1
|
Zhang Y, Cheng Y, Tang H, Yue Q, Cai X, Lu Z, Hao Y, Dai A, Hou T, Liu H, Kong N, Ji X, Lu C, Xu S, Huang K, Zeng X, Wen Y, Ma W, Guan J, Lin Y, Zheng W, Pan H, Wu J, Wu R, Wei N. APOE ε4-associated downregulation of the IL-7/IL-7R pathway in effector memory T cells: Implications for Alzheimer's disease. Alzheimers Dement 2024; 20:6441-6455. [PMID: 39129310 PMCID: PMC11497660 DOI: 10.1002/alz.14173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION The apolipoprotein E (APOE) ε4 allele exerts a significant influence on peripheral inflammation and neuroinflammation, yet the underlying mechanisms remain elusive. METHODS The present study enrolled 54 patients diagnosed with late-onset Alzheimer's disease (AD; including 28 APOE ε4 carriers and 26 non-carriers). Plasma inflammatory cytokine concentration was assessed, alongside bulk RNA sequencing (RNA-seq) and single-cell RNA sequencing (scRNA-seq) analysis of peripheral blood mononuclear cells (PBMCs). RESULTS Plasma tumor necrosis factor α, interferon γ, and interleukin (IL)-33 levels increased in the APOE ε4 carriers but IL-7 expression notably decreased. A negative correlation was observed between plasma IL-7 level and the hippocampal atrophy degree. Additionally, the expression of IL-7R and CD28 also decreased in PBMCs of APOE ε4 carriers. ScRNA-seq data results indicated that the changes were mainly related to the CD4+ Tem (effector memory) and CD8+ Tem T cells. DISCUSSION These findings shed light on the role of the downregulated IL-7/IL-7R pathway associated with the APOE ε4 allele in modulating neuroinflammation and hippocampal atrophy. HIGHLIGHTS The apolipoprotein E (APOE) ε4 allele decreases plasma interleukin (IL)-7 and aggravates hippocampal atrophy in Alzheimer's disease. Plasma IL-7 level is negatively associated with the degree of hippocampal atrophy. The expression of IL-7R signaling decreased in peripheral blood mononuclear cells of APOE ε4 carriers Dysregulation of the IL-7/IL-7R signal pathways enriches T cells.
Collapse
Affiliation(s)
- Ying‐Jie Zhang
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
- Department of RehabilitationThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Yan Cheng
- Department of RadiologyThe Second Affiliated Hospital of Shantou University Medical CollegeShantouChina
- Department of RadiologyThe Second Hospital of Shandong UniversityJinanChina
| | - Hai‐Liang Tang
- Department of NeurosurgeryFudan University Huashan HospitalShanghai Medical College Fudan UniversityShanghaiChina
| | - Qi Yue
- Department of NeurosurgeryFudan University Huashan HospitalShanghai Medical College Fudan UniversityShanghaiChina
| | - Xin‐Yi Cai
- Department of PathologyProvincial Key Laboratory of Infectious Diseases and Molecular ImmunopathologyShantou University Medical CollegeShantouChina
| | - Zhi‐Jie Lu
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Yi‐Xuan Hao
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - An‐Xiang Dai
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Ting Hou
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Hao‐Xin Liu
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Nan Kong
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Xiao‐Yu Ji
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Chang‐Hao Lu
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Sheng‐Liang Xu
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Kai Huang
- Department of RadiologyThe Second Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Xin Zeng
- Department of GeriatricsThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Ya‐Qi Wen
- Department of RadiologyThe Second Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Wan‐Yin Ma
- Department of RadiologyThe Second Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Ji‐Tian Guan
- Department of RadiologyThe Second Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Yan Lin
- Department of RadiologyThe Second Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Wen‐Bin Zheng
- Department of RadiologyThe Second Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Hui Pan
- Department of Family MedicineShantou Longhu People's HospitalShantouChina
| | - Jie Wu
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Ren‐Hua Wu
- Department of RadiologyThe Second Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Nai‐Li Wei
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| |
Collapse
|
2
|
IL-7: Comprehensive review. Cytokine 2022; 160:156049. [DOI: 10.1016/j.cyto.2022.156049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/08/2022] [Accepted: 09/16/2022] [Indexed: 01/08/2023]
|
3
|
Impact of Age on Plasma Inflammatory Biomarkers in the 6 Months Following Mild Traumatic Brain Injury. J Head Trauma Rehabil 2021; 35:324-331. [PMID: 32881766 DOI: 10.1097/htr.0000000000000606] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To compare plasma inflammatory biomarker concentrations to 6 months in young and older adults with and without mild traumatic brain injury (TBI). SETTING Level 1 trauma center. PARTICIPANTS Younger (21-54 years) and older (55+) adults diagnosed with mild TBI along with age-/sex-matched noninjured controls (n = 313). DESIGN Prospective cohort study. MAIN MEASURES Multiplex assays were used to quantify concentrations of selected plasma inflammatory markers at day 0, months 1 and 6. RESULTS Persistent aging-related differences were found between control groups in concentrations of 4 cytokines up to 6 months. At day 0, interleukin-6 (IL-6), IL-8, and fractalkine were higher in the older TBI compared with older control as well as the younger TBI groups, while IL-10 was higher in older TBI compared with controls. At month 1, significantly higher concentrations of IL-8, fractalkine, and tumor necrosis factor-α (TNF-α) were seen. At 6 months postinjury, significantly higher concentrations of IL-6 and IL-8 were seen, while a lower concentration of IL-7 was found in older versus younger TBI groups. CONCLUSION The neuroinflammatory signature that accompanies mild TBI in older adults differs from that of younger adults. The differences seen are notable for their roles in neutrophil attraction (IL-8), neuronal-microglial-immune cell interactions (fractalkine), and chronic inflammation (IL-6).
Collapse
|
4
|
Galera C, Barbosa S, Collet O, Khalfallah O, Aouizerate B, Sutter-Dalley AL, Koehl M, Capuron L, Van der Waerden J, Melchior M, Côté S, Heude B, Glaichenhaus N, Davidovic L. Cord Serum Cytokines at Birth and Children's Anxiety-Depression Trajectories From 3 to 8 Years: The EDEN Mother-Child Cohort. Biol Psychiatry 2021; 89:541-549. [PMID: 33349450 DOI: 10.1016/j.biopsych.2020.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/23/2020] [Accepted: 10/12/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Recent research suggests that immune dysregulation in pregnancy could be a risk factor for anxiety and depression symptoms in offspring. Whereas animal studies have demonstrated the importance of the link between perinatal cytokines and abnormal behaviors in offspring, human epidemiological studies in this area remain limited. The objectives of the study were to describe the network of cord serum cytokines at birth and test whether they are associated with subsequent anxiety and depression symptom trajectories in offspring. METHODS We used data and biological samples from 871 mother-child pairs followed up from pregnancy to 8 years of age and participating in the French mother-child cohort EDEN (a study on the pre- and early postnatal determinants of child health and development). Cord serum cytokines were measured at birth. Children's symptoms of anxiety and depression were assessed with the emotional difficulties subscore of the Strength and Difficulties Questionnaire at ages 3, 5, and 8 years, from which trajectories of anxiety-depression symptoms were derived. RESULTS Results showed a significant association between cord serum interleukin-7 at birth and the trajectories of children's anxiety-depression symptoms between ages 3 to 8 years (adjusted odds ratio, 0.73; 95% confidence interval, 0.57-0.93). The associations considered relevant confounders, including prenatal maternal depressive symptoms. CONCLUSIONS Early immune changes may contribute to subsequent anxiety and depression symptoms in childhood. Beyond the understanding of mechanisms underlying the occurrence of emotional difficulties in children, our findings open avenues for future research in human and animals.
Collapse
Affiliation(s)
- Cédric Galera
- Departments of Public Health and Neurocampus, University of Bordeaux, Bordeaux, France; Institut National de la Santé et de la Recherche Médicale, Bordeaux Population Health Center, Bordeaux, France; Centre Hospitalier Perrens, Bordeaux, France; Research Unit on Children's Psychosocial Maladjustment, Montreal, Quebec, Canada.
| | - Susana Barbosa
- Université Côte d'Azur, Nice, France; Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Ophélie Collet
- Departments of Public Health and Neurocampus, University of Bordeaux, Bordeaux, France; Research Unit on Children's Psychosocial Maladjustment, Montreal, Quebec, Canada
| | - Olfa Khalfallah
- Université Côte d'Azur, Nice, France; Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Bruno Aouizerate
- Departments of Public Health and Neurocampus, University of Bordeaux, Bordeaux, France; Centre Hospitalier Perrens, Bordeaux, France; Bordeaux INP, NutriNeuro UMR 1286, Bordeaux, France
| | - Anne-Laure Sutter-Dalley
- Institut National de la Santé et de la Recherche Médicale, Bordeaux Population Health Center, Bordeaux, France; Centre Hospitalier Perrens, Bordeaux, France
| | - Muriel Koehl
- Departments of Public Health and Neurocampus, University of Bordeaux, Bordeaux, France; Institut national de recherche pour l'agriculture, l'alimentation et l'environnement, Neurocentre Magendie U1215, and Institut National de la Santé et de la Recherche Médicale, Bordeaux, France
| | - Lucile Capuron
- Departments of Public Health and Neurocampus, University of Bordeaux, Bordeaux, France; Bordeaux INP, NutriNeuro UMR 1286, Bordeaux, France
| | - Judith Van der Waerden
- Institut Pierre Louis d'Epidémiologie et de Santé Publique, Sorbonne Université, Paris, France
| | - Maria Melchior
- Institut Pierre Louis d'Epidémiologie et de Santé Publique, Sorbonne Université, Paris, France
| | - Sylvana Côté
- Departments of Public Health and Neurocampus, University of Bordeaux, Bordeaux, France; Institut National de la Santé et de la Recherche Médicale, Bordeaux Population Health Center, Bordeaux, France; Research Unit on Children's Psychosocial Maladjustment, Montreal, Quebec, Canada; Department of Social and Preventive Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Barbara Heude
- Institut National de la Santé et de la Recherche Médicale, INSERM, UMR1153 Epidemiology and Biostatistics Sorbonne Paris Cité Center, ORCHAD Team, Paris, France; Institut National de la Santé et de la Recherche Médicale, Paris, France; Paris University, Paris, France
| | - Nicolas Glaichenhaus
- Université Côte d'Azur, Nice, France; Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Laetitia Davidovic
- Université Côte d'Azur, Nice, France; Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | | |
Collapse
|
5
|
A peripheral inflammatory signature discriminates bipolar from unipolar depression: A machine learning approach. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110136. [PMID: 33045321 DOI: 10.1016/j.pnpbp.2020.110136] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/04/2020] [Accepted: 10/06/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Mood disorders (major depressive disorder, MDD, and bipolar disorder, BD) are considered leading causes of life-long disability worldwide, where high rates of no response to treatment or relapse and delays in receiving a proper diagnosis (~60% of depressed BD patients are initially misdiagnosed as MDD) contribute to a growing personal and socio-economic burden. The immune system may represent a new target to develop novel diagnostic and therapeutic procedures but reliable biomarkers still need to be found. METHODS In our study we predicted the differential diagnosis of mood disorders by considering the plasma levels of 54 cytokines, chemokines and growth factors of 81 BD and 127 MDD depressed patients. Clinical diagnoses were predicted also against 32 healthy controls. Elastic net models, including 5000 non-parametric bootstrapping procedure and inner and outer 10-fold nested cross-validation were performed in order to identify the signatures for the disorders. RESULTS Results showed that the immune-inflammatory signature classifies the two disorders with a high accuracy (AUC = 97%), specifically 92% and 86% respectively for MDD and BD. MDD diagnosis was predicted by high levels of markers related to both pro-inflammatory (i.e. IL-1β, IL-6, IL-7, IL-16) and regulatory responses (IL-2, IL-4, and IL-10), whereas BD by high levels of inflammatory markers (CCL3, CCL4, CCL5, CCL11, CCL25, CCL27, CXCL11, IL-9 and TNF-α). CONCLUSIONS Our findings provide novel tools for early diagnosis of BD, strengthening the impact of biomarkers research into clinical practice, and new insights for the development of innovative therapeutic strategies for depressive disorders.
Collapse
|
6
|
Bednarz-Misa I, Bromke MA, Krzystek-Korpacka M. Interleukin (IL)-7 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1290:9-49. [PMID: 33559853 DOI: 10.1007/978-3-030-55617-4_2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Interleukin (IL)-7 plays an important immunoregulatory role in different types of cells. Therefore, it attracts researcher's attention, but despite the fact, many aspects of its modulatory action, as well as other functionalities, are still poorly understood. The review summarizes current knowledge on the interleukin-7 and its signaling cascade in context of cancer development. Moreover, it provides a cancer-type focused description of the involvement of IL-7 in solid tumors, as well as hematological malignancies.The interleukin has been discovered as a growth factor crucial for the early lymphocyte development and supporting the growth of malignant cells in certain leukemias and lymphomas. Therefore, its targeting has been explored as a treatment modality in hematological malignancies, while the unique ability to expand lymphocyte populations selectively and without hyperinflammation has been used in experimental immunotherapies in patients with lymphopenia. Ever since the early research demonstrated a reduced growth of solid tumors in the presence of IL-7, the interleukin application in boosting up the anticancer immunity has been investigated. However, a growing body of evidence indicative of IL-7 upregulation in carcinomas, facilitating tumor growth and metastasis and aiding drug-resistance, is accumulating. It therefore becomes increasingly apparent that the response to the IL-7 stimulus strongly depends on cell type, their developmental stage, and microenvironmental context. The interleukin exerts its regulatory action mainly through phosphorylation events in JAK/STAT and PI3K/Akt pathways, while the significance of MAPK pathway seems to be limited to solid tumors. Given the unwavering interest in IL-7 application in immunotherapy, a better understanding of interleukin role, source in tumor microenvironment, and signaling pathways, as well as the identification of cells that are likely to respond should be a research priority.
Collapse
Affiliation(s)
- Iwona Bednarz-Misa
- Department of Medical Biochemistry, Wroclaw Medical University, Wroclaw, Poland
| | - Mariusz A Bromke
- Department of Medical Biochemistry, Wroclaw Medical University, Wroclaw, Poland
| | | |
Collapse
|
7
|
Pignataro G, Cataldi M, Taglialatela M. Neurological risks and benefits of cytokine-based treatments in coronavirus disease 2019: from preclinical to clinical evidence. Br J Pharmacol 2021; 179:2149-2174. [PMID: 33512003 DOI: 10.1111/bph.15397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 12/15/2022] Open
Abstract
Immunodeficiency and hyperinflammation are responsible for the most frequent and life-threatening forms of coronavirus disease 2019 (COVID-19). Therefore, cytokine-based treatments targeting immuno-inflammatory mechanisms are currently undergoing clinical scrutiny in COVID-19-affected patients. In addition, COVID-19 patients also exhibit a wide range of neurological manifestations (neuro-COVID), which may also benefit from cytokine-based treatments. In fact, such drugs have shown some clinical efficacy also in neuroinflammatory diseases. On the other hand, anti-cytokine drugs are endowed with significant neurological risks, mainly attributable to their immunodepressant effects. Therefore, the aim of the present manuscript is to briefly describe the role of specific cytokines in neuroinflammation, to summarize the efficacy in preclinical models of neuroinflammatory diseases of drugs targeting these cytokines and to review the clinical data regarding the neurological effects of these drugs currently being investigated against COVID-19, in order to raise awareness about their potentially beneficial and/or detrimental neurological consequences.
Collapse
Affiliation(s)
- Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| | - Mauro Cataldi
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| | - Maurizio Taglialatela
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
8
|
Mortimer N, Sánchez-Mora C, Rovira P, Vilar-Ribó L, Richarte V, Corrales M, Fadeuilhe C, Rivero O, Lesch KP, Casas M, Ramos-Quiroga JA, Artigas MS, Ribasés M. Transcriptome profiling in adult attention-deficit hyperactivity disorder. Eur Neuropsychopharmacol 2020; 41:160-166. [PMID: 33221139 DOI: 10.1016/j.euroneuro.2020.11.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 10/04/2020] [Accepted: 11/02/2020] [Indexed: 01/27/2023]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder with an estimated heritability of around 70%. Although the largest genome-wide association study (GWAS) meta-analysis on ADHD identified independent loci conferring risk to the disorder, the molecular mechanisms underlying the genetic basis of the disorder remain to be elucidated. To explore ADHD biology, we ran a two-step transcriptome profiling in peripheral blood mononuclear cells (PBMCs) of 143 ADHD subjects and 169 healthy controls. Through this exploratory study we found eight differentially expressed genes in ADHD. These results highlight promising candidate genes and gene pathways for ADHD and support the use of peripheral tissues to assess gene expression signatures for ADHD.
Collapse
Affiliation(s)
- Niall Mortimer
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Barcelona 08035, Spain; Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Germany
| | - Cristina Sánchez-Mora
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Barcelona 08035, Spain; Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Department of Genetics, Microbiology & Statistics, University of Barcelona, Barcelona, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Paula Rovira
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Barcelona 08035, Spain; Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Laura Vilar-Ribó
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Barcelona 08035, Spain; Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Vanesa Richarte
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Montse Corrales
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Christian Fadeuilhe
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Olga Rivero
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Germany; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Russia; Department of Neuroscience, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Netherlands
| | - Miguel Casas
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Barcelona 08035, Spain; Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Josep Antoni Ramos-Quiroga
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Barcelona 08035, Spain; Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María Soler Artigas
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Barcelona 08035, Spain; Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Department of Genetics, Microbiology & Statistics, University of Barcelona, Barcelona, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain.
| | - Marta Ribasés
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Barcelona 08035, Spain; Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Department of Genetics, Microbiology & Statistics, University of Barcelona, Barcelona, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
9
|
Guedj F, Siegel AE, Pennings JLA, Alsebaa F, Massingham LJ, Tantravahi U, Bianchi DW. Apigenin as a Candidate Prenatal Treatment for Trisomy 21: Effects in Human Amniocytes and the Ts1Cje Mouse Model. Am J Hum Genet 2020; 107:911-931. [PMID: 33098770 PMCID: PMC7675036 DOI: 10.1016/j.ajhg.2020.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022] Open
Abstract
Human fetuses with trisomy 21 (T21) have atypical brain development that is apparent sonographically in the second trimester. We hypothesize that by analyzing and integrating dysregulated gene expression and pathways common to humans with Down syndrome (DS) and mouse models we can discover novel targets for prenatal therapy. Here, we tested the safety and efficacy of apigenin, identified with this approach, in both human amniocytes from fetuses with T21 and in the Ts1Cje mouse model. In vitro, T21 cells cultured with apigenin had significantly reduced oxidative stress and improved antioxidant defense response. In vivo, apigenin treatment mixed with chow was administered prenatally to the dams and fed to the pups over their lifetimes. There was no significant increase in birth defects or pup deaths resulting from prenatal apigenin treatment. Apigenin significantly improved several developmental milestones and spatial olfactory memory in Ts1Cje neonates. In addition, we noted sex-specific effects on exploratory behavior and long-term hippocampal memory in adult mice, and males showed significantly more improvement than females. We demonstrated that the therapeutic effects of apigenin are pleiotropic, resulting in decreased oxidative stress, activation of pro-proliferative and pro-neurogenic genes (KI67, Nestin, Sox2, and PAX6), reduction of the pro-inflammatory cytokines INFG, IL1A, and IL12P70 through the inhibition of NFκB signaling, increase of the anti-inflammatory cytokines IL10 and IL12P40, and increased expression of the angiogenic and neurotrophic factors VEGFA and IL7. These studies provide proof of principle that apigenin has multiple therapeutic targets in preclinical models of DS.
Collapse
Affiliation(s)
- Faycal Guedj
- Prenatal Genomics and Therapy Section, Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA; Mother Infant Research Institute, Tufts Medical Center and Tufts Children's Hospital, Boston, MA 02111, USA.
| | - Ashley E Siegel
- Prenatal Genomics and Therapy Section, Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA; Mother Infant Research Institute, Tufts Medical Center and Tufts Children's Hospital, Boston, MA 02111, USA
| | - Jeroen L A Pennings
- Center for Health Protection, National Institute for Public Health and the Environment, Bilthoven, BA 3720, the Netherlands
| | - Fatimah Alsebaa
- Mother Infant Research Institute, Tufts Medical Center and Tufts Children's Hospital, Boston, MA 02111, USA
| | - Lauren J Massingham
- Mother Infant Research Institute, Tufts Medical Center and Tufts Children's Hospital, Boston, MA 02111, USA
| | - Umadevi Tantravahi
- Department of Pathology, Women and Infants' Hospital, Providence, RI 02912, USA
| | - Diana W Bianchi
- Prenatal Genomics and Therapy Section, Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA; Mother Infant Research Institute, Tufts Medical Center and Tufts Children's Hospital, Boston, MA 02111, USA.
| |
Collapse
|
10
|
Giloteaux L, O'Neal A, Castro-Marrero J, Levine SM, Hanson MR. Cytokine profiling of extracellular vesicles isolated from plasma in myalgic encephalomyelitis/chronic fatigue syndrome: a pilot study. J Transl Med 2020; 18:387. [PMID: 33046133 PMCID: PMC7552484 DOI: 10.1186/s12967-020-02560-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 10/03/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a debilitating disease of unknown etiology lasting for a minimum of 6 months but usually for many years, with features including fatigue, cognitive impairment, myalgias, post-exertional malaise, and immune system dysfunction. Dysregulation of cytokine signaling could give rise to many of these symptoms. Cytokines are present in both plasma and extracellular vesicles, but little investigation of EVs in ME/CFS has been reported. Therefore, we aimed to characterize the content of extracellular vesicles (EVs) isolated from plasma (including circulating cytokine/chemokine profiling) from individuals with ME/CFS and healthy controls. METHODS We included 35 ME/CFS patients and 35 controls matched for age, sex and BMI. EVs were enriched from plasma by using a polymer-based precipitation method and characterized by Nanoparticle Tracking Analysis (NTA), Transmission Electron Microscopy (TEM) and immunoblotting. A 45-plex immunoassay was used to determine cytokine levels in both plasma and isolated EVs from a subset of 19 patients and controls. Linear regression, principal component analysis and inter-cytokine correlations were analyzed. RESULTS ME/CFS individuals had significantly higher levels of EVs that ranged from 30 to 130 nm in size as compared to controls, but the mean size for total extracellular vesicles did not differ between groups. The enrichment of typical EV markers CD63, CD81, TSG101 and HSP70 was confirmed by Western blot analysis and the morphology assessed by TEM showed a homogeneous population of vesicles in both groups. Comparison of cytokine concentrations in plasma and isolated EVs of cases and controls yielded no significant differences. Cytokine-cytokine correlations in plasma revealed a significant higher number of interactions in ME/CFS cases along with 13 inverse correlations that were mainly driven by the Interferon gamma-induced protein 10 (IP-10), whereas in the plasma of controls, no inverse relationships were found across any of the cytokines. Network analysis in EVs from controls showed 2.5 times more significant inter-cytokine interactions than in the ME/CFS group, and both groups presented a unique negative association. CONCLUSIONS Elevated levels of 30-130 nm EVs were found in plasma from ME/CFS patients and inter-cytokine correlations revealed unusual regulatory relationships among cytokines in the ME/CFS group that were different from the control group in both plasma and EVs. These disturbances in cytokine networks are further evidence of immune dysregulation in ME/CFS.
Collapse
Affiliation(s)
- Ludovic Giloteaux
- Department of Molecular Biology and Genetics, Cornell University, 323 Biotechnology Building, 526 Campus Road, Ithaca, NY, 14853, USA
| | - Adam O'Neal
- Department of Molecular Biology and Genetics, Cornell University, 323 Biotechnology Building, 526 Campus Road, Ithaca, NY, 14853, USA
| | - Jesús Castro-Marrero
- Department of Molecular Biology and Genetics, Cornell University, 323 Biotechnology Building, 526 Campus Road, Ithaca, NY, 14853, USA
- CFS/ME Unit, Division of Rheumatology, Vall d'Hebron University Hospital Research Institute, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain
| | | | - Maureen R Hanson
- Department of Molecular Biology and Genetics, Cornell University, 323 Biotechnology Building, 526 Campus Road, Ithaca, NY, 14853, USA.
| |
Collapse
|
11
|
Immune activity at birth and later psychopathology in childhood. Brain Behav Immun Health 2020; 8:100141. [PMID: 34589885 PMCID: PMC8474670 DOI: 10.1016/j.bbih.2020.100141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 12/28/2022] Open
Abstract
Disruption of neurodevelopmental trajectories can alter brain circuitry and increase the risk of psychopathology later in life. While preclinical studies have demonstrated that the immune system and cytokines influence neurodevelopment, whether immune activity and in particular which cytokines at birth are associated with psychopathology remains poorly explored in children. We used data and biological samples from 869 mother-child pairs participating in the French mother-child cohort EDEN. As proxies for immune activity at birth, we measured the levels of 27 cytokines in umbilical cord blood sera (CBS). We then explored the association between CBS cytokine levels and five psychopathological dimensions assessed in 5-year-old children using the Strengths and Difficulties Questionnaire (SDQ). Five cytokines were positively associated with psychopathology: C-X-C motif chemokine Ligand (CXCL)10, interleukin (IL)-10 and IL-12p40 with emotional symptoms, C–C motif chemokine Ligand (CCL)11 with conduct problems, and CCL11, and IL-17A with peer relationships problems. In contrast, seven cytokines were negatively associated with psychopathology: IL-7, IL-15 and Tumor Necrosis Factor (TNF)-β with emotional symptoms, CCL4 and IL-6 with conduct problems, CCL26 and IL-15 with peer relationships problems, and CCL26, IL-7, IL-15, and TNF-α with abnormal prosocial behavior. Without implying causation, these associations support the notion that cytokines influence neurodevelopment in humans and the risk of psychopathology later in life. Twelve cytokines at birth are associated with psychopathology in 5-year-old children. IL-7, IL-10, IL-12p40, IL-15, TNF-β and CXCL10 are associated with emotional symptoms. IL-6, CCL4 and CCL11 are associated with conduct problems. IL-15, IL-17A, CCL11 and CCL26 are associated with peer relationship problems. IL-7, IL-15, TNF-α and CCL26 are associated with prosocial behavior.
Collapse
|
12
|
Marković I, Savvides SN. Modulation of Signaling Mediated by TSLP and IL-7 in Inflammation, Autoimmune Diseases, and Cancer. Front Immunol 2020; 11:1557. [PMID: 32849527 PMCID: PMC7396566 DOI: 10.3389/fimmu.2020.01557] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/12/2020] [Indexed: 12/30/2022] Open
Abstract
Thymic Stromal Lymphopoietin (TSLP) and Interleukin-7 (IL-7) are widely studied cytokines within distinct branches of immunology. On one hand, TSLP is crucially important for mediating type 2 immunity at barrier surfaces and has been linked to widespread allergic and inflammatory diseases of the airways, skin, and gut. On the other hand, IL-7 operates at the foundations of T-cell and innate lymphoid cell (ILC) development and homeostasis and has been associated with cancer. Yet, TSLP and IL-7 are united by key commonalities in their structure and the structural basis of the receptor assemblies they mediate to initiate cellular signaling, in particular their cross-utilization of IL-7Rα. As therapeutic targeting of TSLP and IL-7 via diverse approaches is reaching advanced stages and in light of the plethora of mechanistic and structural data on receptor signaling mediated by the two cytokines, the time is ripe to provide integrated views of such knowledge. Here, we first discuss the major pathophysiological roles of TSLP and IL-7 in autoimmune diseases, inflammation and cancer. Subsequently, we curate structural and mechanistic knowledge about receptor assemblies mediated by the two cytokines. Finally, we review therapeutic avenues targeting TSLP and IL-7 signaling. We envision that such integrated view of the mechanism, structure, and modulation of signaling assemblies mediated by TSLP and IL-7 will enhance and fine-tune the development of more effective and selective approaches to further interrogate the role of TSLP and IL-7 in physiology and disease.
Collapse
Affiliation(s)
- Iva Marković
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Savvas N Savvides
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
13
|
Martha SR, Cheng Q, Fraser JF, Gong L, Collier LA, Davis SM, Lukins D, Alhajeri A, Grupke S, Pennypacker KR. Expression of Cytokines and Chemokines as Predictors of Stroke Outcomes in Acute Ischemic Stroke. Front Neurol 2020; 10:1391. [PMID: 32010048 PMCID: PMC6974670 DOI: 10.3389/fneur.2019.01391] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/18/2019] [Indexed: 12/30/2022] Open
Abstract
Introduction: Ischemic stroke remains one of the most debilitating diseases and is the fifth leading cause of death in the US. The ability to predict stroke outcomes within the acute period of stroke would be essential for care planning and rehabilitation. The Blood and Clot Thrombectomy Registry and Collaboration (BACTRAC; clinicaltrials.gov NCT03153683) study collects arterial blood immediately distal and proximal to the intracranial thrombus at the time of mechanical thrombectomy. These blood samples are an innovative resource in evaluating acute gene expression changes at the time of ischemic stroke. The purpose of this study was to identify inflammatory genes and important immune factors during mechanical thrombectomy for emergent large vessel occlusion (ELVO) and which patient demographics were predictors for stroke outcomes (infarct and/or edema volume) in acute ischemic stroke patients. Methods: The BACTRAC study is a non-probability sampling of male and female subjects (≥18 year old) treated with mechanical thrombectomy for ELVO. We evaluated 28 subjects (66 ± 15.48 years) relative concentrations of mRNA for gene expression in 84 inflammatory molecules in arterial blood distal and proximal to the intracranial thrombus who underwent thrombectomy. We used the machine learning method, Random Forest to predict which inflammatory genes and patient demographics were important features for infarct and edema volumes. To validate the overlapping genes with outcomes, we perform ordinary least squares regression analysis. Results: Machine learning analyses demonstrated that the genes and subject factors CCR4, IFNA2, IL-9, CXCL3, Age, T2DM, IL-7, CCL4, BMI, IL-5, CCR3, TNFα, and IL-27 predicted infarct volume. The genes and subject factor IFNA2, IL-5, CCL11, IL-17C, CCR4, IL-9, IL-7, CCR3, IL-27, T2DM, and CSF2 predicted edema volume. The overlap of genes CCR4, IFNA2, IL-9, IL-7, IL-5, CCR3, and IL-27 with T2DM predicted both infarct and edema volumes. These genes relate to a microenvironment for chemoattraction and proliferation of autoimmune cells, particularly Th2 cells and neutrophils. Conclusions: Machine learning algorithms can be employed to develop prognostic predictive biomarkers for stroke outcomes in ischemic stroke patients, particularly in regard to identifying acute gene expression changes that occur during stroke.
Collapse
Affiliation(s)
- Sarah R Martha
- School of Nursing, University of Washington, Seattle, WA, United States
| | - Qiang Cheng
- Institute for Biomedical Informatics, University of Kentucky, Lexington, KY, United States
| | - Justin F Fraser
- Department of Neurology, University of Kentucky, Lexington, KY, United States.,College of Medicine, University of Kentucky, Lexington, KY, United States.,Departments of Neurosurgery, University of Kentucky, Lexington, KY, United States.,Neuroscience, University of Kentucky, Lexington, KY, United States.,Radiology, University of Kentucky, Lexington, KY, United States
| | - Liyu Gong
- Institute for Biomedical Informatics, University of Kentucky, Lexington, KY, United States
| | - Lisa A Collier
- Department of Neurology, University of Kentucky, Lexington, KY, United States
| | - Stephanie M Davis
- Department of Neurology, University of Kentucky, Lexington, KY, United States
| | - Doug Lukins
- College of Medicine, University of Kentucky, Lexington, KY, United States.,Departments of Neurosurgery, University of Kentucky, Lexington, KY, United States.,Neuroscience, University of Kentucky, Lexington, KY, United States.,Radiology, University of Kentucky, Lexington, KY, United States
| | - Abdulnasser Alhajeri
- Department of Neurology, University of Kentucky, Lexington, KY, United States.,Radiology, University of Kentucky, Lexington, KY, United States
| | - Stephen Grupke
- Departments of Neurosurgery, University of Kentucky, Lexington, KY, United States.,Radiology, University of Kentucky, Lexington, KY, United States
| | - Keith R Pennypacker
- Department of Neurology, University of Kentucky, Lexington, KY, United States.,Neuroscience, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
14
|
Alterations in the Levels of Growth Factors in Adolescents with Major Depressive Disorder: A Longitudinal Study during the Treatment with Fluoxetine. Mediators Inflamm 2019; 2019:9130868. [PMID: 31827384 PMCID: PMC6885844 DOI: 10.1155/2019/9130868] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/14/2019] [Indexed: 12/26/2022] Open
Abstract
Major depressive disorder (MDD) has a prevalence of 5% in adolescents. Several studies have described the association between the inflammatory response and MDD, but little is known about the relationship between MDD and growth factors, such as IL-7, IL-9, IL-17A, VEGF, basic FGF, G-CSF, and GM-CSF. It must be appointed that there are scarce reports on growth factors in adolescents with MDD and even fewer with a clinical follow-up. In this work, we evaluated the levels of growth factors (IL-7, IL-9, IL-17A, VEGF, basic FGF, G-CSF, and GM-CSF) in MDD adolescents and the clinical follow-up during eight weeks of treatment with fluoxetine. Methods. All patients were diagnosed according to the DSM-IV-TR, and the severity of the symptoms was evaluated using the Hamilton Depression Rating Scale (HDRS). Growth factors IL-7, IL-9, IL-17A, VEGF, basic FGF, G-CSF, and GM-CSF were quantified by cytometric bead array using serum samples from 22 adolescents with MDD and 18 healthy volunteers. Results. All patients showed clinical improvement since the fourth week of pharmacological treatment according to the HDRS. Considerably higher levels of IL-7, IL-9, IL-17A, VEGF, basic FGF, G-CSF, and GM-CSF were detected in MDD adolescents as compared to healthy volunteers. A significant but temporal decrease was detected in basic FGF, G-CSF, and GM-CSF at week four of fluoxetine administration. Conclusions. To the best of our knowledge, this is the first report to show alterations in the levels of growth factors, such as IL-7, IL-9, IL-17A, VEGF, basic FGF, G-CSF, and GM-CSF in MDD adolescents during eight weeks of clinical follow-up. These disturbances might be involved in the physiopathology of MDD since such growth factors have been proven to participate in the neural development and correct functioning of the CNS; therefore, subtle alterations in it may contribute to MDD.
Collapse
|
15
|
Flip the coin: IL-7 and IL-7R in health and disease. Nat Immunol 2019; 20:1584-1593. [PMID: 31745336 DOI: 10.1038/s41590-019-0479-x] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 07/26/2019] [Indexed: 12/14/2022]
Abstract
The cytokine IL-7 and its receptor, IL-7R, are critical for T cell and, in the mouse, B cell development, as well as differentiation and survival of naive T cells, and generation and maintenance of memory T cells. They are also required for innate lymphoid cell (ILC) development and maintenance, and consequently for generation of lymphoid structures and barrier defense. Here we discuss the central role of IL-7 and IL-7R in the lymphoid system and highlight the impact of their deregulation, placing a particular emphasis on their 'dark side' as promoters of cancer development. We also explore therapeutic implications and opportunities associated with either positive or negative modulation of the IL-7-IL-7R signaling axis.
Collapse
|
16
|
Kermarrec L, Durand T, Gonzales J, Pabois J, Hulin P, Neunlist M, Neveu I, Naveilhan P. Rat enteric glial cells express novel isoforms of Interleukine-7 regulated during inflammation. Neurogastroenterol Motil 2019; 31:e13467. [PMID: 30240048 DOI: 10.1111/nmo.13467] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 07/26/2018] [Accepted: 08/14/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Neuroimmune interactions are essential to maintain gut homeostasis and prevent intestinal disorders but so far, the impact of enteric glial cells (EGC) on immune cells remains a relatively unexplored area of research. As a dysregulation of critical cytokines such as interleukine-7 (IL-7) was suggested to exacerbate gut chronic inflammation, we investigated whether EGC could be a source of IL-7 in the gastrointestinal tract. METHODS Expression of IL-7 in the rat enteric nervous system was analyzed by immunochemistry and Q-PCR. IL-7 variants were cloned and specific antibodies against rat IL-7 isoforms were raised to characterize their expression in the submucosal plexus. IL-7 isoforms were produced in vitro to analyze their impact on T-cell survival. KEY RESULTS Neurons and glial cells of the rat enteric nervous system expressed IL-7 at both mRNA and protein levels. Novel rat IL-7 isoforms with distinct C-terminal parts were detected. Three of these isoforms were found in EGC or in both enteric neurons and EGC. Exposure of EGC to pro-inflammatory cytokines (IL-1β and/or TNFα) induced an upregulation of all IL-7 isoforms. Interestingly, time-course and intensity of the upregulation varied according to the presence or absence of exon 5a in IL-7 variants. Functional analysis on T lymphocytes revealed that only canonical IL-7 protects T cells from cell death. CONCLUSIONS AND INFERENCES IL-7 and its variants are expressed by neurons and glial cells in the enteric nervous system. Their distinct expression and upregulation in inflammatory conditions suggest a role in gut homeostasis which could be critical in case of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Laetitia Kermarrec
- Université de Nantes, INSERM, Institut des Maladies de l'Appareil Digestif, The enteric nervous system in gut and brain disorders, Nantes, France
| | - Tony Durand
- Université de Nantes, INSERM, Institut des Maladies de l'Appareil Digestif, The enteric nervous system in gut and brain disorders, Nantes, France
| | - Jacques Gonzales
- Université de Nantes, INSERM, Institut des Maladies de l'Appareil Digestif, The enteric nervous system in gut and brain disorders, Nantes, France
| | - Julie Pabois
- Université de Nantes, INSERM, Institut des Maladies de l'Appareil Digestif, The enteric nervous system in gut and brain disorders, Nantes, France
| | | | - Michel Neunlist
- Université de Nantes, INSERM, Institut des Maladies de l'Appareil Digestif, The enteric nervous system in gut and brain disorders, Nantes, France
| | - Isabelle Neveu
- Université de Nantes, INSERM, Institut des Maladies de l'Appareil Digestif, The enteric nervous system in gut and brain disorders, Nantes, France
| | - Philippe Naveilhan
- Université de Nantes, INSERM, Institut des Maladies de l'Appareil Digestif, The enteric nervous system in gut and brain disorders, Nantes, France
| |
Collapse
|
17
|
Clark SL, Costin BN, Chan RF, Johnson AW, Xie L, Jurmain JL, Kumar G, Shabalin AA, Pandey AK, Aberg KA, Miles MF, van den Oord E. A Whole Methylome Study of Ethanol Exposure in Brain and Blood: An Exploration of the Utility of Peripheral Blood as Proxy Tissue for Brain in Alcohol Methylation Studies. Alcohol Clin Exp Res 2018; 42:2360-2368. [PMID: 30320886 DOI: 10.1111/acer.13905] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 10/06/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND Recent reviews have highlighted the potential use of blood-based methylation biomarkers as diagnostic and prognostic tools of current and future alcohol use and addiction. Due to the substantial overlap that often exists between methylation patterns across different tissues, including blood and brain, blood-based methylation may track methylation changes in brain; however, little work has explored the overlap in alcohol-related methylation in these tissues. METHODS To study the effects of alcohol on the brain methylome and identify possible biomarkers of these changes in blood, we performed a methylome-wide association study in brain and blood from 40 male DBA/2J mice that received either an acute ethanol (EtOH) or saline intraperitoneal injection. To investigate all 22 million CpGs in the mouse genome, we enriched for the methylated genomic fraction using methyl-CpG binding domain (MBD) protein capture followed by next-generation sequencing (MBD-seq). We performed association tests in blood and brain separately followed by enrichment testing to determine whether there was overlapping alcohol-related methylation in the 2 tissues. RESULTS The top result for brain was a CpG located in an intron of Ttc39b (p = 5.65 × 10-08 ), and for blood, the top result was located in Espnl (p = 5.11 × 10-08 ). Analyses implicated pathways involved in inflammation and neuronal differentiation, such as CXCR4, IL-7, and Wnt signaling. Enrichment tests indicated significant overlap among the top results in brain and blood. Pathway analyses of the overlapping genes converge on MAPKinase signaling (p = 5.6 × 10-05 ) which plays a central role in acute and chronic responses to alcohol and glutamate receptor pathways, which can regulate neuroplastic changes underlying addictive behavior. CONCLUSIONS Overall, we have shown some methylation changes in brain and blood after acute EtOH administration and that the changes in blood partly mirror the changes in brain suggesting the potential for DNA methylation in blood to be biomarkers of alcohol use.
Collapse
Affiliation(s)
- Shaunna L Clark
- Department of Psychology , Michigan State University, East Lansing, Michigan.,Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| | - Blair N Costin
- Department of Pharmacology and Toxicology , Virginia Commonwealth University, Richmond, Virginia
| | - Robin F Chan
- Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| | - Alexander W Johnson
- Department of Psychology , Michigan State University, East Lansing, Michigan
| | - Linying Xie
- Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| | - Jessica L Jurmain
- Department of Pharmacology and Toxicology , Virginia Commonwealth University, Richmond, Virginia
| | - Gaurav Kumar
- Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| | - Andrey A Shabalin
- Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| | - Ashutosh K Pandey
- Department of Anatomy and Neurobiology , Center for Integrative and Translational Genomics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Karolina A Aberg
- Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| | - Michael F Miles
- Department of Pharmacology and Toxicology , Virginia Commonwealth University, Richmond, Virginia
| | - Edwin van den Oord
- Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
18
|
Liu YW, Li S, Dai SS. Neutrophils in traumatic brain injury (TBI): friend or foe? J Neuroinflammation 2018; 15:146. [PMID: 29776443 PMCID: PMC5960133 DOI: 10.1186/s12974-018-1173-x] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/23/2018] [Indexed: 12/26/2022] Open
Abstract
Our knowledge of the pathophysiology about traumatic brain injury (TBI) is still limited. Neutrophils, as the most abundant leukocytes in circulation and the first-line transmigrated immune cells at the sites of injury, are highly involved in the initiation, development, and recovery of TBI. Nonetheless, our understanding about neutrophils in TBI is obsolete, and mounting evidences from recent studies have challenged the conventional views. This review summarizes what is known about the relationships between neutrophils and pathophysiology of TBI. In addition, discussions are made on the complex roles as well as the controversial views of neutrophils in TBI.
Collapse
Affiliation(s)
- Yang-Wuyue Liu
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, People's Republic of China.,Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA
| | - Song Li
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA
| | - Shuang-Shuang Dai
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, People's Republic of China. .,Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China.
| |
Collapse
|
19
|
Bal-Price A, Hogberg HT, Crofton KM, Daneshian M, FitzGerald RE, Fritsche E, Heinonen T, Hougaard Bennekou S, Klima S, Piersma AH, Sachana M, Shafer TJ, Terron A, Monnet-Tschudi F, Viviani B, Waldmann T, Westerink RHS, Wilks MF, Witters H, Zurich MG, Leist M. Recommendation on test readiness criteria for new approach methods in toxicology: Exemplified for developmental neurotoxicity. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2018; 35:306-352. [PMID: 29485663 DOI: 10.14573/altex.1712081] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/29/2018] [Indexed: 01/06/2023]
Abstract
Multiple non-animal-based test methods have never been formally validated. In order to use such new approach methods (NAMs) in a regulatory context, criteria to define their readiness are necessary. The field of developmental neurotoxicity (DNT) testing is used to exemplify the application of readiness criteria. The costs and number of untested chemicals are overwhelming for in vivo DNT testing. Thus, there is a need for inexpensive, high-throughput NAMs, to obtain initial information on potential hazards, and to allow prioritization for further testing. A background on the regulatory and scientific status of DNT testing is provided showing different types of test readiness levels, depending on the intended use of data from NAMs. Readiness criteria, compiled during a stakeholder workshop, uniting scientists from academia, industry and regulatory authorities are presented. An important step beyond the listing of criteria, was the suggestion for a preliminary scoring scheme. On this basis a (semi)-quantitative analysis process was assembled on test readiness of 17 NAMs with respect to various uses (e.g. prioritization/screening, risk assessment). The scoring results suggest that several assays are currently at high readiness levels. Therefore, suggestions are made on how DNT NAMs may be assembled into an integrated approach to testing and assessment (IATA). In parallel, the testing state in these assays was compiled for more than 1000 compounds. Finally, a vision is presented on how further NAM development may be guided by knowledge of signaling pathways necessary for brain development, DNT pathophysiology, and relevant adverse outcome pathways (AOP).
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission, Joint Research Centre (EC JRC), Ispra (VA), Italy
| | - Helena T Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Baltimore, MD, USA
| | - Kevin M Crofton
- National Centre for Computational Toxicology, US EPA, RTP, Washington, NC, USA
| | - Mardas Daneshian
- Center for Alternatives to Animal Testing, CAAT-Europe, University of Konstanz, Konstanz, Germany
| | - Rex E FitzGerald
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland
| | - Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine & Heinrich-Heine-University, Düsseldorf, Germany
| | - Tuula Heinonen
- Finnish Centre for Alternative Methods (FICAM), University of Tampere, Tampere, Finland
| | | | - Stefanie Klima
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Aldert H Piersma
- RIVM, National Institute for Public Health and the Environment, Bilthoven, and Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Magdalini Sachana
- Organisation for Economic Co-operation and Development (OECD), Paris, France
| | - Timothy J Shafer
- National Centre for Computational Toxicology, US EPA, RTP, Washington, NC, USA
| | | | - Florianne Monnet-Tschudi
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland.,Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Barbara Viviani
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Tanja Waldmann
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Remco H S Westerink
- Neurotoxicology Research Group, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Martin F Wilks
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland
| | - Hilda Witters
- VITO, Flemish Institute for Technological Research, Unit Environmental Risk and Health, Mol, Belgium
| | - Marie-Gabrielle Zurich
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland.,Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Marcel Leist
- Center for Alternatives to Animal Testing, CAAT-Europe, University of Konstanz, Konstanz, Germany.,In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
20
|
Alternative Pre-mRNA Splicing in Mammals and Teleost Fish: A Effective Strategy for the Regulation of Immune Responses Against Pathogen Infection. Int J Mol Sci 2017; 18:ijms18071530. [PMID: 28714877 PMCID: PMC5536018 DOI: 10.3390/ijms18071530] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/10/2017] [Accepted: 07/12/2017] [Indexed: 12/14/2022] Open
Abstract
Pre-mRNA splicing is the process by which introns are removed and the protein coding elements assembled into mature mRNAs. Alternative pre-mRNA splicing provides an important source of transcriptome and proteome complexity through selectively joining different coding elements to form mRNAs, which encode proteins with similar or distinct functions. In mammals, previous studies have shown the role of alternative splicing in regulating the function of the immune system, especially in the regulation of T-cell activation and function. As lower vertebrates, teleost fish mainly rely on a large family of pattern recognition receptors (PRRs) to recognize pathogen-associated molecular patterns (PAMPs) from various invading pathogens. In this review, we summarize recent advances in our understanding of alternative splicing of piscine PRRs including peptidoglycan recognition proteins (PGRPs), nucleotide binding and oligomerization domain (NOD)-like receptors (NLRs), retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) and their downstream signaling molecules, compared to splicing in mammals. We also discuss what is known and unknown about the function of splicing isoforms in the innate immune responses against pathogens infection in mammals and teleost fish. Finally, we highlight the consequences of alternative splicing in the innate immune system and give our view of important directions for future studies.
Collapse
|
21
|
Rane L, Rahman S, Magalhaes I, Ambati A, Andersson J, Zumla A, Brighenti S, Maeurer MJ. IL-7δ5 protein is expressed in human tissues and induces expression of the oxidized low density lipoprotein receptor 1 (OLR1) in CD14+ monocytes. Int J Infect Dis 2017; 59:29-36. [PMID: 28279736 DOI: 10.1016/j.ijid.2017.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES The 6-exon-spanning 'canonical' Interleukin-7 (IL-7c) is a non-redundant cytokine in human T-cell homeostasis that undergoes extensive alternative pre-mRNA splicing. The IL-7 gene variant lacking, exon 5 (IL-7δ5), exhibits agonistic effects as compared to IL-7c. We studied in this report for the first time the protein expression of IL-7δ5 variant in tissues and its role in monocyte activation. METHODS We visualized the expression of IL-7δ5 protein by immunohistochemistry in both healthy and malignant (human) tissues and investigated the impact of IL-7δ5 stimulation on CD14+ monocytes using gene expression analysis and flow cytometry. RESULTS IL-7δ5 is largely expressed by human epithelial cells, yet also by stromal cells in malignant lesions. Gene expression analysis in CD14+ monocytes, induced by the 6-exon spanning IL-7 or IL-7δ5 showed similar changes resulting in a pro-inflammatory phenotype and increased expression of genes involved in lipid metabolism. IL7δ5 was superior in inducing upregulation of the oxidised low density lipoprotein receptor (OLR), measured by flow cytometry, in CD14+ cells. CONCLUSION IL-7δ5, produced from non-transformed and transformed cells, may contribute to chronic inflammatory responses and development of 'foamy' cells by increased OLR1 expression that mediates increased oxLDL uptake.
Collapse
Affiliation(s)
- Lalit Rane
- Department of Microbiology Tumor and Cell Biology (MTC), Karolinska Institute, 171 77, Stockholm, Sweden.
| | - Sayma Rahman
- Center for Infectious Medicine (CIM), Karolinska Institute, 141 86 Stockholm, Sweden.
| | - Isabelle Magalhaes
- Division of Therapeutic Immunology (TIM), Department of Labroratory Medicine, Karolinska Institute, 141 86, Stockholm, Sweden.
| | - Aditya Ambati
- Division of Therapeutic Immunology (TIM), Department of Labroratory Medicine, Karolinska Institute, 141 86, Stockholm, Sweden; Department of Medicine, Karolinska Institute, 141 86, Stockholm, Sweden.
| | - Jan Andersson
- Center for Infectious Medicine (CIM), Karolinska Institute, 141 86 Stockholm, Sweden.
| | - Alimuddin Zumla
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, and NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, United Kingdom.
| | - Susanna Brighenti
- Center for Infectious Medicine (CIM), Karolinska Institute, 141 86 Stockholm, Sweden.
| | - Markus J Maeurer
- Department of Microbiology Tumor and Cell Biology (MTC), Karolinska Institute, 171 77, Stockholm, Sweden; Division of Therapeutic Immunology (TIM), Department of Labroratory Medicine, Karolinska Institute, 141 86, Stockholm, Sweden.
| |
Collapse
|
22
|
Aschner M, Ceccatelli S, Daneshian M, Fritsche E, Hasiwa N, Hartung T, Hogberg HT, Leist M, Li A, Mundi WR, Padilla S, Piersma AH, Bal-Price A, Seiler A, Westerink RH, Zimmer B, Lein PJ. Reference compounds for alternative test methods to indicate developmental neurotoxicity (DNT) potential of chemicals: example lists and criteria for their selection and use. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2016; 34:49-74. [PMID: 27452664 PMCID: PMC5250586 DOI: 10.14573/altex.1604201] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/09/2016] [Indexed: 11/23/2022]
Abstract
There is a paucity of information concerning the developmental neurotoxicity (DNT) hazard posed by industrial and environmental chemicals. New testing approaches will most likely be based on batteries of alternative and complementary (non-animal) tests. As DNT is assumed to result from the modulation of fundamental neurodevelopmental processes (such as neuronal differentiation, precursor cell migration or neuronal network formation) by chemicals, the first generation of alternative DNT tests target these processes. The advantage of such types of assays is that they capture toxicants with multiple targets and modes-of-action. Moreover, the processes modelled by the assays can be linked to toxicity endophenotypes, i.e. alterations in neural connectivity that form the basis for neurofunctional deficits in man. The authors of this review convened in a workshop to define criteria for the selection of positive/negative controls, to prepare recommendations on their use, and to initiate the setup of a directory of reference chemicals. For initial technical optimization of tests, a set of >50 endpoint-specific control compounds was identified. For further test development, an additional “test” set of 33 chemicals considered to act directly as bona fide DNT toxicants is proposed, and each chemical is annotated to the extent it fulfills these criteria. A tabular compilation of the original literature used to select the test set chemicals provides information on statistical procedures, and toxic/non-toxic doses (both for pups and dams). Suggestions are provided on how to use the >100 compounds (including negative controls) compiled here to address specificity, adversity and use of alternative test systems.
Collapse
Affiliation(s)
| | | | - Mardas Daneshian
- Center for Alternatives to Animal Testing-Europe (CAAT-Europe), University of Konstanz, Germany
| | - Ellen Fritsche
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Nina Hasiwa
- Center for Alternatives to Animal Testing-Europe (CAAT-Europe), University of Konstanz, Germany
| | - Thomas Hartung
- Center for Alternatives to Animal Testing-Europe (CAAT-Europe), University of Konstanz, Germany.,Center for Alternatives to Animal Testing (CAAT), The Johns Hopkins University, Baltimore, MD, USA
| | - Helena T Hogberg
- Center for Alternatives to Animal Testing (CAAT), The Johns Hopkins University, Baltimore, MD, USA
| | - Marcel Leist
- Center for Alternatives to Animal Testing-Europe (CAAT-Europe), University of Konstanz, Germany.,In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden Foundation at the University of Konstanz, Konstanz, Germany.,Konstanz Research School Chemical Biology (KoRS-CB), Konstanz University
| | - Abby Li
- Exponent Inc.,San Francisco, USA
| | - William R Mundi
- United States Environmental Protection Agency (USEPA), NHEERL, Research Triangle Park, NC, USA
| | - Stephanie Padilla
- United States Environmental Protection Agency (USEPA), NHEERL, Research Triangle Park, NC, USA
| | - Aldert H Piersma
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.,Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Anna Bal-Price
- European Commission Joint Research Centre, Institute for Health and Consumer Protection, Ispra, Italy
| | - Andrea Seiler
- Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Remco H Westerink
- Neurotoxicology Research Group, Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, The Netherlands
| | | | - Pamela J Lein
- Center for Research on Occupational and Environmental Toxicology, Oregon Health & Science University, Portland, USA.,Department of Molecular Biosciences, University of California, Davis, USA
| |
Collapse
|
23
|
Splicing Regulation of Pro-Inflammatory Cytokines and Chemokines: At the Interface of the Neuroendocrine and Immune Systems. Biomolecules 2015; 5:2073-100. [PMID: 26371053 PMCID: PMC4598789 DOI: 10.3390/biom5032073] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 08/28/2015] [Indexed: 01/13/2023] Open
Abstract
Alternative splicing plays a key role in posttranscriptional regulation of gene expression, allowing a single gene to encode multiple protein isoforms. As such, alternative splicing amplifies the coding capacity of the genome enormously, generates protein diversity, and alters protein function. More than 90% of human genes undergo alternative splicing, and alternative splicing is especially prevalent in the nervous and immune systems, tissues where cells need to react swiftly and adapt to changes in the environment through carefully regulated mechanisms of cell differentiation, migration, targeting, and activation. Given its prevalence and complexity, this highly regulated mode of gene expression is prone to be affected by disease. In the following review, we look at how alternative splicing of signaling molecules—cytokines and their receptors—changes in different pathological conditions, from chronic inflammation to neurologic disorders, providing means of functional interaction between the immune and neuroendocrine systems. Switches in alternative splicing patterns can be very dynamic and can produce signaling molecules with distinct or antagonistic functions and localization to different subcellular compartments. This newly discovered link expands our understanding of the biology of immune and neuroendocrine cells, and has the potential to open new windows of opportunity for treatment of neurodegenerative disorders.
Collapse
|
24
|
Zai CC, Gonçalves VF, Tiwari AK, Gagliano SA, Hosang G, de Luca V, Shaikh SA, King N, Chen Q, Xu W, Strauss J, Breen G, Lewis CM, Farmer AE, McGuffin P, Knight J, Vincent JB, Kennedy JL. A genome-wide association study of suicide severity scores in bipolar disorder. J Psychiatr Res 2015; 65:23-9. [PMID: 25917933 DOI: 10.1016/j.jpsychires.2014.11.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/16/2014] [Accepted: 11/13/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Suicide claims one million lives worldwide annually, making it a serious public health concern. The risk for suicidal behaviour can be partly explained by genetic factors, as suggested by twin and family studies (reviewed in (Zai et al. 2012)). Recently, genome-wide association studies (GWASs) of suicide attempt on large samples of bipolar disorder (BD) patients from multiple sites have identified a number of novel candidate genes. GWASs of suicide behaviour severity, from suicidal ideation to serious suicide attempt, have not been reported for BD. METHODS We conducted a GWAS of suicide behaviour severity in three independent BD samples:212 small nuclear families with BD probands from Toronto, Canada, 428 BD cases from Toronto, and 483 BD cases from the UK. We carried out imputation with 1000 Genome Project data as reference using IMPUTE2. Quality control and data analysis was conducted using PLINK and R. We conducted the quantitative analyses of suicide behaviour severity in the three samples separately, and derived an overall significance by a meta-analysis using the METAL software. RESULTS We did not find genome-wide significant association of any tested markers in any of the BD samples, but we found a number of suggestive associations, including regions on chromosomes 8 and 10 (p < 1e-5). CONCLUSIONS Our GWAS findings suggest that likely many gene variants of small effects contribute collectively to the risk for suicidal behaviour severity in BD. Larger independent replications are required to strengthen the findings from the GWAS presented here.
Collapse
Affiliation(s)
- Clement C Zai
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Vanessa F Gonçalves
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Arun K Tiwari
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Sarah A Gagliano
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Ontario, Canada
| | - Georgina Hosang
- Department of Psychology, Goldsmiths, University of London, New Cross, London, United Kingdom; MRC Social, Genetic & Developmental Psychiatry Centre, King's College London, London, United Kingdom
| | - Vincenzo de Luca
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Ontario, Canada
| | - Sajid A Shaikh
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Nicole King
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Qian Chen
- Cancer Care Ontario, Toronto, Ontario, Canada
| | - Wei Xu
- Dalla Lana School of Public Health, University of Toronto, Ontario, Canada; Ontario Cancer Institute, Prince Margaret Hospital, Toronto, Ontario, Canada
| | - John Strauss
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Gerome Breen
- MRC Social, Genetic & Developmental Psychiatry Centre, King's College London, London, United Kingdom
| | - Cathryn M Lewis
- MRC Social, Genetic & Developmental Psychiatry Centre, King's College London, London, United Kingdom
| | - Anne E Farmer
- MRC Social, Genetic & Developmental Psychiatry Centre, King's College London, London, United Kingdom
| | - Peter McGuffin
- MRC Social, Genetic & Developmental Psychiatry Centre, King's College London, London, United Kingdom
| | - Jo Knight
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Ontario, Canada
| | - John B Vincent
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Ontario, Canada; Molecular Neuropsychiatry and Development Laboratory (MiND), Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - James L Kennedy
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Ontario, Canada.
| |
Collapse
|
25
|
Radiolabeled semi-quantitative RT-PCR assay for the analysis of alternative splicing of interleukin genes. Methods Mol Biol 2015; 1172:343-62. [PMID: 24908320 DOI: 10.1007/978-1-4939-0928-5_31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alternative splicing evolved as a very efficient way to generate proteome diversity from a limited number of genes, while at the same time modulating posttranscriptional events of gene expression-such as stability, turnover, subcellular localization, binding properties, and general activity of both mRNAs and proteins. Since the vast majority of human genes undergo alternative splicing, it comes to no surprise that interleukin genes also show extensive alternative splicing. In fact, there is a growing body of evidence indicating that alternative splicing plays a central role in modulating the pleiotropic functions of cytokines, and aberrant expression of alternatively spliced interleukin mRNAs has been linked to disease. However, while several interleukin splice variants have been described, their function is still poorly understood. This is particularly relevant, since alternatively spliced cytokine isoforms can act both as disease biomarkers and as candidate entry points for therapeutic intervention. In this chapter we describe a protocol that uses radiolabeled semi-quantitative RT-PCR to efficiently detect, analyze, and quantify alternative splicing patterns of cytokine genes.
Collapse
|
26
|
Yu RY, Brazaitis J, Gallagher G. The human IL-23 receptor rs11209026 A allele promotes the expression of a soluble IL-23R-encoding mRNA species. THE JOURNAL OF IMMUNOLOGY 2014; 194:1062-8. [PMID: 25552541 DOI: 10.4049/jimmunol.1401850] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The human IL23R gene single nucleotide polymorphism rs11209026 A allele confers protection against inflammatory diseases. However, although this difference has been associated with reductions in IL-23-induced IL-17A production and STAT3 phosphorylation, the molecular mechanism underlying these changes remains undefined. Th17 cell maturation depends on IL-23 signaling. Multiple splice forms of the human IL23R transcript exist, and one, Δ9, encodes a soluble form of the receptor. In this study, we asked whether this protective allele was associated with mRNA splicing. Using mini-gene constructs and competitive oligonucleotide binding, we showed that the A allele alters IL-23R α-chain mRNA splicing and favors exon 9 skipping by reducing the binding of the splicing enhancer SF2. This enhances expression of the Δ9 mRNA and consequently diminishes IL-23 signaling. Thus, the presence of the A allele increases expression of the soluble form of IL23R mRNA (which then functions as a decoy receptor) and lowers the ability to develop a Th17 phenotype upon IL-23 stimulation. We further showed that antisense oligonucleotides targeting the SF2 binding site could efficiently induce exon 9 skipping in the presence of the G allele, and thereby replicate the effect of the A allele. Antisense oligonucleotide treatment caused dose-responsive induction of the IL23RΔ9 mRNA and interfered with in vitro differentiation of human Th17 cells, reducing their expression of the signature Th17 cytokines IL-17A and IL-17F. This may represent a novel approach to therapy of Th17-mediated diseases by elevating soluble IL-23R while simultaneously reducing the remaining cell surface receptor density.
Collapse
Affiliation(s)
- Raymond Y Yu
- Genetic Immunology Laboratory, HUMIGEN, The Institute for Genetic Immunology, Genesis Biotechnology Group, Hamilton, NJ 08690
| | - Jonathan Brazaitis
- Genetic Immunology Laboratory, HUMIGEN, The Institute for Genetic Immunology, Genesis Biotechnology Group, Hamilton, NJ 08690
| | - Grant Gallagher
- Genetic Immunology Laboratory, HUMIGEN, The Institute for Genetic Immunology, Genesis Biotechnology Group, Hamilton, NJ 08690
| |
Collapse
|
27
|
Diverse interleukin-7 mRNA transcripts in Chinese tree shrew (Tupaia belangeri chinensis). PLoS One 2014; 9:e99859. [PMID: 24945249 PMCID: PMC4063794 DOI: 10.1371/journal.pone.0099859] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/18/2014] [Indexed: 11/21/2022] Open
Abstract
Interleukin-7 (IL7) is a pleiotropic cytokine that is actively involved in the immune system. The Chinese tree shrew (Tupaia belangeri chinensis) has been proposed as an alternative experimental animal to primates in biomedical research. However, there is a lack of biological knowledge about the immune system of the tree shrew. In this study, we cloned the IL7 gene (tIL7) in the Chinese tree shrew and quantified the expression of mRNA transcripts in eight tissues (heart, liver, spleen, lung, kidney, intestine, skeletal muscle and brain) from 20 individuals. Eleven tIL7 mRNA transcripts were identified in different tissues. The canonical form (tIL7c) had a length of 1817 bp and encoded a predicted gene product with 177 amino acids. Phylogenetic analyses based on the amino acid sequences revealed a considerably large genetic difference between tree shrew and human. Quantification of mRNA expression of transcripts tIL7c, tIL7-sv1, tIL7-sv2 and tIL7-sv3 showed that these transcripts were expressed in all tissues, albeit the expression levels varied in different tissues. Transcripts tIL7c, tIL7-sv1, and tIL7-sv2 had the lowest expression in brain, and tIL7-sv3 had a dramatically high mRNA expression in skeletal muscle and heart. The mRNA expression levels of tIL7c and tIL7-sv1 were significantly increased upon ploy(I:C) stimulation in tree shrew primary renal cells. As with human full-length IL7, tIL7c, tIL7-sv1, tIL7-sv2 and tIL7-sv3 showed similar a subcellular localization pattern. Our results identified diverse tIL7 transcripts in the Chinese tree shrew, which may play a potential role in modulating IL7-regulated biological effects.
Collapse
|
28
|
BDE-47 and 6-OH-BDE-47 modulate calcium homeostasis in primary fetal human neural progenitor cells via ryanodine receptor-independent mechanisms. Arch Toxicol 2014; 88:1537-48. [PMID: 24599297 DOI: 10.1007/s00204-014-1217-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 02/19/2014] [Indexed: 02/04/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) are bioaccumulating flame retardants found in rising concentrations in human tissue. Epidemiological and animal studies have raised concern for their potential to induce developmental neurotoxicity (DNT). Considering the essential role of calcium homeostasis in neurodevelopment, PBDE-induced disturbance of intracellular calcium concentration ([Ca(2+)]i) may underlie PBDE-induced DNT. To test this hypothesis, we investigated acute effects of BDE-47 and 6-OH-BDE-47 on [Ca(2+)]i in human neural progenitor cells (hNPCs) and unraveled involved signaling pathways. Short-time differentiated hNPCs were exposed to BDE-47, 6-OH-BDE-47, and multiple inhibitors/stimulators of presumably involved signaling pathways to determine possible effects on [Ca(2+)]i by single-cell microscopy with the fluorescent dye Fura-2. Initial characterization of calcium signaling pathways confirmed the early developmental stage of hNPCs. In these cells, BDE-47 (2 μM) and 6-OH-BDE-47 (0.2 μM) induce [Ca(2+)]i transients. This increase in [Ca(2+)]i is due to extracellular Ca(2+) influx and intracellular release of Ca(2+), mainly from the endoplasmic reticulum (ER). While extracellular Ca(2+) seems to enter the cytoplasm upon 6-OH-BDE-47 by interfering with the cell membrane and independent of Ca(2+) ion channels, ER-derived Ca(2+) is released following activation of protein lipase C and inositol 1,4,5-trisphosphate receptor, but independently of ryanodine receptors. These findings illustrate that immature developing hNPCs respond to low concentrations of 6-OH-BDE-47 by an increase in [Ca(2+)]i and provide new mechanistic explanations for such BDE-induced calcium disruption. Thus, these data support the possibility of a critical window of PBDE exposure, i.e., early human brain development, which has to be acknowledged in risk assessment.
Collapse
|
29
|
Mizejewski GJ, Lindau-Shepard B, Pass KA. Newborn screening for autism: in search of candidate biomarkers. Biomark Med 2013; 7:247-60. [PMID: 23547820 DOI: 10.2217/bmm.12.108] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) represents a wide range of neurodevelopmental disorders characterized by impairments in social interaction, language, communication and range of interests. Autism is usually diagnosed in children 3-5 years of age using behavioral characteristics; thus, diagnosis shortly after birth would be beneficial for early initiation of treatment. AIM This retrospective study sought to identify newborns at risk for ASD utilizing bloodspot specimens in an immunoassay. MATERIALS & METHODS The present study utilized stored frozen specimens from ASD children already diagnosed at 15-36 months of age. The newborn specimens and controls were analyzed by immunoassay in a multiplex system that included 90 serum biomarkers and subjected to statisical analysis. RESULTS Three sets of five biomarkers associated with ASD were found that differed from control groups. The 15 candidate biomarkers were then discussed regarding their association with ASD. CONCLUSION This study determined that a statistically selected panel of 15 biomarkers successfully discriminated presumptive newborns at risk for ASD from those of nonaffected controls.
Collapse
Affiliation(s)
- Gerald J Mizejewski
- Division of Translational Medicine, Wadsworth Center, NYS Department of Health, PO Box 509, Albany, NY 12201 0509, USA.
| | | | | |
Collapse
|
30
|
Kan AA, de Jager W, de Wit M, Heijnen C, van Zuiden M, Ferrier C, van Rijen P, Gosselaar P, Hessel E, van Nieuwenhuizen O, de Graan PNE. Protein expression profiling of inflammatory mediators in human temporal lobe epilepsy reveals co-activation of multiple chemokines and cytokines. J Neuroinflammation 2012; 9:207. [PMID: 22935090 PMCID: PMC3489559 DOI: 10.1186/1742-2094-9-207] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 07/30/2012] [Indexed: 11/25/2022] Open
Abstract
Mesial temporal lobe epilepsy (mTLE) is a chronic and often treatment-refractory brain disorder characterized by recurrent seizures originating from the hippocampus. The pathogenic mechanisms underlying mTLE remain largely unknown. Recent clinical and experimental evidence supports a role of various inflammatory mediators in mTLE. Here, we performed protein expression profiling of 40 inflammatory mediators in surgical resection material from mTLE patients with and without hippocampal sclerosis, and autopsy controls using a multiplex bead-based immunoassay. In mTLE patients we identified 21 upregulated inflammatory mediators, including 10 cytokines and 7 chemokines. Many of these upregulated mediators have not previously been implicated in mTLE (for example, CCL22, IL-7 and IL-25). Comparing the three patient groups, two main hippocampal expression patterns could be distinguished, pattern I (for example, IL-10 and IL-25) showing increased expression in mTLE + HS patients compared to mTLE-HS and controls, and pattern II (for example, CCL4 and IL-7) showing increased expression in both mTLE groups compared to controls. Upregulation of a subset of inflammatory mediators (for example, IL-25 and IL-7) could not only be detected in the hippocampus of mTLE patients, but also in the neocortex. Principle component analysis was used to cluster the inflammatory mediators into several components. Follow-up analyses of the identified components revealed that the three patient groups could be discriminated based on their unique expression profiles. Immunocytochemistry showed that IL-25 IR (pattern I) and CCL4 IR (pattern II) were localized in astrocytes and microglia, whereas IL-25 IR was also detected in neurons. Our data shows co-activation of multiple inflammatory mediators in hippocampus and neocortex of mTLE patients, indicating activation of multiple pro- and anti-epileptogenic immune pathways in this disease.
Collapse
Affiliation(s)
- Anne A Kan
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Wilco de Jager
- Department of Pediatric Immunology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, The Netherlands
| | - Marina de Wit
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Cobi Heijnen
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Mirjam van Zuiden
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Cyrill Ferrier
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, The Netherlands
| | - Peter van Rijen
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, The Netherlands
| | - Peter Gosselaar
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, The Netherlands
| | - Ellen Hessel
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Onno van Nieuwenhuizen
- Department of Child Neurology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, The Netherlands
| | - Pierre N E de Graan
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| |
Collapse
|
31
|
Pavlica S, Milosevic J, Keller M, Schulze M, Peinemann F, Piscioneri A, De Bartolo L, Darsow K, Bartel S, Lange HA, Bader A. Erythropoietin enhances cell proliferation and survival of human fetal neuronal progenitors in normoxia. Brain Res 2012; 1452:18-28. [DOI: 10.1016/j.brainres.2012.02.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 02/10/2012] [Accepted: 02/17/2012] [Indexed: 12/11/2022]
|
32
|
Expression and function of interleukin-7 in secondary and tertiary lymphoid organs. Semin Immunol 2012; 24:175-89. [PMID: 22444422 DOI: 10.1016/j.smim.2012.02.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 01/06/2012] [Accepted: 02/15/2012] [Indexed: 12/23/2022]
Abstract
Interleukin-7 (IL-7) is known since many years as stromal-cell derived cytokine that plays a key role for the adaptive immune system. It promotes lymphocyte development in the bone marrow and thymus as well as naive and memory T cell homeostasis in the periphery. More recently, IL-7 reporter mice and other approaches have led to the further characterization of the various stromal cell sources of IL-7 in secondary lymphoid organs (SLO) and other tissues. We will review these advances along with a discussion of the regulation of IL-7 and its receptor, and compare the biological effects IL-7 has on adaptive as well as innate immune cells in SLO. Finally, we will review the role of IL-7 in development of SLO and tertiary lymphoid tissues that frequently are associated with sites of chronic inflammation.
Collapse
|
33
|
Chitty DW, Tremblay RG, Ribecco-Lutkiewicz M, Haukenfrers J, Zurakowski B, Massie B, Sikorska M, Bani-Yaghoub M. Development of BMP7-producing human cells, using a third generation lentiviral gene delivery system. J Neurosci Methods 2011; 205:17-27. [PMID: 22209770 DOI: 10.1016/j.jneumeth.2011.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 12/13/2011] [Accepted: 12/14/2011] [Indexed: 12/16/2022]
Abstract
Bone morphogenetic protein 7 (BMP7), a member of the transforming growth factor β (TGF-β) superfamily, plays important roles in the development of various tissues and organs in mouse and human. In particular, BMP7 is critical for the formation of the nervous system and it is considered to have therapeutic potential in brain injury and stroke. One approach to make BMP7 more suitable for therapeutic purposes is the development of efficient vectors that allow the consistent, reliable and cost-effective production of the BMP7 protein. In this study, we developed an efficient BMP7 delivery system, using a third generation lentiviral vector to produce functional BMP7 protein. The lentiviral transduction of several human cell types, including human embryonic kidney 293 (HEK293) cells, amniotic fluid cells, NTera2 neurons (NT2-N) and primary neuronal cultures resulted in BMP7 expression. The production of BMP7 protein was achieved for at least 4 weeks post-transduction, as determined by enzyme-linked immunosorbent assay (ELISA). SMAD phosphorylation and neuronal differentiation assays verified the bioactivity and functionality of the lentiviral-based BMP7 protein, respectively. In addition, the intracerebroventricular injection of the lentivirus resulted in exogenous BMP7 expression in both neurons and astrocytes in the mouse brain. Taken together, this gene delivery system provides a reliable source of functional BMP7 protein for future in vitro and in vivo studies.
Collapse
Affiliation(s)
- David W Chitty
- Neurogenesis and Brain Repair Group, Neurobiology Program, Institute for Biological Sciences, National Research Council Canada, Ottawa, ON K1A 0R6, Canada
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Moors M, Bose R, Johansson-Haque K, Edoff K, Okret S, Ceccatelli S. Dickkopf 1 mediates glucocorticoid-induced changes in human neural progenitor cell proliferation and differentiation. Toxicol Sci 2011; 125:488-95. [PMID: 22048647 DOI: 10.1093/toxsci/kfr304] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Glucocorticoids (GC) are critical for normal development of the fetal brain, and alterations in their levels can induce neurotoxicity with detrimental consequences. Still, there is little information available on the effects of GC on human neural stem/progenitor cells (hNPC). In the present study, we have investigated the effects of the synthetic GC dexamethasone (Dex) on hNPC grown as neurospheres, with special focus on their proliferation and differentiation capacity and the underlying molecular mechanisms. Immunocytochemical stainings showed that Dex markedly decreases proliferation and neuronal differentiation while promoting glia cell formation. Analysis of pathway-specific genes revealed that Dex induces an upregulation of the Wnt-signaling antagonist DKK1. Moreover, Dex- or DKK1-treated hNPCs showed reduced transcriptional levels of the two canonical Wnt target genes cyclin D1 and inhibitor of DNA binding 2 (ID2). Chromatin immunoprecipitation showed that Dex, via the glucocorticoid receptor, interacts with the DKK1 promotor. Treatment of hNPC with recombinant DKK1 or neutralizing antibodies indicated that DKK1 has a critical role in the Dex-induced inhibition of proliferation and neuronal differentiation with a concomitant increase in glial cells. Taken together, our findings show that GC reduce proliferation and interfere with differentiation of hNPCs via the canonical Wnt-signaling pathway.
Collapse
Affiliation(s)
- Michaela Moors
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
35
|
Increased (6 exon) interleukin-7 production after M. tuberculosis infection and soluble interleukin-7 receptor expression in lung tissue. Genes Immun 2011; 12:513-22. [PMID: 21677672 DOI: 10.1038/gene.2011.29] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Interleukin-7 (IL-7) and the IL-7 receptor (IL-7R) have been shown to be alternatively spliced in infectious diseases. We tested IL-7 and IL-7R splicing in a tuberculosis (TB)-vaccine/Mycobacterium tuberculosis (Mtb)-challenge model in non-human primates (NHPs). Differential IL-7 splicing was detected in peripheral blood mononuclear cells (PBMCs) from 15/15 NHPs showing 6 different IL-7 spliced isoforms. This pattern did not change after infection with virulent Mtb. We demonstrated increased IL-7 (6 exon) and IL-17 protein production in lung tissue along with concomitant decreased transforming growth factor-β (TGF-β) from NHPs (vaccinated with a recombinant BCG (rBCG)) who showed increased survival after Mtb challenge. IL-7 increased IL-17 and interferon-γ (IFN-γ) gene and protein expression in PBMCs. Mtb-infected NHPs showed differential IL-7R splicing associated with the anatomical location and tissue origin, that is, in lung tissue, hilus, axillary lymph nodes (LNs) and spleen. Differential splicing of the IL-7R was typical for healthy (non-Mtb infected) and for Mtb-infected lung tissue with a dominant expression of soluble IL-7R (sIL-7R) receptor lacking exon 6 (9:1 ratio of sIL-7R/cell-bound IL-7R). Differential ratios of cell-bound vs sIL-7R could be observed in hilus and axillary LNs from Mtb-infected NHPs with an inversed ratio of 1:9 (sIL-7R/cell-bound IL-7R) in spleen and PBMCs. Soluble IL-7R is exclusively present in lung tissue.
Collapse
|
36
|
Kim GY, Hong C, Park JH. Seeing is believing: illuminating the source of in vivo interleukin-7. Immune Netw 2011; 11:1-10. [PMID: 21494371 PMCID: PMC3072672 DOI: 10.4110/in.2011.11.1.1] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 02/10/2011] [Accepted: 02/11/2011] [Indexed: 12/31/2022] Open
Abstract
Interleukin-7 (IL-7) is an essential cytokine for T cells. However, IL-7 is not produced by T cells themselves such that T cells are dependent on extrinsic IL-7. In fact, in the absence of IL-7, T cell development in the thymus as well as survival of naive T cells in the periphery is severely impaired. Furthermore, modulating IL-7 availability in vivo either by genetic means or other experimental approaches determines the size, composition and function of the T cell pool. Consequently, understanding IL-7 expression is critical for understanding T cell immunity. Until most recently, however, the spatiotemporal expression of in vivo IL-7 has remained obscured. Shortage of such information was partly due to scarce expression of IL-7 itself but mainly due to the lack of adequate reagents to monitor IL-7 expression in vivo. This situation dramatically changed with a recent rush of four independent studies that describe the generation and characterization of IL-7 reporter mice, all utilizing bacterial artificial chromosome transgene technology. The emerging consensus of these studies confirmed thymic stromal cells as the major producers of IL-7 but also identified IL-7 reporter activities in various peripheral tissues including skin, intestine and lymph nodes. Strikingly, developmental and environmental cues actively modulated IL-7 reporter activities in vivo suggesting that IL-7 regulation might be a new mechanism of shaping T cell development and homeostasis. Collectively, the availability of these new tools opens up new venues to assess unanswered questions in IL-7 biology in T cells and beyond.
Collapse
Affiliation(s)
- Grace Yoonhee Kim
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | |
Collapse
|
37
|
Ponchel F, Cuthbert RJ, Goëb V. IL-7 and lymphopenia. Clin Chim Acta 2010; 412:7-16. [PMID: 20850425 DOI: 10.1016/j.cca.2010.09.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 09/01/2010] [Accepted: 09/01/2010] [Indexed: 01/10/2023]
Abstract
Interleukin-7 (IL-7) is a growth and anti-apoptotic factor for T-lymphocytes, with potential for clinical use in the treatment of immunodeficiencies due to loss of T-cells. Lymphopenia induced by disease (HIV infection, hemodialysis or Idiopathic CD4+ lymphopenia) or by treatment (high dose chemotherapy or depleting antibodies) for cancer or auto-immune diseases results in increased circulating levels of IL-7 which decline with T-cell recovery, however, the mechanism of such response remains to be elucidated. Furthermore, IL-7 is a major player in the regulation of peripheral T-cell homeostasis and as such is an important candidate cytokine for therapy aimed at improving T-cell reconstitution following lymphopenia. Anti- IL-7 is on the other hand proposed to treat conditions where IL-7 may play a more direct role in pathogenesis such as autoimmune disease like Rheumatoid Arthritis, Multiple Sclerosis or Inflammatory Bowel disease.
Collapse
Affiliation(s)
- Frederique Ponchel
- Leeds Institute of Molecular Medicine, Section of Musculoskeletal disease, the University of Leeds, Leeds, UK.
| | | | | |
Collapse
|
38
|
Schreiber T, Gassmann K, Götz C, Hübenthal U, Moors M, Krause G, Merk HF, Nguyen NH, Scanlan TS, Abel J, Rose CR, Fritsche E. Polybrominated diphenyl ethers induce developmental neurotoxicity in a human in vitro model: evidence for endocrine disruption. ENVIRONMENTAL HEALTH PERSPECTIVES 2010; 118:572-8. [PMID: 20368126 PMCID: PMC2854737 DOI: 10.1289/ehp.0901435] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Accepted: 12/07/2009] [Indexed: 05/18/2023]
Abstract
BACKGROUND Polybrominated diphenyl ethers (PBDEs) are persistent and bioaccumulative flame retardants, which are found in rising concentrations in human tissues. They are of concern for human health because animal studies have shown that they possess the potential to be developmentally neurotoxic. OBJECTIVE Because there is little knowledge of the effects of PBDEs on human brain cells, we investigated their toxic potential for human neural development in vitro. Moreover, we studied the involvement of thyroid hormone (TH) disruption in the effects caused by PBDEs. METHODS We used the two PBDE congeners BDE-47 and BDE-99 (0.1-10 microM), which are most prominent in human tissues. As a model of neural development, we employed primary fetal human neural progenitor cells (hNPCs), which are cultured as neurospheres and mimic basic processes of brain development in vitro: proliferation, migration, and differentiation. RESULTS PBDEs do not disturb hNPC proliferation but decrease migration distance of hNPCs. Moreover, they cause a reduction of differentiation into neurons and oligodendrocytes. Simultaneous exposure with the TH receptor (THR) agonist triiodothyronine rescues these effects on migration and differentiation, whereas the THR antagonist NH-3 does not exert an additive effect. CONCLUSION PBDEs disturb development of hNPCs in vitro via endocrine disruption of cellular TH signaling at concentrations that might be of relevance for human exposure.
Collapse
Affiliation(s)
- Timm Schreiber
- Group of Toxicology, Institut für umweltmedizinische Forschung gGmbH an der Heinrich Heine-Universität, Düsseldorf, Germany
| | - Kathrin Gassmann
- Group of Toxicology, Institut für umweltmedizinische Forschung gGmbH an der Heinrich Heine-Universität, Düsseldorf, Germany
| | - Christine Götz
- Group of Toxicology, Institut für umweltmedizinische Forschung gGmbH an der Heinrich Heine-Universität, Düsseldorf, Germany
| | - Ulrike Hübenthal
- Group of Toxicology, Institut für umweltmedizinische Forschung gGmbH an der Heinrich Heine-Universität, Düsseldorf, Germany
| | - Michaela Moors
- Group of Toxicology, Institut für umweltmedizinische Forschung gGmbH an der Heinrich Heine-Universität, Düsseldorf, Germany
| | - Guido Krause
- Institute for Neurobiology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hans F. Merk
- Department of Dermatology and Allergology, University clinic, RWTH Aachen University, Aachen, Germany
| | - Ngoc-Ha Nguyen
- Departments of Pharmaceutical Chemistry and Cellular and Molecular Pharmacology, University of California–San Francisco, San Francisco, California, USA
| | - Thomas S. Scanlan
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon, USA
| | - Josef Abel
- Group of Toxicology, Institut für umweltmedizinische Forschung gGmbH an der Heinrich Heine-Universität, Düsseldorf, Germany
| | - Christine R. Rose
- Institute for Neurobiology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ellen Fritsche
- Group of Toxicology, Institut für umweltmedizinische Forschung gGmbH an der Heinrich Heine-Universität, Düsseldorf, Germany
- Department of Dermatology and Allergology, University clinic, RWTH Aachen University, Aachen, Germany
- Address correspondence to E. Fritsche, Institut für umweltmedizinische Forschung gGmbH an der Heinrich Heine-Universität, Toxicology, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany. Telephone: 00492113389217. Fax: 00492113190910. E-mail:
| |
Collapse
|