C. These two peptides are implicated in the activation of neuroprotective pathways exploitable in the amelioration of pathological outcomes related to MNDs.
Collapse
Affiliation(s)
- Emanuela Zuccaro
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
- Padova Neuroscience Center, Padua, Italy
| | - Diana Piol
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Manuela Basso
- Department of Cellular, Computational and Integrative Biology – CIBIO, University of Trento, Trento, Italy
| | - Maria Pennuto
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
- Padova Neuroscience Center, Padua, Italy
| |
Collapse
|
36
|
Levy JA, LaFlamme CW, Tsaprailis G, Crynen G, Page DT. Dyrk1a Mutations Cause Undergrowth of Cortical Pyramidal Neurons via Dysregulated Growth Factor Signaling. Biol Psychiatry 2021; 90:295-306. [PMID: 33840455 PMCID: PMC8787822 DOI: 10.1016/j.biopsych.2021.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND Mutations in DYRK1A are a cause of microcephaly, autism spectrum disorder, and intellectual disability; however, the underlying cellular and molecular mechanisms are not well understood. METHODS We generated a conditional mouse model using Emx1-cre, including conditional heterozygous and homozygous knockouts, to investigate the necessity of Dyrk1a in the cortex during development. We used unbiased, high-throughput phosphoproteomics to identify dysregulated signaling mechanisms in the developing Dyrk1a mutant cortex as well as classic genetic modifier approaches and pharmacological therapeutic intervention to rescue microcephaly and neuronal undergrowth caused by Dyrk1a mutations. RESULTS We found that cortical deletion of Dyrk1a in mice causes decreased brain mass and neuronal size, structural hypoconnectivity, and autism-relevant behaviors. Using phosphoproteomic screening, we identified growth-associated signaling cascades dysregulated upon Dyrk1a deletion, including TrkB-BDNF (tyrosine receptor kinase B-brain-derived neurotrophic factor), an important regulator of ERK/MAPK (extracellular signal-regulated kinase/mitogen-activated protein kinase) and mTOR (mammalian target of rapamycin) signaling. Genetic suppression of Pten or pharmacological treatment with IGF-1 (insulin-like growth factor-1), both of which impinge on these signaling cascades, rescued microcephaly and neuronal undergrowth in neonatal mutants. CONCLUSIONS Altogether, these findings identify a previously unknown mechanism through which Dyrk1a mutations disrupt growth factor signaling in the developing brain, thus influencing neuronal growth and connectivity. Our results place DYRK1A as a critical regulator of a biological pathway known to be dysregulated in humans with autism spectrum disorder and intellectual disability. In addition, these data position Dyrk1a within a larger group of autism spectrum disorder/intellectual disability risk genes that impinge on growth-associated signaling cascades to regulate brain size and connectivity, suggesting a point of convergence for multiple autism etiologies.
Collapse
Affiliation(s)
- Jenna A Levy
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida; Doctoral Program in Chemical and Biological Sciences, The Skaggs Graduate School of Chemical and Biological Sciences at Scripps Research, Jupiter, Florida
| | - Christy W LaFlamme
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida; The Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, Florida
| | | | - Gogce Crynen
- Center for Computational Biology and Bioinformatics, The Scripps Research Institute, Jupiter, Florida
| | - Damon T Page
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida; Doctoral Program in Chemical and Biological Sciences, The Skaggs Graduate School of Chemical and Biological Sciences at Scripps Research, Jupiter, Florida.
| |
Collapse
|
37
|
Consorti A, Di Marco I, Sansevero G. Physical Exercise Modulates Brain Physiology Through a Network of Long- and Short-Range Cellular Interactions. Front Mol Neurosci 2021; 14:710303. [PMID: 34489641 PMCID: PMC8417110 DOI: 10.3389/fnmol.2021.710303] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/23/2021] [Indexed: 12/14/2022] Open
Abstract
In the last decades, the effects of sedentary lifestyles have emerged as a critical aspect of modern society. Interestingly, recent evidence demonstrated that physical exercise plays an important role not only in maintaining peripheral health but also in the regulation of central nervous system function. Many studies have shown that physical exercise promotes the release of molecules, involved in neuronal survival, differentiation, plasticity and neurogenesis, from several peripheral organs. Thus, aerobic exercise has emerged as an intriguing tool that, on one hand, could serve as a therapeutic protocol for diseases of the nervous system, and on the other hand, could help to unravel potential molecular targets for pharmacological approaches. In the present review, we will summarize the cellular interactions that mediate the effects of physical exercise on brain health, starting from the factors released in myocytes during muscle contraction to the cellular pathways that regulate higher cognitive functions, in both health and disease.
Collapse
Affiliation(s)
- Alan Consorti
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
- NEUROFARBA, University of Florence, Florence, Italy
| | | | | |
Collapse
|
38
|
HSP60 reduction protects against diet-induced obesity by modulating energy metabolism in adipose tissue. Mol Metab 2021; 53:101276. [PMID: 34153520 PMCID: PMC8319365 DOI: 10.1016/j.molmet.2021.101276] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/06/2021] [Accepted: 06/15/2021] [Indexed: 11/29/2022] Open
Abstract
Objective Insulin regulates mitochondrial function, thereby propagating an efficient metabolism. Conversely, diabetes and insulin resistance are linked to mitochondrial dysfunction with a decreased expression of the mitochondrial chaperone HSP60. The aim of this investigation was to determine the effect of a reduced HSP60 expression on the development of obesity and insulin resistance. Methods Control and heterozygous whole-body HSP60 knockout (Hsp60+/−) mice were fed a high-fat diet (HFD, 60% calories from fat) for 16 weeks and subjected to extensive metabolic phenotyping. To understand the effect of HSP60 on white adipose tissue, microarray analysis of gonadal WAT was performed, ex vivo experiments were performed, and a lentiviral knockdown of HSP60 in 3T3-L1 cells was conducted to gain detailed insights into the effect of reduced HSP60 levels on adipocyte homeostasis. Results Male Hsp60+/− mice exhibited lower body weight with lower fat mass. These mice exhibited improved insulin sensitivity compared to control, as assessed by Matsuda Index and HOMA-IR. Accordingly, insulin levels were significantly reduced in Hsp60+/− mice in a glucose tolerance test. However, Hsp60+/− mice exhibited an altered adipose tissue metabolism with elevated insulin-independent glucose uptake, adipocyte hyperplasia in the presence of mitochondrial dysfunction, altered autophagy, and local insulin resistance. Conclusions We discovered that the reduction of HSP60 in mice predominantly affects adipose tissue homeostasis, leading to beneficial alterations in body weight, body composition, and adipocyte morphology, albeit exhibiting local insulin resistance. Mice with reduced HSP60 levels are protected from diet-induced obesity. Hsp60+/− mice exhibit altered adipose tissue energy metabolism. WAT of Hsp60+/− mice exhibit elevated insulin-independent glucose uptake. Hsp60+/− mice show improved global, but impaired WAT insulin action.
Collapse
|
39
|
Rana T, Behl T, Sehgal A, Mehta V, Singh S, Sharma N, Bungau S. Elucidating the Possible Role of FoxO in Depression. Neurochem Res 2021; 46:2761-2775. [PMID: 34075521 DOI: 10.1007/s11064-021-03364-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 12/21/2022]
Abstract
Forkhead box-O (FoxO) transcriptional factors perform essential functions in several physiological and biological processes. Recent studies have shown that FoxO is implicated in the pathophysiology of depression. Changes in the upstream mediators of FoxOs including brain-derived neurotrophic factor (BDNF) and protein kinase B have been associated with depressive disorder and the antidepressant agents are known to alter the phosphorylation of FoxOs. Moreover, FoxOs might be regulated by serotonin or noradrenaline signaling and the hypothalamic-pituitary-adrenal (HPA)-axis,both of them are associated with the development of the depressive disorder. FoxO also regulates neural morphology, synaptogenesis, and neurogenesis in the hippocampus, which accounts for the pathogenesis of the depressive disorder. The current article underlined the potential functions of FoxOs in the etiology of depressive disorder and formulate few essential proposals for further investigation. The review also proposes that FoxO and its signal pathway might establish possible therapeutic mediators for the management of depressive disorder.
Collapse
Affiliation(s)
- Tarapati Rana
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India.,Government Pharmacy College, Seraj, Mandi, Himachal Pradesh, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Vineet Mehta
- Government College of Pharmacy, Rohru, Distt., Shimla, Himachal Pradesh, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
40
|
Onesto MM, Short CA, Rempel SK, Catlett TS, Gomez TM. Growth Factors as Axon Guidance Molecules: Lessons From in vitro Studies. Front Neurosci 2021; 15:678454. [PMID: 34093120 PMCID: PMC8175860 DOI: 10.3389/fnins.2021.678454] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
Growth cones at the tips of extending axons navigate through developing organisms by probing extracellular cues, which guide them through intermediate steps and onto final synaptic target sites. Widespread focus on a few guidance cue families has historically overshadowed potentially crucial roles of less well-studied growth factors in axon guidance. In fact, recent evidence suggests that a variety of growth factors have the ability to guide axons, affecting the targeting and morphogenesis of growth cones in vitro. This review summarizes in vitro experiments identifying responses and signaling mechanisms underlying axon morphogenesis caused by underappreciated growth factors.
Collapse
Affiliation(s)
| | | | | | | | - Timothy M. Gomez
- Neuroscience Training Program and Cell and Molecular Biology Program, Department of Neuroscience, University of Wisconsin–Madison, Madison, WI, United States
| |
Collapse
|
41
|
Shandilya A, Mehan S. Dysregulation of IGF-1/GLP-1 signaling in the progression of ALS: potential target activators and influences on neurological dysfunctions. Neurol Sci 2021; 42:3145-3166. [PMID: 34018075 DOI: 10.1007/s10072-021-05328-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/17/2021] [Indexed: 12/31/2022]
Abstract
The prominent causes for motor neuron diseases like ALS are demyelination, immune dysregulation, and neuroinflammation. Numerous research studies indicate that the downregulation of IGF-1 and GLP-1 signaling pathways plays a significant role in the progression of ALS pathogenesis and other neurological disorders. In the current review, we discussed the dysregulation of IGF-1/GLP-1 signaling in neurodegenerative manifestations of ALS like a genetic anomaly, oligodendrocyte degradation, demyelination, glial overactivation, immune deregulation, and neuroexcitation. In addition, the current review reveals the IGF-1 and GLP-1 activators based on the premise that the restoration of abnormal IGF-1/GLP-1 signaling could result in neuroprotection and neurotrophic effects for the clinical-pathological presentation of ALS and other brain diseases. Thus, the potential benefits of IGF-1/GLP-1 signal upregulation in the development of disease-modifying therapeutic strategies may prevent ALS and associated neurocomplications.
Collapse
Affiliation(s)
- Ambika Shandilya
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
42
|
Yuan T, Ying J, Li C, Jin L, Kang J, Shi Y, Gui S, Liu C, Wang R, Zuo Z, Zhang Y. In Vivo Characterization of Cortical and White Matter Microstructural Pathology in Growth Hormone-Secreting Pituitary Adenoma. Front Oncol 2021; 11:641359. [PMID: 33912457 PMCID: PMC8072046 DOI: 10.3389/fonc.2021.641359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/03/2021] [Indexed: 11/13/2022] Open
Abstract
Background The growth hormone (GH) and insulin-like-growth factor 1 (IGF-1) axis has long been recognized for its critical role in brain growth, development. This study was designed to investigate microstructural pathology in the cortex and white matter in growth hormone-secreting pituitary adenoma, which characterized by excessive secretion of GH and IGF-1. Methods 29 patients with growth hormone-secreting pituitary adenoma (acromegaly) and 31 patients with non-functional pituitary adenoma as controls were recruited and assessed using neuropsychological test, surface-based morphometry, T1/T2-weighted myelin-sensitive magnetic resonance imaging, neurite orientation dispersion and density imaging, and diffusion tensor imaging. Results Compared to controls, we found 1) acromegaly had significantly increased cortical thickness throughout the bilateral cortex (pFDR < 0.05). 2) T1/T2-weighted ratio in the cortex were decreased in the bilateral occipital cortex and pre/postcentral central gyri but increased in the bilateral fusiform, insular, and superior temporal gyri in acromegaly (pFDR < 0.05). 3) T1/T2-weighted ratio were decreased in most bundles, and only a few areas showed increases in acromegaly (pFDR < 0.05). 4) Neurite density index (NDI) was significantly lower throughout the cortex and bundles in acromegaly (pTFCE < 0.05). 5) lower fractional anisotropy (FA) and higher mean diffusivity (MD), axial diffusivity (AD) and radial diffusivity (RD) in extensive bundles in acromegaly (pTFCE < 0.05). 6) microstructural pathology in the cortex and white matter were associated with neuropsychological dysfunction in acromegaly. Conclusions Our findings suggested that long-term persistent and excess serum GH/IGF-1 levels alter the microstructure in the cortex and white matter in acromegaly, which may be responsible for neuropsychological dysfunction.
Collapse
Affiliation(s)
- Taoyang Yuan
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jianyou Ying
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Lu Jin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jie Kang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuanyu Shi
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chunhui Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Rui Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhentao Zuo
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Beijing, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders Brain Tumour Center, China National Clinical Research Center for Neurological Diseases, Key Laboratory of Central Nervous System Injury Research, Beijing, China
| |
Collapse
|
43
|
Long KLP, Breton JM, Barraza MK, Perloff OS, Kaufer D. Hormonal Regulation of Oligodendrogenesis I: Effects across the Lifespan. Biomolecules 2021; 11:biom11020283. [PMID: 33672939 PMCID: PMC7918364 DOI: 10.3390/biom11020283] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 02/07/2023] Open
Abstract
The brain’s capacity to respond to changing environments via hormonal signaling is critical to fine-tuned function. An emerging body of literature highlights a role for myelin plasticity as a prominent type of experience-dependent plasticity in the adult brain. Myelin plasticity is driven by oligodendrocytes (OLs) and their precursor cells (OPCs). OPC differentiation regulates the trajectory of myelin production throughout development, and importantly, OPCs maintain the ability to proliferate and generate new OLs throughout adulthood. The process of oligodendrogenesis, the creation of new OLs, can be dramatically influenced during early development and in adulthood by internal and environmental conditions such as hormones. Here, we review the current literature describing hormonal regulation of oligodendrogenesis within physiological conditions, focusing on several classes of hormones: steroid, peptide, and thyroid hormones. We discuss hormonal regulation at each stage of oligodendrogenesis and describe mechanisms of action, where known. Overall, the majority of hormones enhance oligodendrogenesis, increasing OPC differentiation and inducing maturation and myelin production in OLs. The mechanisms underlying these processes vary for each hormone but may ultimately converge upon common signaling pathways, mediated by specific receptors expressed across the OL lineage. However, not all of the mechanisms have been fully elucidated, and here, we note the remaining gaps in the literature, including the complex interactions between hormonal systems and with the immune system. In the companion manuscript in this issue, we discuss the implications of hormonal regulation of oligodendrogenesis for neurological and psychiatric disorders characterized by white matter loss. Ultimately, a better understanding of the fundamental mechanisms of hormonal regulation of oligodendrogenesis across the entire lifespan, especially in vivo, will progress both basic and translational research.
Collapse
Affiliation(s)
- Kimberly L. P. Long
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; (J.M.B.); (D.K.)
- Correspondence:
| | - Jocelyn M. Breton
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; (J.M.B.); (D.K.)
| | - Matthew K. Barraza
- Department of Molecular and Cellular Biology, University of California, Berkeley, CA 94720, USA;
| | - Olga S. Perloff
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA 94143, USA;
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; (J.M.B.); (D.K.)
- Department of Integrative Biology, University of California, Berkeley, CA 94720, USA
- Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
| |
Collapse
|
44
|
Deficiency of Microglial Autophagy Increases the Density of Oligodendrocytes and Susceptibility to Severe Forms of Seizures. eNeuro 2021; 8:ENEURO.0183-20.2021. [PMID: 33472865 PMCID: PMC7890520 DOI: 10.1523/eneuro.0183-20.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/11/2020] [Accepted: 01/02/2021] [Indexed: 12/14/2022] Open
Abstract
Excessive activation of mTOR in microglia impairs CNS homeostasis and causes severe epilepsy. Autophagy constitutes an important part of mTOR signaling. The contribution of microglial autophagy to CNS homeostasis and epilepsy remains to be determined. Here, we report that ATG7KO mice deficient for autophagy in microglia display a marked increase of myelination markers, a higher density of mature oligodendrocytes (ODCs), and altered lengths of the nodes of Ranvier. Moreover, we found that deficiency of microglial autophagy (ATG7KO) leads to increased seizure susceptibility in three seizure models (pilocarpine, kainic acid, and amygdala kindling). We demonstrated that ATG7KO mice develop severe generalized seizures and display nearly 100% mortality to convulsions induced by pilocarpine and kainic acid. In the amygdala kindling model, we observed significant facilitation of contralateral propagation of seizures, a process underlying the development of generalized seizures. Taken together, our results reveal impaired microglial autophagy as a novel mechanism underlying altered homeostasis of ODCs and increased susceptibility to severe and fatal generalized seizures.
Collapse
|
45
|
Alibazi RJ, Pearce AJ, Rostami M, Frazer AK, Brownstein C, Kidgell DJ. Determining the Intracortical Responses After a Single Session of Aerobic Exercise in Young Healthy Individuals: A Systematic Review and Best Evidence Synthesis. J Strength Cond Res 2021; 35:562-575. [PMID: 33201155 DOI: 10.1519/jsc.0000000000003884] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
ABSTRACT Alibazi, RJ, Pearce, AJ, Rostami, M, Frazer, AK, Brownstein, C, and Kidgell, DJ. Determining the intracortical responses after a single session of aerobic exercise in young healthy individuals: a systematic review and best evidence synthesis. J Strength Cond Res 35(2): 562-575, 2021-A single bout of aerobic exercise (AE) may induce changes in the excitability of the intracortical circuits of the primary motor cortex (M1). Similar to noninvasive brain stimulation techniques, such as transcranial direct current stimulation, AE could be used as a priming technique to facilitate motor learning. This review examined the effect of AE on modulating intracortical excitability and inhibition in human subjects. A systematic review, according to PRISMA guidelines, identified studies by database searching, hand searching, and citation tracking between inception and the last week of February 2020. Methodological quality of included studies was determined using the Downs and Black quality index and Cochrane Collaboration of risk of bias tool. Data were synthesized and analyzed using best-evidence synthesis. There was strong evidence for AE not to change corticospinal excitability and conflicting evidence for increasing intracortical facilitation and reducing silent period and long-interval cortical inhibition. Aerobic exercise did reduce short-interval cortical inhibition, which suggests AE modulates the excitability of the short-latency inhibitory circuits within the M1; however, given the small number of included studies, it remains unclear how AE affects all circuits. In light of the above, AE may have important implications during periods of rehabilitation, whereby priming AE could be used to facilitate motor learning.
Collapse
Affiliation(s)
- Razie J Alibazi
- Non-invasive Brain Stimulation & Neuroplasticity Laboratory, Department of Physiotherapy, School of Primary and Allied Health Care, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria, Australia
| | - Alan J Pearce
- College of Science, Health and Engineering, La Trobe University, Melbourne, Victoria, Australia
| | - Mohamad Rostami
- Department of Physiotherapy, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran; and
| | - Ashlyn K Frazer
- Non-invasive Brain Stimulation & Neuroplasticity Laboratory, Department of Physiotherapy, School of Primary and Allied Health Care, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria, Australia
| | - Callum Brownstein
- University of Lyon, University Jean Monnet Saint-Etienne, Inter-university Laboratory of Human Movement Biology, Saint-Etienne, France
| | - Dawson J Kidgell
- Non-invasive Brain Stimulation & Neuroplasticity Laboratory, Department of Physiotherapy, School of Primary and Allied Health Care, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
46
|
Şimşek F, Işık Ü, Aktepe E, Kılıç F, Şirin FB, Bozkurt M. Comparison of Serum VEGF, IGF-1, and HIF-1α Levels in Children with Autism Spectrum Disorder and Healthy Controls. J Autism Dev Disord 2021; 51:3564-3574. [PMID: 33389301 DOI: 10.1007/s10803-020-04820-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2020] [Indexed: 01/13/2023]
Abstract
The aim of this study was to determine whether serum VEGF, IGF-1, and HIF-1α levels differed between Autism Spectrum Disorder (ASD) patients and healthy controls. A total of 40 children with ASD and 40 healthy controls aged 4-12 years were included. Serum levels of VEGF, IGF-1, and HIF-1α were measured using commercial enzyme-linked immunosorbent assay kits. Serum IGF-1 levels were found to be statistically significantly higher in the ASD group than in the control group. Serum HIF-1α levels were borderline significantly lower in the ASD group. There was no statistically significant difference in serum VEGF levels between the two groups. IGF-1 and HIF-1α may play a potential role in the etiopathogenesis of ASD.
Collapse
Affiliation(s)
- Fulya Şimşek
- Department of Child and Adolescent Psychiatry, Suleyman Demirel University Medicine Faculty, Çünür, East Campus, Isparta, 32260, Turkey
| | - Ümit Işık
- Department of Child and Adolescent Psychiatry, Suleyman Demirel University Medicine Faculty, Çünür, East Campus, Isparta, 32260, Turkey.
| | - Evrim Aktepe
- Department of Child and Adolescent Psychiatry, Suleyman Demirel University Medicine Faculty, Çünür, East Campus, Isparta, 32260, Turkey
| | - Faruk Kılıç
- Department of Psychiatry, Suleyman Demirel University Medicine Faculty, Isparta, Turkey
| | - Fevziye Burcu Şirin
- Department of Biochemistry, Suleyman Demirel University Medicine Faculty, Isparta, Turkey
| | - Mustafa Bozkurt
- Department of Biochemistry, Suleyman Demirel University Medicine Faculty, Isparta, Turkey
| |
Collapse
|
47
|
Guevara-Aguirre J, Torres C, Peña G, Palacios M, Bautista C, Guevara A, Gavilanes AW. IGF-I deficiency and enhanced insulin sensitivity due to a mutated growth hormone receptor gene in humans. Mol Cell Endocrinol 2021; 519:111044. [PMID: 33053393 DOI: 10.1016/j.mce.2020.111044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/19/2022]
Abstract
Human size is achieved by the coordinated expression of many genes. From conception to adulthood, a given genomic endowment is modified by highly variable environmental circumstances. During each stage of a person's life, distinct nutritional and hormonal influences continuously shape growing physical features until mature characteristics are attained. Underlying processes depend on precise provision of substrates and energy extracted by insulin action from nutrients, which allows cell proliferation, differentiation, and survival, under the concerted actions of growth hormone and insulin-like growth factor-I (IGF-I). It should be noted that growth and metabolic signaling pathways are interdependent and superimposed at multiple levels. Attainment of a fully developed human phenotype should be considered as a harmonious increment in body size rather than a simple increase in height. From this perspective we herein analyze adult features of individuals with an inactive growth hormone receptor, who consequently have severely diminished concentrations of serum insulin and endocrine IGF-I.
Collapse
Affiliation(s)
- Jaime Guevara-Aguirre
- Colegio de Ciencias de La Salud, Universidad San Francisco de Quito, Diego de Robles s/n y Pampite, Cumbayá, Quito, Ecuador; Maastricht University, Maastricht, the Netherlands; Instituto de Endocrinología IEMYR, Quito, Ecuador.
| | - Carlos Torres
- Colegio de Ciencias de La Salud, Universidad San Francisco de Quito, Diego de Robles s/n y Pampite, Cumbayá, Quito, Ecuador
| | - Gabriela Peña
- Colegio de Ciencias de La Salud, Universidad San Francisco de Quito, Diego de Robles s/n y Pampite, Cumbayá, Quito, Ecuador
| | - María Palacios
- Colegio de Ciencias de La Salud, Universidad San Francisco de Quito, Diego de Robles s/n y Pampite, Cumbayá, Quito, Ecuador
| | - Camila Bautista
- Colegio de Ciencias de La Salud, Universidad San Francisco de Quito, Diego de Robles s/n y Pampite, Cumbayá, Quito, Ecuador
| | | | | |
Collapse
|
48
|
Vaes JEG, Kosmeijer CM, Kaal M, van Vliet R, Brandt MJV, Benders MJNL, Nijboer CH. Regenerative Therapies to Restore Interneuron Disturbances in Experimental Models of Encephalopathy of Prematurity. Int J Mol Sci 2020; 22:ijms22010211. [PMID: 33379239 PMCID: PMC7795049 DOI: 10.3390/ijms22010211] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/22/2022] Open
Abstract
Encephalopathy of Prematurity (EoP) is a major cause of morbidity in (extreme) preterm neonates. Though the majority of EoP research has focused on failure of oligodendrocyte maturation as an underlying pathophysiological mechanism, recent pioneer work has identified developmental disturbances in inhibitory interneurons to contribute to EoP. Here we investigated interneuron abnormalities in two experimental models of EoP and explored the potential of two promising treatment strategies, namely intranasal mesenchymal stem cells (MSCs) or insulin-like growth factor I (IGF1), to restore interneuron development. In rats, fetal inflammation and postnatal hypoxia led to a transient increase in total cortical interneuron numbers, with a layer-specific deficit in parvalbumin (PV)+ interneurons. Additionally, a transient excess of total cortical cell density was observed, including excitatory neuron numbers. In the hippocampal cornu ammonis (CA) 1 region, long-term deficits in total interneuron numbers and PV+ subtype were observed. In mice subjected to postnatal hypoxia/ischemia and systemic inflammation, total numbers of cortical interneurons remained unaffected; however, subtype analysis revealed a global, transient reduction in PV+ cells and a long-lasting layer-specific increase in vasoactive intestinal polypeptide (VIP)+ cells. In the dentate gyrus, a long-lasting deficit of somatostatin (SST)+ cells was observed. Both intranasal MSC and IGF1 therapy restored the majority of interneuron abnormalities in EoP mice. In line with the histological findings, EoP mice displayed impaired social behavior, which was partly restored by the therapies. In conclusion, induction of experimental EoP is associated with model-specific disturbances in interneuron development. In addition, intranasal MSCs and IGF1 are promising therapeutic strategies to aid interneuron development after EoP.
Collapse
Affiliation(s)
- Josine E. G. Vaes
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands; (J.E.G.V.); (C.M.K.); (M.K.); (R.v.V.); (M.J.V.B.)
- Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands;
| | - Chantal M. Kosmeijer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands; (J.E.G.V.); (C.M.K.); (M.K.); (R.v.V.); (M.J.V.B.)
| | - Marthe Kaal
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands; (J.E.G.V.); (C.M.K.); (M.K.); (R.v.V.); (M.J.V.B.)
| | - Rik van Vliet
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands; (J.E.G.V.); (C.M.K.); (M.K.); (R.v.V.); (M.J.V.B.)
| | - Myrna J. V. Brandt
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands; (J.E.G.V.); (C.M.K.); (M.K.); (R.v.V.); (M.J.V.B.)
| | - Manon J. N. L. Benders
- Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands;
| | - Cora H. Nijboer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands; (J.E.G.V.); (C.M.K.); (M.K.); (R.v.V.); (M.J.V.B.)
- Correspondence: ; Tel.: +31-88-755-4360
| |
Collapse
|
49
|
Role of Oligodendrocytes and Myelin in the Pathophysiology of Autism Spectrum Disorder. Brain Sci 2020; 10:brainsci10120951. [PMID: 33302549 PMCID: PMC7764453 DOI: 10.3390/brainsci10120951] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/12/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is an early neurodevelopmental disorder that involves deficits in interpersonal communication, social interaction, and repetitive behaviors. Although ASD pathophysiology is still uncertain, alterations in the abnormal development of the frontal lobe, limbic areas, and putamen generate an imbalance between inhibition and excitation of neuronal activity. Interestingly, recent findings suggest that a disruption in neuronal connectivity is associated with neural alterations in white matter production and myelination in diverse brain regions of patients with ASD. This review is aimed to summarize the most recent evidence that supports the notion that abnormalities in the oligodendrocyte generation and axonal myelination in specific brain regions are involved in the pathophysiology of ASD. Fundamental molecular mediators of these pathological processes are also examined. Determining the role of alterations in oligodendrogenesis and myelination is a fundamental step to understand the pathophysiology of ASD and identify possible therapeutic targets.
Collapse
|
50
|
Vaes JEG, Brandt MJV, Wanders N, Benders MJNL, de Theije CGM, Gressens P, Nijboer CH. The impact of trophic and immunomodulatory factors on oligodendrocyte maturation: Potential treatments for encephalopathy of prematurity. Glia 2020; 69:1311-1340. [PMID: 33595855 PMCID: PMC8246971 DOI: 10.1002/glia.23939] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022]
Abstract
Encephalopathy of prematurity (EoP) is a major cause of morbidity in preterm neonates, causing neurodevelopmental adversities that can lead to lifelong impairments. Preterm birth-related insults, such as cerebral oxygen fluctuations and perinatal inflammation, are believed to negatively impact brain development, leading to a range of brain abnormalities. Diffuse white matter injury is a major hallmark of EoP and characterized by widespread hypomyelination, the result of disturbances in oligodendrocyte lineage development. At present, there are no treatment options available, despite the enormous burden of EoP on patients, their families, and society. Over the years, research in the field of neonatal brain injury and other white matter pathologies has led to the identification of several promising trophic factors and cytokines that contribute to the survival and maturation of oligodendrocytes, and/or dampening neuroinflammation. In this review, we discuss the current literature on selected factors and their therapeutic potential to combat EoP, covering a wide range of in vitro, preclinical and clinical studies. Furthermore, we offer a future perspective on the translatability of these factors into clinical practice.
Collapse
Affiliation(s)
- Josine E G Vaes
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands.,Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Myrna J V Brandt
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Nikki Wanders
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Caroline G M de Theije
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | | | - Cora H Nijboer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|