1
|
Wei J, Wang M, Li S, Han R, Xu W, Zhao A, Yu Q, Li H, Li M, Chi G. Reprogramming of astrocytes and glioma cells into neurons for central nervous system repair and glioblastoma therapy. Biomed Pharmacother 2024; 176:116806. [PMID: 38796971 DOI: 10.1016/j.biopha.2024.116806] [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: 02/09/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
Central nervous system (CNS) damage is usually irreversible owing to the limited regenerative capability of neurons. Following CNS injury, astrocytes are reactively activated and are the key cells involved in post-injury repair mechanisms. Consequently, research on the reprogramming of reactive astrocytes into neurons could provide new directions for the restoration of neural function after CNS injury and in the promotion of recovery in various neurodegenerative diseases. This review aims to provide an overview of the means through which reactive astrocytes around lesions can be reprogrammed into neurons, to elucidate the intrinsic connection between the two cell types from a neurogenesis perspective, and to summarize what is known about the neurotranscription factors, small-molecule compounds and MicroRNA that play major roles in astrocyte reprogramming. As the malignant proliferation of astrocytes promotes the development of glioblastoma multiforme (GBM), this review also examines the research advances on and the theoretical basis for the reprogramming of GBM cells into neurons and discusses the advantages of such approaches over traditional treatment modalities. This comprehensive review provides new insights into the field of GBM therapy and theoretical insights into the mechanisms of neurological recovery following neurological injury and in GBM treatment.
Collapse
Affiliation(s)
- Junyuan Wei
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Miaomiao Wang
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Shilin Li
- School of Public Health, Jilin University, Changchun 130021, China.
| | - Rui Han
- Department of Neurovascular Surgery, First Hospital of Jilin University, 1xinmin Avenue, Changchun, Jilin Province 130021, China.
| | - Wenhong Xu
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Anqi Zhao
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Qi Yu
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Haokun Li
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
2
|
Cornejo F, Franchini N, Cortés BI, Elgueta D, Cancino GI. Neural conditional ablation of the protein tyrosine phosphatase receptor Delta PTPRD impairs gliogenesis in the developing mouse brain cortex. Front Cell Dev Biol 2024; 12:1357862. [PMID: 38487272 PMCID: PMC10937347 DOI: 10.3389/fcell.2024.1357862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/12/2024] [Indexed: 03/17/2024] Open
Abstract
Neurodevelopmental disorders are characterized by alterations in the development of the cerebral cortex, including aberrant changes in the number and function of neural cells. Although neurogenesis is one of the most studied cellular processes in these pathologies, little evidence is known about glial development. Genetic association studies have identified several genes associated with neurodevelopmental disorders. Indeed, variations in the PTPRD gene have been associated with numerous brain disorders, including autism spectrum disorder, restless leg syndrome, and schizophrenia. We previously demonstrated that constitutive loss of PTPRD expression induces significant alterations in cortical neurogenesis, promoting an increase in intermediate progenitors and neurons in mice. However, its role in gliogenesis has not been evaluated. To assess this, we developed a conditional knockout mouse model lacking PTPRD expression in telencephalon cells. Here, we found that the lack of PTPRD in the mouse cortex reduces glial precursors, astrocytes, and oligodendrocytes. According to our results, this decrease in gliogenesis resulted from a reduced number of radial glia cells at gliogenesis onset and a lower gliogenic potential in cortical neural precursors due to less activation of the JAK/STAT pathway and reduced expression of gliogenic genes. Our study shows PTPRD as a regulator of the glial/neuronal balance during cortical neurodevelopment and highlights the importance of studying glial development to understand the etiology of neurodevelopmental diseases.
Collapse
Affiliation(s)
- Francisca Cornejo
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Nayhara Franchini
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Bastián I. Cortés
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Elgueta
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gonzalo I. Cancino
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
3
|
Zhang G, Ren Q, Lin Y, Zhou D, Huang L, Li W, Chang H, Huang G, Li Z, Yan J. Parental folic acid deficiency delays neurobehavioral development in rat offspring by inhibiting the differentiation of neural stem cells into neurons. J Nutr Biochem 2023; 122:109455. [PMID: 37788724 DOI: 10.1016/j.jnutbio.2023.109455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/29/2023] [Accepted: 09/27/2023] [Indexed: 10/05/2023]
Abstract
Maternal folate status during pregnancy is associated with the neurodevelopment of offspring; however, study results on the association between paternal folate status and offspring neurodevelopment are inconsistent. This study aimed to explore whether parental folic acid deficiency affects the neurobehavioral development of offspring by affecting the differentiation of neural stem cells (NSCs) into neurons. In the present study, the offspring were divided into four groups: parental folic acid deficient group (D-D), maternal folic acid deficient and paternal folic acid normal group (D-N), maternal folic acid normal and paternal folic acid deficient group (N-D), and parental folic acid normal group (N-N). For in vivo study, neurobehavioral indexes, and neuron-specific nuclear protein (NeuN) and glial fibrillary acidic protein (GFAP) expression in the brain hippocampus and cerebral cortex of offspring were measured at different time points. For in vitro study, NSCs were cultured from the hippocampus and striatum, and neuronal and astrocytic differentiation were measured. The results demonstrated that parental folic acid deficiency decreased the brain folate level in offspring, delayed early sensory-motor reflex development, impaired spatial learning and memory ability in adolescence and adulthood, decreased differentiation of NSCs into neurons and increased differentiation of NSCs into astrocytes in vivo and in vitro. These impacts on the neurodevelopment of offspring were most pronounced in D-D group, followed by D-N group and N-D group. In conclusion, parental folic acid deficiency inhibits the neurobehavioral development of offspring, possibly by inhibiting the differentiation of NSCs into neurons.
Collapse
Affiliation(s)
- Guoquan Zhang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Qinghan Ren
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Ying Lin
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Children's Hospital, Tianjin, China
| | - Dezheng Zhou
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Li Huang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Wen Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Hong Chang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Guowei Huang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Zhenshu Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China.
| | - Jing Yan
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China; Department of Social Medicine and Health Administration, School of Public Health, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
4
|
Sarapultsev A, Gusev E, Komelkova M, Utepova I, Luo S, Hu D. JAK-STAT signaling in inflammation and stress-related diseases: implications for therapeutic interventions. MOLECULAR BIOMEDICINE 2023; 4:40. [PMID: 37938494 PMCID: PMC10632324 DOI: 10.1186/s43556-023-00151-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/26/2023] [Indexed: 11/09/2023] Open
Abstract
The Janus kinase-signal transducer and transcription activator pathway (JAK-STAT) serves as a cornerstone in cellular signaling, regulating physiological and pathological processes such as inflammation and stress. Dysregulation in this pathway can lead to severe immunodeficiencies and malignancies, and its role extends to neurotransduction and pro-inflammatory signaling mechanisms. Although JAK inhibitors (Jakinibs) have successfully treated immunological and inflammatory disorders, their application has generally been limited to diseases with similar pathogenic features. Despite the modest expression of JAK-STAT in the CNS, it is crucial for functions in the cortex, hippocampus, and cerebellum, making it relevant in conditions like Parkinson's disease and other neuroinflammatory disorders. Furthermore, the influence of the pathway on serotonin receptors and phospholipase C has implications for stress and mood disorders. This review expands the understanding of JAK-STAT, moving beyond traditional immunological contexts to explore its role in stress-related disorders and CNS function. Recent findings, such as the effectiveness of Jakinibs in chronic conditions such as rheumatoid arthritis, expand their therapeutic applicability. Advances in isoform-specific inhibitors, including filgotinib and upadacitinib, promise greater specificity with fewer off-target effects. Combination therapies, involving Jakinibs and monoclonal antibodies, aiming to enhance therapeutic specificity and efficacy also give great hope. Overall, this review bridges the gap between basic science and clinical application, elucidating the complex influence of the JAK-STAT pathway on human health and guiding future interventions.
Collapse
Affiliation(s)
- Alexey Sarapultsev
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080, Chelyabinsk, Russia.
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049, Ekaterinburg, Russia.
| | - Evgenii Gusev
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080, Chelyabinsk, Russia
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049, Ekaterinburg, Russia
| | - Maria Komelkova
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080, Chelyabinsk, Russia
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049, Ekaterinburg, Russia
| | - Irina Utepova
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049, Ekaterinburg, Russia
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 620002, Ekaterinburg, Russian Federation
| | - Shanshan Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, 430022, China
- Clinical Research Center of Cancer Immunotherapy, Hubei Wuhan, 430022, China
| |
Collapse
|
5
|
Huang T, Fakurazi S, Cheah PS, Ling KH. REST Targets JAK-STAT and HIF-1 Signaling Pathways in Human Down Syndrome Brain and Neural Cells. Int J Mol Sci 2023; 24:9980. [PMID: 37373133 DOI: 10.3390/ijms24129980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Down syndrome (DS) is the most frequently diagnosed chromosomal disorder of chromosome 21 (HSA21) aneuploidy, characterized by intellectual disability and reduced lifespan. The transcription repressor, Repressor Element-1 Silencing Transcription factor (REST), which acts as an epigenetic regulator, is a crucial regulator of neuronal and glial gene expression. In this study, we identified and investigated the role of REST-target genes in human brain tissues, cerebral organoids, and neural cells in Down syndrome. Gene expression datasets generated from healthy controls and DS samples of human brain tissues, cerebral organoids, NPC, neurons, and astrocytes were retrieved from the Gene Ontology (GEO) and Sequence Read Archive (SRA) databases. Differential expression analysis was performed on all datasets to produce differential expression genes (DEGs) between DS and control groups. REST-targeted DEGs were subjected to functional ontologies, pathways, and network analyses. We found that REST-targeted DEGs in DS were enriched for the JAK-STAT and HIF-1 signaling pathways across multiple distinct brain regions, ages, and neural cell types. We also identified REST-targeted DEGs involved in nervous system development, cell differentiation, fatty acid metabolism and inflammation in the DS brain. Based on the findings, we propose REST as the critical regulator and a promising therapeutic target to modulate homeostatic gene expression in the DS brain.
Collapse
Affiliation(s)
- Tan Huang
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Pike-See Cheah
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - King-Hwa Ling
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Malaysian Research Institute on Ageing (MyAgeingTM), Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
6
|
Hydroxychloroquine lowers Alzheimer's disease and related dementias risk and rescues molecular phenotypes related to Alzheimer's disease. Mol Psychiatry 2023; 28:1312-1326. [PMID: 36577843 PMCID: PMC10005941 DOI: 10.1038/s41380-022-01912-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/29/2022]
Abstract
We recently nominated cytokine signaling through the Janus-kinase-signal transducer and activator of transcription (JAK/STAT) pathway as a potential AD drug target. As hydroxychloroquine (HCQ) has recently been shown to inactivate STAT3, we hypothesized that it may impact AD pathogenesis and risk. Among 109,124 rheumatoid arthritis patients from routine clinical care, HCQ initiation was associated with a lower risk of incident AD compared to methotrexate initiation across 4 alternative analyses schemes addressing specific types of biases including informative censoring, reverse causality, and outcome misclassification (hazard ratio [95% confidence interval] of 0.92 [0.83-1.00], 0.87 [0.81-0.93], 0.84 [0.76-0.93], and 0.87 [0.75-1.01]). We additionally show that HCQ exerts dose-dependent effects on late long-term potentiation (LTP) and rescues impaired hippocampal synaptic plasticity prior to significant accumulation of amyloid plaques and neurodegeneration in APP/PS1 mice. Additionally, HCQ treatment enhances microglial clearance of Aβ1-42, lowers neuroinflammation, and reduces tau phosphorylation in cell culture-based phenotypic assays. Finally, we show that HCQ inactivates STAT3 in microglia, neurons, and astrocytes suggesting a plausible mechanism associated with its observed effects on AD pathogenesis. HCQ, a relatively safe and inexpensive drug in current use may be a promising disease-modifying AD treatment. This hypothesis merits testing through adequately powered clinical trials in at-risk individuals during preclinical stages of disease progression.
Collapse
|
7
|
Liu Y, Li S, Liu D, Wei H, Wang X, Yan F. Exploration of the potential mechanism of Pushen capsule in the treatment of vascular dementia based on network pharmacology and experimental verification. JOURNAL OF ETHNOPHARMACOLOGY 2022; 298:115632. [PMID: 35964821 DOI: 10.1016/j.jep.2022.115632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/03/2022] [Accepted: 08/06/2022] [Indexed: 06/15/2023]
Abstract
HEADINGS ETHNOPHARMACOLOGICAL RELEVANCE Pushen capsule is a traditional Chinese medicine compound functioning as 'stimulating blood circulation to remove blood stasis', which widely used to treat hyperlipidemia. Recent clinical research showed that Pushen capsule ameliorated cognitive function in patients with vascular mild cognitive impairment. AIM OF THE STUDY Explore the potential mechanism of Pushen capsule in vascular dementia (VaD) using network pharmacology analysis and experimental verification. MATERIALS AND METHODS Active ingredients and their related targets of Pushen capsule, and VaD-related targets were searched in public databases. Core targets, potential functions and mechanisms of Pushen capsule on VaD were predicted by protein-protein interaction (PPI), Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. In vivo experiments were conducted to demonstrate the potential mechanisms of Pushen capsule in the treatment of VaD. RESULTS In total, 155 active ingredients, 273 related targets of Pushen capsule, and 1035 VaD-related targets were selected from the public databases. 147 common targets of Pushen capsule against VaD were obtained. The PPI network, GO and KEGG enrichment analyses revealed that some core targets and signaling pathways are related to inflammation. The experimental results showed that Pushen capsule treatment largely alleviated hippocampal glial activation, accelerated the polarization of activated microglia from the M1 to the M2 phenotype and reduced associated inflammatory factor expression to protect against VaD-induced neuronal loss, synaptic protein reduction and cognitive defects in a dose-dependent manner. Moreover, Pushen capsule reduced the mRNA expression of NF-κB p65; and STAT1. CONCLUSION Our study demonstrates that Pushen capsule alleviates hippocampal neuroinflammation to protect against VaD-induced cognitive impairment in a dose-dependent manner. The neuroprotective effect of Pushen capsule on VaD might be regulated by the NF-κB; and JAK-STAT pathway.
Collapse
Affiliation(s)
- Yun Liu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Shuo Li
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Dandan Liu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, China; The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Jiangsu, 212300, China
| | - Hong Wei
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xingzhi Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Fuling Yan
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, China; Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
8
|
Enhanced Cognition and Neurogenesis in miR-146b Deficient Mice. Cells 2022; 11:cells11132002. [PMID: 35805086 PMCID: PMC9265316 DOI: 10.3390/cells11132002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
The miR-146 family consists of two microRNAs (miRNAs), miR-146a and miR-146b, which are both known to suppress a variety of immune responses. Here in this study, we show that miR-146b is abundantly expressed in neuronal cells, while miR-146a is mainly expressed in microglia and astroglia of adult mice. Accordingly, miR-146b deficient (Mir146b-/-) mice exhibited anxiety-like behaviors and enhanced cognition. Characterization of cellular composition of Mir146b-/- mice using flow cytometry revealed an increased number of neurons and a decreased abundancy of astroglia in the hippocampus and frontal cortex, whereas microglia abundancy remained unchanged. Immunohistochemistry showed a higher density of neurons in the frontal cortex of Mir146b-/- mice, enhanced hippocampal neurogenesis as evidenced by an increased proliferation, and survival of newly generated cells with enhanced maturation into neuronal phenotype. No microglial activation or signs of neuroinflammation were observed in Mir146b-/- mice. Further analysis demonstrated that miR-146b deficiency is associated with elevated expression of glial cell line-derived neurotrophic factor (Gdnf) mRNA in the hippocampus, which might be at least in part responsible for the observed neuronal expansion and the behavioral phenotype. This hypothesis is partially supported by the positive correlation between performance of mice in the object recognition test and Gdnf mRNA expression in Mir146b-/- mice. Together, these results show the distinct function of miR-146b in controlling behaviors and provide new insights in understanding cell-specific function of miR-146b in the neuronal and astroglial organization of the mouse brain.
Collapse
|
9
|
Prutton KM, Marentette JO, Leifheit BA, Esquer H, LaBarbera DV, Anderson CC, Maclean KN, Roede JR. Oxidative stress as a candidate mechanism for accelerated neuroectodermal differentiation due to trisomy 21. Free Radic Biol Med 2022; 186:32-42. [PMID: 35537597 DOI: 10.1016/j.freeradbiomed.2022.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/21/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022]
Abstract
The ubiquity of cognitive deficits and early onset Alzheimer's disease in Down syndrome (DS) has focused much DS iPSC-based research on neuron degeneration and regeneration. Despite reports of elevated oxidative stress in DS brains, few studies assess the impact of this oxidative burden on iPSC differentiation. Here, we evaluate cellular specific redox differences in DS and euploid iPSCs and neural progenitor cells (NPCs) during critical intermediate stages of differentiation. Despite successful generation of NPCs, our results indicate accelerated neuroectodermal differentiation of DS iPSCs compared to isogenic, euploid controls. Specifically, DS embryoid bodies (EBs) and neural rosettes prematurely develop with distinct morphological differences from controls. Additionally, we observed developmental stage-specific alterations in mitochondrial superoxide production and SOD1/2 abundance, coupled with modulations in thioredoxin, thioredoxin reductase, and peroxiredoxin isoforms. Disruption of intracellular redox state and its associated signaling has the potential to disrupt cellular differentiation and development in DS lending to DS-specific phenotypes.
Collapse
Affiliation(s)
- Kendra M Prutton
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, 80045, USA; Linda Crnic Institute for Down Syndrome, Aurora, CO, 80045, USA
| | - John O Marentette
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, 80045, USA; Linda Crnic Institute for Down Syndrome, Aurora, CO, 80045, USA
| | - Brice A Leifheit
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, 80045, USA
| | - Hector Esquer
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, 80045, USA; Center for Drug Discovery, University of Colorado, Aurora, CO, 80045, USA
| | - Daniel V LaBarbera
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, 80045, USA; Center for Drug Discovery, University of Colorado, Aurora, CO, 80045, USA
| | - Colin C Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, 80045, USA; Linda Crnic Institute for Down Syndrome, Aurora, CO, 80045, USA
| | - Kenneth N Maclean
- Linda Crnic Institute for Down Syndrome, Aurora, CO, 80045, USA; Department of Pediatrics, School of Medicine, University of Colorado, Aurora, CO, 80045, USA
| | - James R Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, 80045, USA; Linda Crnic Institute for Down Syndrome, Aurora, CO, 80045, USA.
| |
Collapse
|
10
|
Stagni F, Bartesaghi R. The Challenging Pathway of Treatment for Neurogenesis Impairment in Down Syndrome: Achievements and Perspectives. Front Cell Neurosci 2022; 16:903729. [PMID: 35634470 PMCID: PMC9130961 DOI: 10.3389/fncel.2022.903729] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/19/2022] [Indexed: 12/17/2022] Open
Abstract
Down syndrome (DS), also known as trisomy 21, is a genetic disorder caused by triplication of Chromosome 21. Gene triplication may compromise different body functions but invariably impairs intellectual abilities starting from infancy. Moreover, after the fourth decade of life people with DS are likely to develop Alzheimer’s disease. Neurogenesis impairment during fetal life stages and dendritic pathology emerging in early infancy are thought to be key determinants of alterations in brain functioning in DS. Although the progressive improvement in medical care has led to a notable increase in life expectancy for people with DS, there are currently no treatments for intellectual disability. Increasing evidence in mouse models of DS reveals that pharmacological interventions in the embryonic and neonatal periods may greatly benefit brain development and cognitive performance. The most striking results have been obtained with pharmacotherapies during embryonic life stages, indicating that it is possible to pharmacologically rescue the severe neurodevelopmental defects linked to the trisomic condition. These findings provide hope that similar benefits may be possible for people with DS. This review summarizes current knowledge regarding (i) the scope and timeline of neurogenesis (and dendritic) alterations in DS, in order to delineate suitable windows for treatment; (ii) the role of triplicated genes that are most likely to be the key determinants of these alterations, in order to highlight possible therapeutic targets; and (iii) prenatal and neonatal treatments that have proved to be effective in mouse models, in order to rationalize the choice of treatment for human application. Based on this body of evidence we will discuss prospects and challenges for fetal therapy in individuals with DS as a potential means of drastically counteracting the deleterious effects of gene triplication.
Collapse
Affiliation(s)
- Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- *Correspondence: Renata Bartesaghi,
| |
Collapse
|
11
|
Sanchez-Petidier M, Guerri C, Moreno-Manzano V. Toll-like receptors 2 and 4 differentially regulate the self-renewal and differentiation of spinal cord neural precursor cells. Stem Cell Res Ther 2022; 13:117. [PMID: 35314006 PMCID: PMC8935849 DOI: 10.1186/s13287-022-02798-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 03/07/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Toll-like receptors (TLRs) represent critical effectors in the host defense response against various pathogens; however, their known function during development has also highlighted a potential role in cell fate determination and neural differentiation. While glial cells and neural precursor cells (NPCs) of the spinal cord express both TLR2 and TLR4, their influence on self-renewal and cell differentiation remains incompletely described. METHODS TLR2, TLR4 knock-out and the wild type mice were employed for spinal cord tissue analysis and NPCs isolation at early post-natal stage. Sox2, FoxJ1 and Ki67 expression among others served to identify the undifferentiated and proliferative NPCs; GFAP, Olig2 and β-III-tubulin markers served to identify astrocytes, oligodendrocytes and neurons respectively after NPC spontaneous differentiation. Multiple comparisons were analyzed using one-way ANOVA, with appropriate corrections such as Tukey's post hoc tests used for comparisons. RESULTS We discovered that the deletion of TLR2 or TLR4 significantly reduced the number of Sox2-expressing NPCs in the neonatal mouse spinal cord. While TLR2-knockout NPCs displayed enhanced self-renewal, increased proliferation and apoptosis, and delayed neural differentiation, the absence of TLR4 promoted the neural differentiation of NPCs without affecting proliferation, producing long projecting neurons. TLR4 knock-out NPCs showed significantly higher expression of Neurogenin1, that would be involved in the activation of this neurogenic program by a ligand and microenvironment-independent mechanism. Interestingly, the absence of both TLR2 and TLR4, which induces also a significant reduction in the expression of TLR1, in NPCs impeded oligodendrocyte precursor cell maturation to a similar degree. CONCLUSIONS Our data suggest that Toll-like receptors are needed to maintain Sox2 positive neural progenitors in the spinal cord, however possess distinct regulatory roles in mouse neonatal spinal cord NPCs-while TLR2 and TLR4 play a similar role in oligodendrocytic differentiation, they differentially influence neural differentiation.
Collapse
Affiliation(s)
- Marina Sanchez-Petidier
- Neuronal and Tissue Regeneration Laboratory, Prince Felipe Research Institute, Valencia, Spain.,Neuropathology Laboratory, Prince Felipe Research Institute, Valencia, Spain
| | - Consuelo Guerri
- Neuropathology Laboratory, Prince Felipe Research Institute, Valencia, Spain.
| | - Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Laboratory, Prince Felipe Research Institute, Valencia, Spain.
| |
Collapse
|
12
|
Mollo N, Esposito M, Aurilia M, Scognamiglio R, Accarino R, Bonfiglio F, Cicatiello R, Charalambous M, Procaccini C, Micillo T, Genesio R, Calì G, Secondo A, Paladino S, Matarese G, Vita GD, Conti A, Nitsch L, Izzo A. Human Trisomic iPSCs from Down Syndrome Fibroblasts Manifest Mitochondrial Alterations Early during Neuronal Differentiation. BIOLOGY 2021; 10:biology10070609. [PMID: 34209429 PMCID: PMC8301075 DOI: 10.3390/biology10070609] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND The presence of mitochondrial alterations in Down syndrome suggests that it might affect neuronal differentiation. We established a model of trisomic iPSCs, differentiating into neural precursor cells (NPCs) to monitor the occurrence of differentiation defects and mitochondrial dysfunction. METHODS Isogenic trisomic and euploid iPSCs were differentiated into NPCs in monolayer cultures using the dual-SMAD inhibition protocol. Expression of pluripotency and neural differentiation genes was assessed by qRT-PCR and immunofluorescence. Meta-analysis of expression data was performed on iPSCs. Mitochondrial Ca2+, reactive oxygen species (ROS) and ATP production were investigated using fluorescent probes. Oxygen consumption rate (OCR) was determined by Seahorse Analyzer. RESULTS NPCs at day 7 of induction uniformly expressed the differentiation markers PAX6, SOX2 and NESTIN but not the stemness marker OCT4. At day 21, trisomic NPCs expressed higher levels of typical glial differentiation genes. Expression profiles indicated that mitochondrial genes were dysregulated in trisomic iPSCs. Trisomic NPCs showed altered mitochondrial Ca2+, reduced OCR and ATP synthesis, and elevated ROS production. CONCLUSIONS Human trisomic iPSCs can be rapidly and efficiently differentiated into NPC monolayers. The trisomic NPCs obtained exhibit greater glial-like differentiation potential than their euploid counterparts and manifest mitochondrial dysfunction as early as day 7 of neuronal differentiation.
Collapse
Affiliation(s)
- Nunzia Mollo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (M.E.); (M.A.); (R.S.); (R.A.); (R.C.); (R.G.); (S.P.); (G.M.); (G.D.V.); (A.C.); (L.N.)
| | - Matteo Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (M.E.); (M.A.); (R.S.); (R.A.); (R.C.); (R.G.); (S.P.); (G.M.); (G.D.V.); (A.C.); (L.N.)
| | - Miriam Aurilia
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (M.E.); (M.A.); (R.S.); (R.A.); (R.C.); (R.G.); (S.P.); (G.M.); (G.D.V.); (A.C.); (L.N.)
| | - Roberta Scognamiglio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (M.E.); (M.A.); (R.S.); (R.A.); (R.C.); (R.G.); (S.P.); (G.M.); (G.D.V.); (A.C.); (L.N.)
| | - Rossella Accarino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (M.E.); (M.A.); (R.S.); (R.A.); (R.C.); (R.G.); (S.P.); (G.M.); (G.D.V.); (A.C.); (L.N.)
| | - Ferdinando Bonfiglio
- CEINGE-Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy;
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, 80125 Naples, Italy
| | - Rita Cicatiello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (M.E.); (M.A.); (R.S.); (R.A.); (R.C.); (R.G.); (S.P.); (G.M.); (G.D.V.); (A.C.); (L.N.)
| | - Maria Charalambous
- Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council, 80131 Naples, Italy; (M.C.); (C.P.); (G.C.)
| | - Claudio Procaccini
- Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council, 80131 Naples, Italy; (M.C.); (C.P.); (G.C.)
- Neuroimmunology Unit, IRCCS, Fondazione Santa Lucia, 00143 Rome, Italy;
| | - Teresa Micillo
- Neuroimmunology Unit, IRCCS, Fondazione Santa Lucia, 00143 Rome, Italy;
| | - Rita Genesio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (M.E.); (M.A.); (R.S.); (R.A.); (R.C.); (R.G.); (S.P.); (G.M.); (G.D.V.); (A.C.); (L.N.)
| | - Gaetano Calì
- Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council, 80131 Naples, Italy; (M.C.); (C.P.); (G.C.)
| | - Agnese Secondo
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy;
| | - Simona Paladino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (M.E.); (M.A.); (R.S.); (R.A.); (R.C.); (R.G.); (S.P.); (G.M.); (G.D.V.); (A.C.); (L.N.)
| | - Giuseppe Matarese
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (M.E.); (M.A.); (R.S.); (R.A.); (R.C.); (R.G.); (S.P.); (G.M.); (G.D.V.); (A.C.); (L.N.)
- Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council, 80131 Naples, Italy; (M.C.); (C.P.); (G.C.)
| | - Gabriella De Vita
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (M.E.); (M.A.); (R.S.); (R.A.); (R.C.); (R.G.); (S.P.); (G.M.); (G.D.V.); (A.C.); (L.N.)
| | - Anna Conti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (M.E.); (M.A.); (R.S.); (R.A.); (R.C.); (R.G.); (S.P.); (G.M.); (G.D.V.); (A.C.); (L.N.)
| | - Lucio Nitsch
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (M.E.); (M.A.); (R.S.); (R.A.); (R.C.); (R.G.); (S.P.); (G.M.); (G.D.V.); (A.C.); (L.N.)
- Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council, 80131 Naples, Italy; (M.C.); (C.P.); (G.C.)
| | - Antonella Izzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (M.E.); (M.A.); (R.S.); (R.A.); (R.C.); (R.G.); (S.P.); (G.M.); (G.D.V.); (A.C.); (L.N.)
- Correspondence: ; Tel.: +39-081-746-3237
| |
Collapse
|
13
|
Hosseini S, Michaelsen-Preusse K, Grigoryan G, Chhatbar C, Kalinke U, Korte M. Type I Interferon Receptor Signaling in Astrocytes Regulates Hippocampal Synaptic Plasticity and Cognitive Function of the Healthy CNS. Cell Rep 2021; 31:107666. [PMID: 32433975 DOI: 10.1016/j.celrep.2020.107666] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/23/2020] [Accepted: 04/28/2020] [Indexed: 02/04/2023] Open
Abstract
Type I interferon receptor (IFNAR) signaling is a hallmark of viral control and host protection. Here, we show that, in the hippocampus of healthy IFNAR-deficient mice, synapse number and synaptic plasticity, as well as spatial learning, are impaired. This is also the case for IFN-β-deficient animals. Moreover, antibody-mediated IFNAR blocking acutely interferes with neuronal plasticity, whereas a low-dose application of IFN-β has a positive effect on dendritic spine structure. Interfering with IFNAR signaling in different cell types shows a role for cognitive function and synaptic plasticity specifically mediated by astrocytes. Intriguingly, levels of the astrocytic glutamate-aspartate transporter (GLAST) are reduced significantly upon IFN-β treatment and increase following inhibition of IFNAR signaling. These results indicate that, besides the prominent role for host defense, IFNAR is important for synaptic plasticity as well as cognitive function. Astrocytes are at the center stage of this so-far-unknown signaling cascade.
Collapse
Affiliation(s)
- Shirin Hosseini
- Department of Cellular Neurobiology, Zoological Institute, TU Braunschweig, 38106 Braunschweig, Germany; Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration Group, 38124 Braunschweig, Germany
| | | | - Gayane Grigoryan
- Department of Systemic and Cellular Neurophysiology, Institute for Physiology I, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Chintan Chhatbar
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | - Martin Korte
- Department of Cellular Neurobiology, Zoological Institute, TU Braunschweig, 38106 Braunschweig, Germany; Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration Group, 38124 Braunschweig, Germany.
| |
Collapse
|
14
|
Shi W, Yang F, Dai R, Sun Y, Chu Y, Liao S, Hao B. METTL3-Mediated N6-Methyladenosine Modification Is Involved in the Dysregulation of NRIP1 Expression in Down Syndrome. Front Cell Dev Biol 2021; 9:621374. [PMID: 33869171 PMCID: PMC8047211 DOI: 10.3389/fcell.2021.621374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 03/08/2021] [Indexed: 11/13/2022] Open
Abstract
Down syndrome (DS) is a common genetic condition in which a person is born with an extra copy of chromosome 21. Intellectual disability is the most common characteristic of DS. N6-methyladenosine (m6A) is a common RNA modification that is implicated in many biological processes. It is highly enriched within the brain and plays an essential role in human brain development. However, the mRNA m6A modification in the fetal brain of DS has not been explored. Here, we report m6A mRNA profiles and mRNA expression profiles of fetal brain cortex tissue from DSs and controls. We observed that the m6A modification in DS brain tissues was reduced genome-wide, which may be due to decreased the m6A methyltransferase like 3 (METTL3) protein expression. The nuclear receptor-interacting protein 1 (NRIP1/RIP140) is coded by a highly conserved chromosome 21 (Hsa21) gene. Overexpression of NRIP1 is associated with mitochondrial dysfunction in DS. The NRIP1 mRNA increased in fetal brain tissues of DS, whereas the m6A modification of the NRIP1 mRNA significantly decreased. METTL3 knockdown reduced the m6A modification of NRIP1 mRNA and increased its expression, and an increase in NRIP1 m6A modification and a decrease in its expression were observed in METTL3-overexpressed cells. The Luciferase reporter assay confirmed that METTL3 regulates NRIP1 expression in an m6A-dependent manner. The decay rate of NRIP1 mRNA was significantly reduced in METTL3-knockdown cells but increased in METTL3-overexpressed cells. We proposed that the m6A modification of NRIP1 mRNA in DS fetal brain tissue is reduced, reducing its transcript degradation rate, resulting in abnormally increased expression of NRIP1, at least partially, in the DS brain. It provides a new mechanism for the molecular pathology of DS and leads to a new insight that may become therapeutically relevant.
Collapse
Affiliation(s)
- Weili Shi
- Henan Provincial People's Hospital, Medical Genetics Institute of Henan Province, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, People's Hospital of Zhengzhou University, Zhengzhou, China.,National Health Commission Key Laboratory of Birth Defect Prevention, Henan Key Laboratory of Population Defects Prevention, Zhengzhou, China
| | - Fan Yang
- Zhengzhou Central Hospital, Department of Neurology, Zhengzhou, China
| | - Ranran Dai
- Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yafei Sun
- National Health Commission Key Laboratory of Birth Defect Prevention, Henan Key Laboratory of Population Defects Prevention, Zhengzhou, China
| | - Yan Chu
- Henan Provincial People's Hospital, Medical Genetics Institute of Henan Province, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Shixiu Liao
- Henan Provincial People's Hospital, Medical Genetics Institute of Henan Province, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, People's Hospital of Zhengzhou University, Zhengzhou, China.,National Health Commission Key Laboratory of Birth Defect Prevention, Henan Key Laboratory of Population Defects Prevention, Zhengzhou, China
| | - Bingtao Hao
- Henan Provincial People's Hospital, Medical Genetics Institute of Henan Province, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, People's Hospital of Zhengzhou University, Zhengzhou, China.,National Health Commission Key Laboratory of Birth Defect Prevention, Henan Key Laboratory of Population Defects Prevention, Zhengzhou, China.,Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Duchon A, Del Mar Muniz Moreno M, Martin Lorenzo S, Silva de Souza MP, Chevalier C, Nalesso V, Meziane H, Loureiro de Sousa P, Noblet V, Armspach JP, Brault V, Herault Y. Multi-influential genetic interactions alter behaviour and cognition through six main biological cascades in Down syndrome mouse models. Hum Mol Genet 2021; 30:771-788. [PMID: 33693642 PMCID: PMC8161522 DOI: 10.1093/hmg/ddab012] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Down syndrome (DS) is the most common genetic form of intellectual disability caused by the presence of an additional copy of human chromosome 21 (Hsa21). To provide novel insights into genotype–phenotype correlations, we used standardized behavioural tests, magnetic resonance imaging and hippocampal gene expression to screen several DS mouse models for the mouse chromosome 16 region homologous to Hsa21. First, we unravelled several genetic interactions between different regions of chromosome 16 and how they contribute significantly to altering the outcome of the phenotypes in brain cognition, function and structure. Then, in-depth analysis of misregulated expressed genes involved in synaptic dysfunction highlighted six biological cascades centred around DYRK1A, GSK3β, NPY, SNARE, RHOA and NPAS4. Finally, we provide a novel vision of the existing altered gene–gene crosstalk and molecular mechanisms targeting specific hubs in DS models that should become central to better understanding of DS and improving the development of therapies.
Collapse
Affiliation(s)
- Arnaud Duchon
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Maria Del Mar Muniz Moreno
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Sandra Martin Lorenzo
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Marcia Priscilla Silva de Souza
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Claire Chevalier
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Valérie Nalesso
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Hamid Meziane
- Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris (ICS), CELPHEDIA, PHENOMIN, 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | | | - Vincent Noblet
- Université de Strasbourg, CNRS UMR 7357, ICube, FMTS, 67000 Strasbourg, France
| | - Jean-Paul Armspach
- Université de Strasbourg, CNRS UMR 7357, ICube, FMTS, 67000 Strasbourg, France
| | - Veronique Brault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France.,Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris (ICS), CELPHEDIA, PHENOMIN, 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| |
Collapse
|
16
|
Lee HC, Hamzah H, Leong MPY, Md Yusof H, Habib O, Zainal Abidin S, Seth EA, Lim SM, Vidyadaran S, Mohd Moklas MA, Abdullah MA, Nordin N, Hassan Z, Cheah PS, Ling KH. Transient prenatal ruxolitinib treatment suppresses astrogenesis during development and improves learning and memory in adult mice. Sci Rep 2021; 11:3847. [PMID: 33589712 PMCID: PMC7884429 DOI: 10.1038/s41598-021-83222-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 01/20/2021] [Indexed: 01/08/2023] Open
Abstract
Ruxolitinib is the first janus kinase 1 (JAK1) and JAK2 inhibitor that was approved by the United States Food and Drug Administration (FDA) agency for the treatment of myeloproliferative neoplasms. The drug targets the JAK/STAT signalling pathway, which is critical in regulating the gliogenesis process during nervous system development. In the study, we assessed the effect of non-maternal toxic dosages of ruxolitinib (0-30 mg/kg/day between E7.5-E20.5) on the brain of the developing mouse embryos. While the pregnant mice did not show any apparent adverse effects, the Gfap protein marker for glial cells and S100β mRNA marker for astrocytes were reduced in the postnatal day (P) 1.5 pups' brains. Gfap expression and Gfap+ cells were also suppressed in the differentiating neurospheres culture treated with ruxolitinib. Compared to the control group, adult mice treated with ruxolitinib prenatally showed no changes in motor coordination, locomotor function, and recognition memory. However, increased explorative behaviour within an open field and improved spatial learning and long-term memory retention were observed in the treated group. We demonstrated transplacental effects of ruxolitinib on astrogenesis, suggesting the potential use of ruxolitinib to revert pathological conditions caused by gliogenic-shift in early brain development such as Down and Noonan syndromes.
Collapse
Affiliation(s)
- Han-Chung Lee
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Hamizun Hamzah
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Melody Pui-Yee Leong
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Hadri Md Yusof
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Department of Biotechnology, Faculty of Science, Technology, Engineering and Mathematics, International University of Malaya-Wales, 50480, Kuala Lumpur, Malaysia
| | - Omar Habib
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Shahidee Zainal Abidin
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030, Kuala Nerus, Terengganu, Malaysia
| | - Eryse Amira Seth
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Siong-Meng Lim
- Collaborative Drug Discovery Research, Faculty of Pharmacy, Universiti Teknologi MARA, Cawangan Selangor, Kampus Puncak Alam, 42300, Bandar Puncak Alam, Selangor, Malaysia
| | - Sharmili Vidyadaran
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Mohamad Aris Mohd Moklas
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Maizaton Atmadini Abdullah
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Institute of Biosciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Norshariza Nordin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Pike-See Cheah
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
| | - King-Hwa Ling
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
| |
Collapse
|
17
|
Pellegrini M, Zoghi M, Jaberzadeh S. Genetic Polymorphisms Do Not Predict Interindividual Variability to Cathodal Transcranial Direct Current Stimulation of the Primary Motor Cortex. Brain Connect 2020; 11:56-72. [PMID: 33198509 DOI: 10.1089/brain.2020.0762] [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] [Indexed: 11/13/2022] Open
Abstract
Introduction: High variability between individuals (i.e., interindividual variability) in response to transcranial direct current stimulation (tDCS) has become a commonly reported issue in the tDCS literature in recent years. Inherent genetic differences between individuals have been proposed as a contributing factor to observed response variability. This study investigated whether tDCS interindividual variability was genetically mediated. Methods: A large sample size of 61 healthy males received cathodal tDCS (c-tDCS) and sham-tDCS of the primary motor cortex at 1 mA and 10 min via 6 × 4 cm active and 7 × 5 cm return electrodes. Corticospinal excitability (CSE) was assessed via 25 single-pulse transcranial magnetic stimulation motor-evoked potentials (MEPs). Intracortical inhibition was assessed via twenty-five 3 msec interstimulus interval (ISI) paired-pulse MEPs, known as short-interval intracortical inhibition (SICI). Intracortical facilitation (ICF) was assessed via twenty-five 10 msec ISI paired-pulse MEPs. Gene variants encoding for excitatory and inhibitory neuroreceptors were determined via saliva samples. Predetermined thresholds and statistical cluster analyses were used to subgroup individuals. Results: Two distinct subgroups were identified, "responders" reducing CSE following c-tDCS and "nonresponders" showing no reduction or even increase in CSE. Differences in CSE between responders and nonresponders following c-tDCS were not explained by changes in SICI or ICF. Conclusions: No significant relationships were reported between gene variants and interindividual variability to c-tDCS, suggesting that the chosen gene variants did not influence the activity of the neuroreceptors involved in eliciting changes in CSE in responders following c-tDCS. In this largest c-tDCS study of its kind, novel insights were reported into the contribution genetic factors may play in observed interindividual variability to c-tDCS. Impact statement This study adds insight into the issue of interindividual variability to c-tDCS. It highlights not all individuals respond to c-tDCS similarly when exposed to the same stimulus parameters. This disparity in response to c-tDCS between individuals does not appear to be genetically mediated. For c-tDCS to progress to large-scale clinical application, reliability, predictability and reproducibility are essential. Systematically investigating factors contributing to interindividual variability take steps towards this progress the c-tDCS field towards the potential development of screening tools to determine clinical suitability to c-tDCS to ensure its application in those who may benefit the most.
Collapse
Affiliation(s)
- Michael Pellegrini
- Non-Invasive Brain Stimulation and Neuroplasticity Laboratory, Department of Physiotherapy, School of Primary and Allied Health Care, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Australia
| | - Maryam Zoghi
- Department of Rehabilitation, Nutrition and Sport, School of Allied Health, Discipline of Physiotherapy, La Trobe University, Melbourne, Australia
| | - Shapour Jaberzadeh
- Non-Invasive Brain Stimulation and Neuroplasticity Laboratory, Department of Physiotherapy, School of Primary and Allied Health Care, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Australia
| |
Collapse
|
18
|
Kipper FC, Angolano C, Vissapragada R, Contreras MA, Moore J, Bhasin M, Ferran C, Thomas AJ. Embryonic periventricular endothelial cells demonstrate a unique pro-neurodevelopment and anti-inflammatory gene signature. Sci Rep 2020; 10:20393. [PMID: 33230288 PMCID: PMC7683543 DOI: 10.1038/s41598-020-77297-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/02/2020] [Indexed: 01/08/2023] Open
Abstract
Brain embryonic periventricular endothelial cells (PVEC) crosstalk with neural progenitor cells (NPC) promoting mutual proliferation, formation of tubular-like structures in the former and maintenance of stemness in the latter. To better characterize this interaction, we conducted a comparative transcriptome analysis of mouse PVEC vs. adult brain endothelial cells (ABEC) in mono-culture or NPC co-culture. We identified > 6000 differentially expressed genes (DEG), regardless of culture condition. PVEC exhibited a 30-fold greater response to NPC than ABEC (411 vs. 13 DEG). Gene Ontology (GO) analysis of DEG that were higher or lower in PVEC vs. ABEC identified "Nervous system development" and "Response to Stress" as the top significantly different biological process, respectively. Enrichment in canonical pathways included HIF1A, FGF/stemness, WNT signaling, interferon signaling and complement. Solute carriers (SLC) and ABC transporters represented an important subset of DEG, underscoring PVEC's implication in blood-brain barrier formation and maintenance of nutrient-rich/non-toxic environment. Our work characterizes the gene signature of PVEC and their important partnership with NPC, underpinning their unique role in maintaining a healthy neurovascular niche, and in supporting brain development. This information may pave the way for additional studies to explore their therapeutic potential in neuro-degenerative diseases, such as Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Franciele Cristina Kipper
- Division of Neurosurgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02215, USA
| | - Cleide Angolano
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02215, USA
| | - Ravi Vissapragada
- Division of Neurosurgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Discipline of Surgery, College of Medicine and Public Health, Flinders University, Adelaide, 5042, SA, Australia
| | - Mauricio A Contreras
- Division of Vascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Justin Moore
- Division of Neurosurgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02215, USA
| | - Manoj Bhasin
- BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Christiane Ferran
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Division of Vascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02215, USA
| | - Ajith J Thomas
- Division of Neurosurgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA.
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA.
- Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
19
|
Phenotype microarrays reveal metabolic dysregulations of neurospheres derived from embryonic Ts1Cje mouse model of Down syndrome. PLoS One 2020; 15:e0236826. [PMID: 32730314 PMCID: PMC7392322 DOI: 10.1371/journal.pone.0236826] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/14/2020] [Indexed: 11/20/2022] Open
Abstract
Down syndrome (DS), is the most common cause of intellectual disability, and is characterized by defective neurogenesis during perinatal development. To identify metabolic aberrations in early neurogenesis, we profiled neurospheres derived from the embryonic brain of Ts1Cje, a mouse model of Down syndrome. High-throughput phenotypic microarray revealed a significant decrease in utilisation of 17 out of 367 substrates and significantly higher utilisation of 6 substrates in the Ts1Cje neurospheres compared to controls. Specifically, Ts1Cje neurospheres were less efficient in the utilisation of glucose-6-phosphate suggesting a dysregulation in the energy-producing pathway. T Cje neurospheres were significantly smaller in diameter than the controls. Subsequent preliminary study on supplementation with 6-phosphogluconic acid, an intermediate of glucose-6-phosphate metabolism, was able to rescue the Ts1Cje neurosphere size. This study confirmed the perturbed pentose phosphate pathway, contributing to defects observed in Ts1Cje neurospheres. We show for the first time that this comprehensive energetic assay platform facilitates the metabolic characterisation of Ts1Cje cells and confirmed their distinguishable metabolic profiles compared to the controls.
Collapse
|
20
|
Creisher PS, Chandwani MN, Kamte YS, Covvey JR, Ganesan P, O’Donnell LA. Type II interferon signaling in the brain during a viral infection with age-dependent pathogenesis. Dev Neurobiol 2020; 80:213-228. [PMID: 32866337 PMCID: PMC8513332 DOI: 10.1002/dneu.22778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 06/13/2020] [Accepted: 08/05/2020] [Indexed: 01/10/2023]
Abstract
Viral infections of the central nervous system (CNS) often cause disease in an age-dependent manner, with greater neuropathology during the fetal and neonatal periods. Transgenic CD46+ mice model these age-dependent outcomes through a measles virus infection of CNS neurons. Adult CD46+ mice control viral spread and survive the infection in an interferon gamma (IFNγ)-dependent manner, whereas neonatal CD46+ mice succumb despite similar IFNγ expression in the brain. Thus, we hypothesized that IFNγ signaling in the adult brain may be more robust, potentially due to greater basal expression of IFNγ signaling proteins. To test this hypothesis, we evaluated the expression of canonical IFNγ signaling proteins in the neonatal and adult brain, including the IFNγ receptor, Janus kinase (JAK) 1/2, and signal transducer and activator of transcription-1 (STAT1) in the absence of infection. We also analyzed the expression and activation of STAT1 and IFNγ-stimulated genes during MV infection. We found that neonatal brains have equivalent or greater JAK/STAT1 expression in the hippocampus and the cerebellum than adults. IFNγ receptor expression varied by cell type in the brain but was widely expressed on neuronal and glial cells. During MV infection, increased STAT1 expression and activation correlated with viral load in the hippocampus regardless of age, but not in the cerebellum where viral load was consistently undetectable in adults. These results suggest the neonatal brain is capable of initiating IFNγ signaling during a viral infection, but that downstream STAT1 activation is insufficient to limit viral spread.
Collapse
Affiliation(s)
- Patrick S. Creisher
- Duquesne University, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Pittsburgh, PA 15282
| | - Manisha N. Chandwani
- Duquesne University, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Pittsburgh, PA 15282
| | - Yashika S. Kamte
- Duquesne University, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Pittsburgh, PA 15282
| | - Jordan R. Covvey
- Duquesne University, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Pittsburgh, PA 15282
| | - Priya Ganesan
- Duquesne University, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Pittsburgh, PA 15282
| | - Lauren A. O’Donnell
- Duquesne University, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Pittsburgh, PA 15282
| |
Collapse
|
21
|
Zhou G, Xu Y, He B, Ma R, Wang Y, Chang Y, Xie Y, Wu L, Huang J, Xiao Z. Ionizing radiation modulates vascular endothelial growth factor expression through STAT3 signaling pathway in rat neonatal primary astrocyte cultures. Brain Behav 2020; 10:e01529. [PMID: 32106359 PMCID: PMC7177558 DOI: 10.1002/brb3.1529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/30/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Radiation-induced brain injury (RBI) usually occurs six months to three years after irradiation, often shows cognitive dysfunction, epilepsy, and other neurological dysfunction. In severe cases, it can cause a wide range of cerebral edema, even herniation. It seriously threatens the survival of patients and their quality of life, and it becomes a key factor in limiting the radiation dose and lowering the therapeutic efficacy in recent years. Therefore, studying the pathogenesis of RBI and exploring new therapeutic targets are of great significance. METHODS In our study, we observed the activation and secretory function in astrocytes as well as the intracellular signal transducer and activator of transcription 3 (STAT3) signal transduction pathway activation status after exposing different doses of X-ray irradiation by using MTT, Immunocytologic analysis, and Western blot analysis. Further, we used the same way to explore the role of vascular endothelial growth factor (VEGF) in signal transduction pathways playing in the activation of astrocytes after irradiating through the use of specificInhivascular endothelial growth factorbitors of STAT3. RESULTS Ast can be directly activated, reactive hyperplasia and hypertrophy, the expression of the activation marker glial fibrillary acidic protein is increased, and the expression of vascular endothelial growth factor (VEGF) in the cells is increased, which may lead to RBI. After the addition of STAT3 pathway inhibitor, most of the Ast radiation activation was suppressed, and the expression of high-level expression of VEGF decreased after irradiation. CONCLUSION Our findings demonstrated that X-ray irradiation directly induced the activation of astrocytes in a persistent manner and X-ray irradiation activated STAT3 signaling pathway. As the same time, we found that X-ray irradiation induced the activation of astrocytes and secretion cytokine. The STAT3 signaling pathway may participate in the pathogenesis of radiation-induced brain injury.
Collapse
Affiliation(s)
- Guijuan Zhou
- The First Afliated Hospital of University of South China, University of South China, Hengyang, China
| | - Yan Xu
- The First Afliated Hospital of University of South China, University of South China, Hengyang, China
| | - Bing He
- The First Afliated Hospital of University of South China, University of South China, Hengyang, China
| | - Rundong Ma
- The First Afliated Hospital of University of South China, University of South China, Hengyang, China
| | - Yilin Wang
- The First Afliated Hospital of University of South China, University of South China, Hengyang, China
| | - Yunqian Chang
- The First Afliated Hospital of University of South China, University of South China, Hengyang, China
| | - Yangzhi Xie
- The First Afliated Hospital of University of South China, University of South China, Hengyang, China.,Leiyang People's Hospital, Leiyang, China
| | - Lin Wu
- The First Afliated Hospital of University of South China, University of South China, Hengyang, China
| | - Jianghua Huang
- The First Afliated Hospital of University of South China, University of South China, Hengyang, China
| | - Zijian Xiao
- The First Afliated Hospital of University of South China, University of South China, Hengyang, China
| |
Collapse
|
22
|
Zarei-Kheirabadi M, Vaccaro AR, Rahimi-Movaghar V, Kiani S, Baharvand H. An Overview of Extrinsic and Intrinsic Mechanisms Involved in Astrocyte Development in the Central Nervous System. Stem Cells Dev 2020; 29:266-280. [PMID: 31847709 DOI: 10.1089/scd.2019.0189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Over the past few decades, our knowledge about the function of the central nervous system (CNS) and astrocytes has improved, and research has confirmed the key roles that astrocytes play in the physiology and pathology of the CNS. Here, we reviewed the intrinsic and extrinsic mechanisms that regulate the development of astrocytes, which are generated from radial glial cells. These regulatory systems modulate various signaling pathways and transcription factors. In this review, four stages of astrocyte development-specification (patterning and switch), migration, proliferation, and maturation, are discussed. In astrocyte patterning, VA1-VA3 domains create the astrocyte subtypes by differential expression of Slit1 and Reelin in the spinal cord. In the brain, patterning creates several astrocyte subtypes by different organizing centers. At the switch step, the janus kinase-signal transducer and activator of transcription pathway governs the transition of neurogenesis to gliogenesis. Bone marrow protein and Notch pathways are also important players of the progliogenic switch. Intrinsic regulation is mediated by DNA methylation transferases, and polycomb group complexes can intrinsically affect the development of astrocytes. In the next stage, these cells proliferate and migrate to their final location. Astrocyte maturation is accomplished through the development of cellular processes, molecular markers, and functions.
Collapse
Affiliation(s)
- Masoumeh Zarei-Kheirabadi
- Department of Brain, Cognitive Sciences and Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Alexander R Vaccaro
- Department of Orthopedics, Rothman Orthopedic Institute, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Vafa Rahimi-Movaghar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Kiani
- Department of Brain, Cognitive Sciences and Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
23
|
Li H, Liu Z, Wu Y, Chen Y, Wang J, Wang Z, Huang D, Wang M, Yu M, Fei J, Huang F. The deficiency of NRSF/REST enhances the pro-inflammatory function of astrocytes in a model of Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165590. [DOI: 10.1016/j.bbadis.2019.165590] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/11/2019] [Accepted: 09/30/2019] [Indexed: 12/15/2022]
|
24
|
Aberrant Oligodendrogenesis in Down Syndrome: Shift in Gliogenesis? Cells 2019; 8:cells8121591. [PMID: 31817891 PMCID: PMC6953000 DOI: 10.3390/cells8121591] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/25/2022] Open
Abstract
Down syndrome (DS), or trisomy 21, is the most prevalent chromosomal anomaly accounting for cognitive impairment and intellectual disability (ID). Neuropathological changes of DS brains are characterized by a reduction in the number of neurons and oligodendrocytes, accompanied by hypomyelination and astrogliosis. Recent studies mainly focused on neuronal development in DS, but underestimated the role of glial cells as pathogenic players. Aberrant or impaired differentiation within the oligodendroglial lineage and altered white matter functionality are thought to contribute to central nervous system (CNS) malformations. Given that white matter, comprised of oligodendrocytes and their myelin sheaths, is vital for higher brain function, gathering knowledge about pathways and modulators challenging oligodendrogenesis and cell lineages within DS is essential. This review article discusses to what degree DS-related effects on oligodendroglial cells have been described and presents collected evidence regarding induced cell-fate switches, thereby resulting in an enhanced generation of astrocytes. Moreover, alterations in white matter formation observed in mouse and human post-mortem brains are described. Finally, the rationale for a better understanding of pathways and modulators responsible for the glial cell imbalance as a possible source for future therapeutic interventions is given based on current experience on pro-oligodendroglial treatment approaches developed for demyelinating diseases, such as multiple sclerosis.
Collapse
|
25
|
McCarthy GD, Drewell RA, Dresch JM. Analyzing the stability of gene expression using a simple reaction-diffusion model in an early Drosophila embryo. Math Biosci 2019; 316:108239. [PMID: 31454629 DOI: 10.1016/j.mbs.2019.108239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 08/20/2019] [Accepted: 08/22/2019] [Indexed: 11/28/2022]
Abstract
In all complex organisms, the precise levels and timing of gene expression controls vital biological processes. In higher eukaryotes, including the fruit fly Drosophila melanogaster, the complex molecular control of transcription (the synthesis of RNA from DNA) and translation (the synthesis of proteins from RNA) events driving this gene expression are not fully understood. In particular, for Drosophila melanogaster, there is a plethora of experimental data, including quantitative measurements of both RNA and protein concentrations, but the precise mechanisms that control the dynamics of gene expression during early development and the processes which lead to steady-state levels of certain proteins remain elusive. This study analyzes a current mathematical modeling approach in an attempt to better understand the long-term behavior of gene regulation. The model is a modified reaction-diffusion equation which has been previously employed in predicting gene expression levels and studying the relative contributions of transcription and translation events to protein abundance [10,11,24]. Here, we use Matrix Algebra and Analysis techniques to study the stability of the gene expression system and analyze equilibria, using very general assumptions regarding the parameter values incorporated into the model. We prove that, given realistic biological parameter values, the system will result in a unique, stable equilibrium solution. Additionally, we give an example of this long-term behavior using the model alongside actual experimental data obtained from Drosophila embryos.
Collapse
Affiliation(s)
- Gregory D McCarthy
- School of Natural Science, Hampshire College, Amherst, MA 01002, United States.
| | - Robert A Drewell
- Biology Department, Clark University, Worcester, MA 01610, United States.
| | - Jacqueline M Dresch
- Department of Mathematics and Computer Science, Clark University, Worcester, MA 01610, United States.
| |
Collapse
|
26
|
Lee HC, Md Yusof HH, Leong MPY, Zainal Abidin S, Seth EA, Hewitt CA, Vidyadaran S, Nordin N, Scott HS, Cheah PS, Ling KH. Gene and protein expression profiles of JAK-STAT signalling pathway in the developing brain of the Ts1Cje down syndrome mouse model. Int J Neurosci 2019; 129:871-881. [DOI: 10.1080/00207454.2019.1580280] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Han-Chung Lee
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Hadri Hadi Md Yusof
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Melody Pui-Yee Leong
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Shahidee Zainal Abidin
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Eryse Amira Seth
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Chelsee A. Hewitt
- Department of Pathology, The Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Sharmili Vidyadaran
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Pathology, Immunology Unit, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Norshariza Nordin
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Hamish S. Scott
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, An alliance between SA Pathology and the University of South Australia, SA Pathology, Adelaide, Australia
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, Australia
- School of Medicine, School of Biological Sciences, University of Adelaide, Adelaide, South Australia
- Centre for Cancer Biology, SA Pathology, Australian Cancer Research Foundation Genomics Facility, Adelaide, Australia
| | - Pike-See Cheah
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - King-Hwa Ling
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| |
Collapse
|
27
|
Nabavi SM, Ahmed T, Nawaz M, Devi KP, Balan DJ, Pittalà V, Argüelles-Castilla S, Testai L, Khan H, Sureda A, de Oliveira MR, Vacca RA, Xu S, Yousefi B, Curti V, Daglia M, Sobarzo-Sánchez E, Filosa R, Nabavi SF, Majidinia M, Dehpour AR, Shirooie S. Targeting STATs in neuroinflammation: The road less traveled! Pharmacol Res 2018; 141:73-84. [PMID: 30550953 DOI: 10.1016/j.phrs.2018.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/01/2018] [Accepted: 12/10/2018] [Indexed: 12/16/2022]
Abstract
JAK/STAT transduction pathway is a highly conserved pathway implicated in regulating cellular proliferation, differentiation, survival and apoptosis. Dysregulation of this pathway is involved in the onset of autoimmune, haematological, oncological, metabolic and neurological diseases. Over the last few years, the research of anti-neuroinflammatory agents has gained considerable attention. The ability to diminish the STAT-induced transcription of inflammatory genes is documented for both natural compounds (such as polyphenols) and chemical drugs. Among polyphenols, quercetin and curcumin directly inhibit STAT, while Berberis vulgaris L. and Sophora alopecuroides L extracts act indirectly. Also, the Food and Drug Administration has approved several JAK/STAT inhibitors (direct or indirect) for treating inflammatory diseases, indicating STAT can be considered as a therapeutic target for neuroinflammatory pathologies. Considering the encouraging data obtained so far, clinical trials are warranted to demonstrate the effectiveness and potential use in the clinical practice of STAT inhibitors to treat inflammation-associated neurodegenerative pathologies.
Collapse
Affiliation(s)
- Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Touqeer Ahmed
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad, 44000, Pakistan
| | - Maheen Nawaz
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad, 44000, Pakistan
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University (Science Campus), Karaikudi 630 003, Tamil Nadu, India
| | - Devasahayam Jaya Balan
- Department of Biotechnology, Alagappa University (Science Campus), Karaikudi 630 003, Tamil Nadu, India
| | - Valeria Pittalà
- Department of Drug Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | | | - Lara Testai
- Department of Pharmacy, University of Pisa, Pisa, via Bonanno 6 - 56126, Pisa, Italy
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress and CIBEROBN (Physiopathology of Obesity and Nutrition), University of Balearic Islands, E-07122 Palma de Mallorca, Spain.
| | - Marcos Roberto de Oliveira
- Department of Chemistry/ICET, Federal University of Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, 78060-900, Brazil
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, I-70126, Bari, Italy
| | - Suowen Xu
- University of Rochester, Aab Cardiovascular Research Institute, Rochester, NY, 14623, USA
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Valeria Curti
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Maria Daglia
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Eduardo Sobarzo-Sánchez
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782, Spain; Instituto de Investigación e Innovación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago, Chile
| | - Rosanna Filosa
- Consorzio Sannio Tech, Appia Str, Apollosa, BN 82030, Italy
| | - Seyed Fazel Nabavi
- Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran; Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Shirooie
- Department of Pharmacology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
28
|
Lucas ED, Finlon JM, Burchill MA, McCarthy MK, Morrison TE, Colpitts TM, Tamburini BAJ. Type 1 IFN and PD-L1 Coordinate Lymphatic Endothelial Cell Expansion and Contraction during an Inflammatory Immune Response. THE JOURNAL OF IMMUNOLOGY 2018; 201:1735-1747. [PMID: 30045970 DOI: 10.4049/jimmunol.1800271] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/09/2018] [Indexed: 12/16/2022]
Abstract
Lymph node (LN) expansion during an immune response is a complex process that involves the relaxation of the fibroblastic network, germinal center formation, and lymphatic vessel growth. These processes require the stromal cell network of the LN to act deliberately to accommodate the influx of immune cells to the LN. The molecular drivers of these processes are not well understood. Therefore, we asked whether the immediate cytokines type 1 IFN produced during viral infection influence the lymphatic network of the LN in mice. We found that following an IFN-inducing stimulus such as viral infection or polyI:C, programmed cell death ligand 1 (PD-L1) expression is dynamically upregulated on lymphatic endothelial cells (LECs). We found that reception of type 1 IFN by LECs is important for the upregulation of PD-L1 of mouse and human LECs and the inhibition of LEC expansion in the LN. Expression of PD-L1 by LECs is also important for the regulation of LN expansion and contraction after an IFN-inducing stimulus. We demonstrate a direct role for both type 1 IFN and PD-L1 in inhibiting LEC division and in promoting LEC survival. Together, these data reveal a novel mechanism for the coordination of type 1 IFN and PD-L1 in manipulating LEC expansion and survival during an inflammatory immune response.
Collapse
Affiliation(s)
- Erin D Lucas
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Jeffrey M Finlon
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Matthew A Burchill
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Mary K McCarthy
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Thomas E Morrison
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Tonya M Colpitts
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118; and.,Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| | - Beth A Jirón Tamburini
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; .,Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
29
|
Stagni F, Giacomini A, Emili M, Guidi S, Bartesaghi R. Neurogenesis impairment: An early developmental defect in Down syndrome. Free Radic Biol Med 2018; 114:15-32. [PMID: 28756311 DOI: 10.1016/j.freeradbiomed.2017.07.026] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 07/24/2017] [Accepted: 07/25/2017] [Indexed: 02/06/2023]
Abstract
Down syndrome (DS) is characterized by brain hypotrophy and intellectual disability starting from early life stages. Accumulating evidence shows that the phenotypic features of the DS brain can be traced back to the fetal period since the DS brain exhibits proliferation potency reduction starting from the critical time window of fetal neurogenesis. This defect is worsened by the fact that neural progenitor cells exhibit reduced acquisition of a neuronal phenotype and an increase in the acquisition of an astrocytic phenotype. Consequently, the DS brain has fewer neurons in comparison with the typical brain. Although apoptotic cell death may be increased in DS, this does not seem to be the major cause of brain hypocellularity. Evidence obtained in brains of individuals with DS, DS-derived induced pluripotent stem cells (iPSCs), and DS mouse models has provided some insight into the mechanisms underlying the developmental defects due to the trisomic condition. Although many triplicated genes may be involved, in the light of the studies reviewed here, DYRK1A, APP, RCAN1 and OLIG1/2 appear to be particularly important determinants of many neurodevelopmental alterations that characterize DS because their triplication affects both the proliferation and fate of neural precursor cells as well as apoptotic cell death. Based on the evidence reviewed here, pathways downstream to these genes may represent strategic targets, for the design of possible interventions.
Collapse
Affiliation(s)
- Fiorenza Stagni
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Andrea Giacomini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
30
|
Oswald F, Klöble P, Ruland A, Rosenkranz D, Hinz B, Butter F, Ramljak S, Zechner U, Herlyn H. The FOXP2-Driven Network in Developmental Disorders and Neurodegeneration. Front Cell Neurosci 2017; 11:212. [PMID: 28798667 PMCID: PMC5526973 DOI: 10.3389/fncel.2017.00212] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/04/2017] [Indexed: 12/24/2022] Open
Abstract
The transcription repressor FOXP2 is a crucial player in nervous system evolution and development of humans and songbirds. In order to provide an additional insight into its functional role we compared target gene expression levels between human neuroblastoma cells (SH-SY5Y) stably overexpressing FOXP2 cDNA of either humans or the common chimpanzee, Rhesus monkey, and marmoset, respectively. RNA-seq led to identification of 27 genes with differential regulation under the control of human FOXP2, which were previously reported to have FOXP2-driven and/or songbird song-related expression regulation. RT-qPCR and Western blotting indicated differential regulation of additional 13 new target genes in response to overexpression of human FOXP2. These genes may be directly regulated by FOXP2 considering numerous matches of established FOXP2-binding motifs as well as publicly available FOXP2-ChIP-seq reads within their putative promoters. Ontology analysis of the new and reproduced targets, along with their interactors in a network, revealed an enrichment of terms relating to cellular signaling and communication, metabolism and catabolism, cellular migration and differentiation, and expression regulation. Notably, terms including the words "neuron" or "axonogenesis" were also enriched. Complementary literature screening uncovered many connections to human developmental (autism spectrum disease, schizophrenia, Down syndrome, agenesis of corpus callosum, trismus-pseudocamptodactyly, ankyloglossia, facial dysmorphology) and neurodegenerative diseases and disorders (Alzheimer's, Parkinson's, and Huntington's diseases, Lewy body dementia, amyotrophic lateral sclerosis). Links to deafness and dyslexia were detected, too. Such relations existed for single proteins (e.g., DCDC2, NURR1, PHOX2B, MYH8, and MYH13) and groups of proteins which conjointly function in mRNA processing, ribosomal recruitment, cell-cell adhesion (e.g., CDH4), cytoskeleton organization, neuro-inflammation, and processing of amyloid precursor protein. Conspicuously, many links pointed to an involvement of the FOXP2-driven network in JAK/STAT signaling and the regulation of the ezrin-radixin-moesin complex. Altogether, the applied phylogenetic perspective substantiated FOXP2's importance for nervous system development, maintenance, and functioning. However, the study also disclosed new regulatory pathways that might prove to be useful for understanding the molecular background of the aforementioned developmental disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Franz Oswald
- Center for Internal Medicine, Department of Internal Medicine I, University Medical Center UlmUlm, Germany
| | - Patricia Klöble
- Center for Internal Medicine, Department of Internal Medicine I, University Medical Center UlmUlm, Germany
| | - André Ruland
- Center for Internal Medicine, Department of Internal Medicine I, University Medical Center UlmUlm, Germany
| | - David Rosenkranz
- Institut für Organismische und Molekulare Evolutionsbiologie, Johannes Gutenberg-University MainzMainz, Germany
| | - Bastian Hinz
- Institut für Organismische und Molekulare Evolutionsbiologie, Johannes Gutenberg-University MainzMainz, Germany
- Institute of Human Genetics, University Medical Center MainzMainz, Germany
| | - Falk Butter
- Institute of Molecular BiologyMainz, Germany
| | | | - Ulrich Zechner
- Institute of Human Genetics, University Medical Center MainzMainz, Germany
- Dr. Senckenbergisches Zentrum für HumangenetikFrankfurt, Germany
| | - Holger Herlyn
- Institut für Organismische und Molekulare Evolutionsbiologie, Johannes Gutenberg-University MainzMainz, Germany
| |
Collapse
|
31
|
|
32
|
Sullivan KD, Lewis HC, Hill AA, Pandey A, Jackson LP, Cabral JM, Smith KP, Liggett LA, Gomez EB, Galbraith MD, DeGregori J, Espinosa JM. Trisomy 21 consistently activates the interferon response. eLife 2016; 5:e16220. [PMID: 27472900 PMCID: PMC5012864 DOI: 10.7554/elife.16220] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 07/28/2016] [Indexed: 12/12/2022] Open
Abstract
Although it is clear that trisomy 21 causes Down syndrome, the molecular events acting downstream of the trisomy remain ill defined. Using complementary genomics analyses, we identified the interferon pathway as the major signaling cascade consistently activated by trisomy 21 in human cells. Transcriptome analysis revealed that trisomy 21 activates the interferon transcriptional response in fibroblast and lymphoblastoid cell lines, as well as circulating monocytes and T cells. Trisomy 21 cells show increased induction of interferon-stimulated genes and decreased expression of ribosomal proteins and translation factors. An shRNA screen determined that the interferon-activated kinases JAK1 and TYK2 suppress proliferation of trisomy 21 fibroblasts, and this defect is rescued by pharmacological JAK inhibition. Therefore, we propose that interferon activation, likely via increased gene dosage of the four interferon receptors encoded on chromosome 21, contributes to many of the clinical impacts of trisomy 21, and that interferon antagonists could have therapeutic benefits.
Collapse
Affiliation(s)
- Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - Hannah C Lewis
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, United States
| | - Amanda A Hill
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, United States
| | - Ahwan Pandey
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - Leisa P Jackson
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - Joseph M Cabral
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
| | - L Alexander Liggett
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, United States
| | - Eliana B Gomez
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - James DeGregori
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, United States
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, United States
- Integrated Department of Immunology, University of Colorado School of Medicine, Aurora, United States
- Section of Hematology, University of Colorado School of Medicine, Aurora, United States
- Department of Medicine, University of Colorado School of Medicine, Aurora, United States
| | - Joaquín M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| |
Collapse
|