1
|
Singh S, Sutkus L, Li Z, Baker S, Bear J, Dilger RN, Miller DJ. Standardization of a silver stain to reveal mesoscale myelin in histological preparations of the mammalian brain. J Neurosci Methods 2024; 407:110139. [PMID: 38626852 DOI: 10.1016/j.jneumeth.2024.110139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/26/2024] [Accepted: 04/12/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND The brain is built of neurons supported by myelin, a fatty substance that improves cellular communication. Noninvasive magnetic resonance imaging (MRI) is now able to measure brain structure like myelin and requires histological validation. NEW METHOD Here we present work in small and large biomedical model mammals to standardize a silver impregnation method as a high-throughput histological myelin visualization procedure. Specifically, we built a new staining well plate to increase batch size, and then systematically varied the staining and clearing cycles to describe the staining response curve across taxa and conditions. We compared tissues fixed by immersion or perfusion, mounted versus free-floating, and cut as thicker or thinner slices, with two-weeks of post-fixation. RESULTS The staining response curves show optimal staining with a single exposure across taxa when incubation and clearing epochs are held to within 3-9 min. We show that clearing was slower in mounted vs free-floating tissue, and that staining was faster and caused fracturing earlier in thinner sliced and smaller volumes of tissue. COMPARISON WITH EXISTING METHODS We developed a batch processing approach to increase throughput while ensuring reproducibility and demonstrate the optimal conditions for fine myelinated fiber morphology visualization with short cycles (<9 minutes). CONCLUSIONS We present our optimized protocol to reveal mesoscale neuroanatomical myelin content in histology across mammals. This standard staining procedure will facilitate multiscale analyses of myelin content across development as well as in the presence of injury or disease.
Collapse
Affiliation(s)
- S Singh
- Department of Evolution, Ecology, and Behavior, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - L Sutkus
- Neuroscience Program, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - Z Li
- Neuroscience Program, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - S Baker
- Machine Shop, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - J Bear
- Machine Shop, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - R N Dilger
- Department of Animal Sciences, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America; Neuroscience Program, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America; Beckman Institute for Advanced Science and Technology, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - D J Miller
- Department of Evolution, Ecology, and Behavior, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America; Neuroscience Program, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America; Beckman Institute for Advanced Science and Technology, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America.
| |
Collapse
|
2
|
Schilliger Z, Alemán-Gómez Y, Magnus Smith M, Celen Z, Meuleman B, Binz PA, Steullet P, Do KQ, Conus P, Merglen A, Piguet C, Dwir D, Klauser P. Sex-specific interactions between stress axis and redox balance are associated with internalizing symptoms and brain white matter microstructure in adolescents. Transl Psychiatry 2024; 14:30. [PMID: 38233401 PMCID: PMC10794182 DOI: 10.1038/s41398-023-02728-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/19/2024] Open
Abstract
Adolescence is marked by the maturation of systems involved in emotional regulation and by an increased risk for internalizing disorders (anxiety/depression), especially in females. Hypothalamic-pituitary-adrenal (HPA)-axis function and redox homeostasis (balance between reactive oxygen species and antioxidants) have both been associated with internalizing disorders and may represent critical factors for the development of brain networks of emotional regulation. However, sex-specific interactions between these factors and internalizing symptoms and their link with brain maturation remain unexplored. We investigated in a cohort of adolescents aged 13-15 from the general population (n = 69) whether sex-differences in internalizing symptoms were associated with the glutathione (GSH)-redox cycle homeostasis and HPA-axis function and if these parameters were associated with brain white matter microstructure development. Female adolescents displayed higher levels of internalizing symptoms, GSH-peroxidase (GPx) activity and cortisol/11-deoxycortisol ratio than males. There was a strong correlation between GPx and GSH-reductase (Gred) activities in females only. The cortisol/11-deoxycortisol ratio, related to the HPA-axis activity, was associated with internalizing symptoms in both sexes, whereas GPx activity was associated with internalizing symptoms in females specifically. The cortisol/11-deoxycortisol ratio mediated sex-differences in internalizing symptoms and the association between anxiety and GPx activity in females specifically. In females, GPx activity was positively associated with generalized fractional anisotropy in widespread white matter brain regions. We found that higher levels of internalizing symptoms in female adolescents than in males relate to sex-differences in HPA-axis function. In females, our results suggest an important interplay between HPA-axis function and GSH-homeostasis, a parameter strongly associated with brain white matter microstructure.
Collapse
Affiliation(s)
- Zoé Schilliger
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Yasser Alemán-Gómez
- Connectomics Lab, Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Mariana Magnus Smith
- Division of General Pediatrics, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Zeynep Celen
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ben Meuleman
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pierre-Alain Binz
- Service of Clinical Chemistry, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| | - Pascal Steullet
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Philippe Conus
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Arnaud Merglen
- Division of General Pediatrics, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Camille Piguet
- Division of General Pediatrics, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Daniella Dwir
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Paul Klauser
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
- Service of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
3
|
Romaniuk L, MacSweeney N, Atkinson K, Chan SWY, Barbu MC, Lawrie SM, Whalley HC. Striatal correlates of Bayesian beliefs in self-efficacy in adolescents and their relation to mood and autonomy: a pilot study. Cereb Cortex Commun 2023; 4:tgad020. [PMID: 38089939 PMCID: PMC10712445 DOI: 10.1093/texcom/tgad020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 10/25/2023] [Indexed: 02/02/2024] Open
Abstract
Major depressive disorder often originates in adolescence and is associated with long-term functional impairment. Mechanistically characterizing this heterogeneous illness could provide important leads for optimizing treatment. Importantly, reward learning is known to be disrupted in depression. In this pilot fMRI study of 21 adolescents (16-20 years), we assessed how reward network disruption impacts specifically on Bayesian belief representations of self-efficacy (SE-B) and their associated uncertainty (SE-U), using a modified instrumental learning task probing activation induced by the opportunity to choose, and an optimal Hierarchical Gaussian Filter computational model. SE-U engaged caudate, nucleus accumbens (NAcc), precuneus, posterior parietal and dorsolateral prefrontal cortex (PFWE < 0.005). Sparse partial least squares analysis identified SE-U striatal activation as associating with one's sense of perceived choice and depressive symptoms, particularly anhedonia and negative feelings about oneself. As Bayesian uncertainty modulates belief flexibility and their capacity to steer future actions, this suggests that these striatal signals may be informative developmentally, longitudinally and in assessing response to treatment.
Collapse
Affiliation(s)
- Liana Romaniuk
- Division of Psychiatry, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Morningside Park, Edinburgh EH10 5H, United Kingdom
| | - Niamh MacSweeney
- Division of Psychiatry, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Morningside Park, Edinburgh EH10 5H, United Kingdom
| | - Kimberley Atkinson
- Division of Psychiatry, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Morningside Park, Edinburgh EH10 5H, United Kingdom
| | - Stella W Y Chan
- School of Psychology & Clinical Language Sciences, University of Reading, Earley Gate, Whiteknights, Reading RG6 6ES, United Kingdom
| | - Miruna C Barbu
- Division of Psychiatry, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Morningside Park, Edinburgh EH10 5H, United Kingdom
| | - Stephen M Lawrie
- Division of Psychiatry, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Morningside Park, Edinburgh EH10 5H, United Kingdom
| | - Heather C Whalley
- Division of Psychiatry, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Morningside Park, Edinburgh EH10 5H, United Kingdom
| |
Collapse
|
4
|
Duan J, Gong X, Womer FY, Sun K, Tang L, Liu J, Zheng J, Zhu Y, Tang Y, Zhang X, Wang F. Neurodevelopmental trajectories, polygenic risk, and lipometabolism in vulnerability and resilience to schizophrenia. BMC Psychiatry 2023; 23:153. [PMID: 36894907 PMCID: PMC9999573 DOI: 10.1186/s12888-023-04597-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/07/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Schizophrenia (SZ) arises from a complex interplay involving genetic and molecular factors. Early intervention of SZ hinges upon understanding its vulnerability and resiliency factors in study of SZ and genetic high risk for SZ (GHR). METHODS Herein, using integrative and multimodal strategies, we first performed a longitudinal study of neural function as measured by amplitude of low frequency function (ALFF) in 21 SZ, 26 GHR, and 39 healthy controls to characterize neurodevelopmental trajectories of SZ and GHR. Then, we examined the relationship between polygenic risk score for SZ (SZ-PRS), lipid metabolism, and ALFF in 78 SZ, and 75 GHR in cross-sectional design to understand its genetic and molecular substrates. RESULTS Across time, SZ and GHR diverge in ALFF alterations of the left medial orbital frontal cortex (MOF). At baseline, both SZ and GHR had increased left MOF ALFF compared to HC (P < 0.05). At follow-up, increased ALFF persisted in SZ, yet normalized in GHR. Further, membrane genes and lipid species for cell membranes predicted left MOF ALFF in SZ; whereas in GHR, fatty acids best predicted and were negatively correlated (r = -0.302, P < 0.05) with left MOF. CONCLUSIONS Our findings implicate divergence in ALFF alteration in left MOF between SZ and GHR with disease progression, reflecting vulnerability and resiliency to SZ. They also indicate different influences of membrane genes and lipid metabolism on left MOF ALFF in SZ and GHR, which have important implications for understanding mechanisms underlying vulnerability and resiliency in SZ and contribute to translational efforts for early intervention.
Collapse
Affiliation(s)
- Jia Duan
- Department of Psychiatry. Early Intervention Unit, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210000, Jiangsu, PR China.,Department of Psychiatry and Gerontology, The First Affiliated Hospital, China Medical University, 155 Nanjing North Street, Shenyang, 110001, Liaoning, PR China
| | - Xiaohong Gong
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China
| | - Fay Y Womer
- Dept of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kaijin Sun
- Department of Psychiatry. Early Intervention Unit, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210000, Jiangsu, PR China
| | - Lili Tang
- Department of Psychiatry. Early Intervention Unit, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210000, Jiangsu, PR China.,Department of Psychiatry and Gerontology, The First Affiliated Hospital, China Medical University, 155 Nanjing North Street, Shenyang, 110001, Liaoning, PR China
| | - Juan Liu
- Department of Psychiatry. Early Intervention Unit, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210000, Jiangsu, PR China.,Department of Psychiatry and Gerontology, The First Affiliated Hospital, China Medical University, 155 Nanjing North Street, Shenyang, 110001, Liaoning, PR China
| | - Junjie Zheng
- Department of Psychiatry. Early Intervention Unit, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210000, Jiangsu, PR China
| | - Yue Zhu
- Department of Psychiatry. Early Intervention Unit, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210000, Jiangsu, PR China.,Department of Psychiatry and Gerontology, The First Affiliated Hospital, China Medical University, 155 Nanjing North Street, Shenyang, 110001, Liaoning, PR China
| | - Yanqing Tang
- Department of Psychiatry and Gerontology, The First Affiliated Hospital, China Medical University, 155 Nanjing North Street, Shenyang, 110001, Liaoning, PR China.
| | - Xizhe Zhang
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 210000, Jiangsu, PR China.
| | - Fei Wang
- Department of Psychiatry. Early Intervention Unit, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210000, Jiangsu, PR China. .,Department of Psychiatry and Gerontology, The First Affiliated Hospital, China Medical University, 155 Nanjing North Street, Shenyang, 110001, Liaoning, PR China.
| |
Collapse
|
5
|
Maklad A, Sedeeq M, Wilson R, Heath JA, Gueven N, Azimi I. LIN28 expression and function in medulloblastoma. J Cell Physiol 2023; 238:533-548. [PMID: 36649308 DOI: 10.1002/jcp.30946] [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: 06/08/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2023]
Abstract
Medulloblastoma (MB) is the most common malignant pediatric brain tumor. Current treatment modalities are not completely effective and can lead to severe neurological and cognitive adverse effects. In addition to urgently needing better treatment approaches, new diagnostic and prognostic biomarkers are required to improve the therapy outcomes of MB patients. The RNA-binding proteins, LIN28A and LIN28B, are known to regulate invasive phenotypes in many different cancer types. However, the expression and function of these proteins in MB had not been studied to date. This study identified the expression of LIN28A and LIN28B in MB patient samples and cell lines and assessed the effect of LIN28 inhibition on MB cell growth, metabolism and stemness. LIN28B expression was significantly upregulated in MB tissues compared to normal brain tissues. This upregulation, which was not observed in other brain tumors, was specific for the aggressive MB subgroups and correlated with patient survival and metastasis rates. Functionally, pharmacological inhibition of LIN28 activity concentration-dependently reduced LIN28B expression, as well as the growth of D283 MB cells. While LIN28 inhibition did not affect the levels of intracellular ATP, it reduced the expression of the stemness marker CD133 in D283 cells and the sphere formation of CHLA-01R cells. LIN28B, which is highly expressed in the human cerebellum during the first few months after birth, subsequently decreased with age. The results of this study highlight the potential of LIN28B as a diagnostic and prognostic marker for MB and open the possibility to utilize LIN28 as a pharmacological target to suppress MB cell growth and stemness.
Collapse
Affiliation(s)
- Ahmed Maklad
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Mohammed Sedeeq
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Richard Wilson
- Central Science Laboratory, College of Science and Engineering, University of Tasmania, Hobart, Tasmania, Australia
| | - John A Heath
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
- Department of Paediatrics, Royal Hobart Hospital, Hobart, Australia
| | - Nuri Gueven
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Iman Azimi
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
6
|
Qi D, Geng Y, Cardenas J, Gu J, Yi SS, Huang JH, Fonkem E, Wu E. Transcriptomic analyses of patient peripheral blood with hemoglobin depletion reveal glioblastoma biomarkers. NPJ Genom Med 2023; 8:2. [PMID: 36697401 PMCID: PMC9877004 DOI: 10.1038/s41525-022-00348-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 12/21/2022] [Indexed: 01/26/2023] Open
Abstract
Peripheral blood is gaining prominence as a noninvasive alternative to tissue biopsy to develop biomarkers for glioblastoma (GBM); however, widely utilized blood-based biomarkers in clinical settings have not yet been identified due to the lack of a robust detection approach. Here, we describe the application of globin reduction in RNA sequencing of whole blood (i.e., WBGR) and perform transcriptomic analysis to identify GBM-associated transcriptomic changes. By using WBGR, we improved the detection sensitivity of informatic reads and identified differential gene expression in GBM blood. By analyzing tumor tissues, we identified transcriptomic traits of GBM blood. Further functional enrichment analyses retained the most changed genes in GBM. Subsequent validation elicited a 10-gene panel covering mRNA, long noncoding RNA, and microRNA (i.e., GBM-Dx panel) that has translational potential to aid in the early detection or clinical management of GBM. Here, we report an integrated approach, WBGR, with comprehensive analytic capacity for blood-based marker identification.
Collapse
Affiliation(s)
- Dan Qi
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX, 76508, USA
| | - Yiqun Geng
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX, 76508, USA
- Laboratory of Molecular Pathology, Shantou University Medical College, 515041, Shantou, China
| | - Jacob Cardenas
- Baylor Scott & White Research Institute, Dallas, TX, 75204, USA
| | - Jinghua Gu
- Baylor Scott & White Research Institute, Dallas, TX, 75204, USA
| | - S Stephen Yi
- Institute for Cellular and Molecular Biology (ICMB), College of Natural Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
- Oden Institute for Computational Engineering and Sciences (ICES), The University of Texas at Austin, Austin, TX, 78712, USA
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
- Department of Oncology, LIVESTRONG Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Jason H Huang
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX, 76508, USA.
- Texas A & M University School of Medicine, Temple, TX, 76508, USA.
| | - Ekokobe Fonkem
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX, 76508, USA.
- Texas A & M University School of Medicine, Temple, TX, 76508, USA.
| | - Erxi Wu
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX, 76508, USA.
- Department of Oncology, LIVESTRONG Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, 78712, USA.
- Texas A & M University School of Medicine, Temple, TX, 76508, USA.
- Texas A & M University School of Pharmacy, College Station, TX, 77843, USA.
| |
Collapse
|
7
|
Petanjek Z, Banovac I, Sedmak D, Hladnik A. Dendritic Spines: Synaptogenesis and Synaptic Pruning for the Developmental Organization of Brain Circuits. ADVANCES IN NEUROBIOLOGY 2023; 34:143-221. [PMID: 37962796 DOI: 10.1007/978-3-031-36159-3_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Synaptic overproduction and elimination is a regular developmental event in the mammalian brain. In the cerebral cortex, synaptic overproduction is almost exclusively correlated with glutamatergic synapses located on dendritic spines. Therefore, analysis of changes in spine density on different parts of the dendritic tree in identified classes of principal neurons could provide insight into developmental reorganization of specific microcircuits.The activity-dependent stabilization and selective elimination of the initially overproduced synapses is a major mechanism for generating diversity of neural connections beyond their genetic determination. The largest number of overproduced synapses was found in the monkey and human cerebral cortex. The highest (exceeding adult values by two- to threefold) and most protracted overproduction (up to third decade of life) was described for associative layer IIIC pyramidal neurons in the human dorsolateral prefrontal cortex.Therefore, the highest proportion and extraordinarily extended phase of synaptic spine overproduction is a hallmark of neural circuitry in human higher-order associative areas. This indicates that microcircuits processing the most complex human cognitive functions have the highest level of developmental plasticity. This finding is the backbone for understanding the effect of environmental impact on the development of the most complex, human-specific cognitive and emotional capacities, and on the late onset of human-specific neuropsychiatric disorders, such as autism and schizophrenia.
Collapse
Affiliation(s)
- Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia.
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia.
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia.
| | - Ivan Banovac
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dora Sedmak
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ana Hladnik
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
8
|
Perica MI, Calabro FJ, Larsen B, Foran W, Yushmanov VE, Hetherington H, Tervo-Clemmens B, Moon CH, Luna B. Development of frontal GABA and glutamate supports excitation/inhibition balance from adolescence into adulthood. Prog Neurobiol 2022; 219:102370. [DOI: 10.1016/j.pneurobio.2022.102370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/22/2022] [Accepted: 10/24/2022] [Indexed: 11/05/2022]
|
9
|
Zhang L, Nesvick CL, Day CA, Choi J, Lu VM, Peterson T, Power EA, Anderson JB, Hamdan FH, Decker PA, Simons R, Welby JP, Siada R, Ge J, Kaptzan T, Johnsen SA, Hinchcliffe EH, Daniels DJ. STAT3 is a biologically relevant therapeutic target in H3K27M-mutant diffuse midline glioma. Neuro Oncol 2022; 24:1700-1711. [PMID: 35397475 PMCID: PMC9527528 DOI: 10.1093/neuonc/noac093] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background H3K27M-mutant diffuse midline glioma (DMG) is a lethal brain tumor that usually occurs in children. Despite advances in our understanding of its underlying biology, efficacious therapies are severely lacking. Methods We screened a library of drugs either FDA-approved or in clinical trial using a library of patient-derived H3K27M-mutant DMG cell lines with cell viability as the outcome. Results were validated for clinical relevance and mechanistic importance using patient specimens from biopsy and autopsy, patient-derived cell lines, inhibition by gene knockdown and small molecule inhibitors, and patient-derived xenografts. Results Kinase inhibitors were highly toxic to H3K27M-mutant DMG cells. Within this class, STAT3 inhibitors demonstrated robust cytotoxic activity in vitro. Mechanistic analyses revealed one form of activated STAT3, phospho-tyrosine- 705 STAT3 (pSTAT3), was selectively upregulated in H3K27M-mutant cell lines and clinical specimens. STAT3 inhibition by CRISPR/Cas9 knockout, shRNA or small molecule inhibition reduced cell viability in vitro, and partially restored expression of the polycomb repressive mark H3K27me3, which is classically lost in H3K27M-mutant DMG. Putative STAT3-regulated genes were enriched in an H3K27M-knockout DMG cell line, indicating relative gain of STAT3 signaling in K27M-mutant cells. Treatment of patient-derived intracranial xenografts with WP1066, a STAT3 pathway inhibitor currently in clinical use for pediatric brain tumors, resulted in stasis of tumor growth, and increased overall survival. Finally, pSTAT3(Y705) was detected in circulating plasma extracellular vesicles of patients with H3K27M-mutant DMG. Conclusions STAT3 is a biologically relevant therapeutic target in H3K27M-mutant DMG. STAT3 inhibition should be considered in future clinical trials.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Cody L Nesvick
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Charlie A Day
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Jonghoon Choi
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Victor M Lu
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Timothy Peterson
- Department of Cardiac Regeneration Program, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Jacob B Anderson
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Feda H Hamdan
- Department of Gastroenterology, Mayo Clinic, Rochester, Minnesota, USA
| | - Paul A Decker
- Department of Biostatistics, Mayo Clinic, Rochester, Minnesota, USA
| | - Renae Simons
- Campbell University Jerry M. Wallace School of Osteopathic Medicine
| | - John P Welby
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Ruby Siada
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jizhi Ge
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Tatiana Kaptzan
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Steven A Johnsen
- Department of Gastroenterology, Mayo Clinic, Rochester, Minnesota, USA.,Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | | | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA.,Molecular Pharmacology and Experimental Therapeutics Program
| |
Collapse
|
10
|
Thomas KT, Zakharenko SS. MicroRNAs in the Onset of Schizophrenia. Cells 2021; 10:2679. [PMID: 34685659 PMCID: PMC8534348 DOI: 10.3390/cells10102679] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/30/2021] [Accepted: 10/02/2021] [Indexed: 12/14/2022] Open
Abstract
Mounting evidence implicates microRNAs (miRNAs) in the pathology of schizophrenia. These small noncoding RNAs bind to mRNAs containing complementary sequences and promote their degradation and/or inhibit protein synthesis. A single miRNA may have hundreds of targets, and miRNA targets are overrepresented among schizophrenia-risk genes. Although schizophrenia is a neurodevelopmental disorder, symptoms usually do not appear until adolescence, and most patients do not receive a schizophrenia diagnosis until late adolescence or early adulthood. However, few studies have examined miRNAs during this critical period. First, we examine evidence that the miRNA pathway is dynamic throughout adolescence and adulthood and that miRNAs regulate processes critical to late neurodevelopment that are aberrant in patients with schizophrenia. Next, we examine evidence implicating miRNAs in the conversion to psychosis, including a schizophrenia-associated single nucleotide polymorphism in MIR137HG that is among the strongest known predictors of age of onset in patients with schizophrenia. Finally, we examine how hemizygosity for DGCR8, which encodes an obligate component of the complex that synthesizes miRNA precursors, may contribute to the onset of psychosis in patients with 22q11.2 microdeletions and how animal models of this disorder can help us understand the many roles of miRNAs in the onset of schizophrenia.
Collapse
Affiliation(s)
- Kristen T. Thomas
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Stanislav S. Zakharenko
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
11
|
Ventriglio A, Bellomo A, Favale D, Bonfitto I, Vitrani G, Di Sabatino D, Cuozzo E, Di Gioia I, Mauro P, Giampaolo P, Alessandro V, De Berardis D. Oxidative Stress in the Early Stage of Psychosis. Curr Top Med Chem 2021; 21:1457-1470. [PMID: 34218786 DOI: 10.2174/1568026621666210701105839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/25/2021] [Accepted: 05/05/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND In the past few decades, increasing evidence in the literature has appeared describing the role of the antioxidant defense system and redox signaling in the multifactorial pathophysiology of psychosis. It is of interest to clinicians and researchers alike that abnormalities of the antioxidant defense system are associated with alterations of cellular membranes, immune functions and neurotransmission, all of which have some clinical implications. METHODS This narrative review summarizes the evidence regarding oxidative stress in the early stages of psychosis. We included 136 peer-reviewed articles published from 2007 to 2020 on PubMed EMBASE, The Cochrane Library and Google Scholar. RESULTS Patients affected by psychotic disorders show a decreased level of non-enzymatic antioxidants, an increased level of lipid peroxides, nitric oxides, and a homeostatic imbalance of purine catabolism. In particular, a significantly reduced antioxidant defense has been described in the early onset first episode of psychosis, including reduced levels of glutathione. Also, it has been shown that a decreased basal low -antioxidant capacity correlates with cognitive deficits and negative symptoms, mostly related to glutamate-receptor hypofunction. In addition, atypical antipsychotic drugs seem to show significant antioxidant activity. These factors are critical in order to treat cases of first-onset psychosis effectively. CONCLUSION This systematic review indicates the importance that must be given to anti-oxidant defense systems.
Collapse
Affiliation(s)
- Antonio Ventriglio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonello Bellomo
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Donato Favale
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Iris Bonfitto
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Giovanna Vitrani
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Dario Di Sabatino
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Edwige Cuozzo
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Ilaria Di Gioia
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Pettorruso Mauro
- Department of Neurosciences, Imaging and Clinical Sciences, Univerity of Chieti, Italy
| | - Perna Giampaolo
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | | | |
Collapse
|
12
|
Lloyd K, Reyes T. High fat diet consumption restricted to adolescence has minimal effects on adult executive function that vary by sex. Nutr Neurosci 2020; 25:801-811. [PMID: 32840166 DOI: 10.1080/1028415x.2020.1809879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Early life environment can have a lasting effect on brain development and behavior. Diet is a potent environmental factor that can positively or negatively affect neurodevelopment, and unfortunately, the likelihood of a poor diet is high during adolescence. Adverse effects of adolescent high fat diet have been observed on reward-related behaviors, reversal learning, and hippocampal-dependent learning tasks in rodents when tested in adulthood. The prefrontal cortex (PFC) continues to develop throughout adolescence and is thus vulnerable to environmental insults such as poor diet. Therefore, we sought to examine the effects of a high fat diet (HFD) consumed only during adolescence on later life adult PFC-dependent executive function. Male and female mice were fed a HFD (60% energy from fat) during either early or late adolescence then switched to standard chow and tested in a battery of touchscreen-based operant tests of executive function in adulthood. Contrary to our prediction of an adverse effect of HFD, there was no effect of adolescent HFD in males, and females showed faster learning and decreased inattention in adulthood. We conclude that the effects of adolescent-limited HFD on adult executive function are mild, positive, and vary by sex.
Collapse
Affiliation(s)
- Kelsey Lloyd
- Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Teresa Reyes
- Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
13
|
Jung E, Alfonso J, Monyer H, Wick W, Winkler F. Neuronal signatures in cancer. Int J Cancer 2020; 147:3281-3291. [PMID: 32510582 DOI: 10.1002/ijc.33138] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/26/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022]
Abstract
Despite advances in the treatment of solid tumors, the prognosis of patients with many cancers remains poor, particularly of those with primary and metastatic brain tumors. In the last years, "Cancer Neuroscience" emerged as novel field of research at the crossroads of oncology and classical neuroscience. In primary brain tumors, including glioblastoma (GB), communicating networks that render tumor cells resistant against cytotoxic therapies were identified. To build these networks, GB cells extend neurite-like protrusions called tumor microtubes (TMs). Synapses on TMs allow tumor cells to retrieve neuronal input that fosters growth. Single cell sequencing further revealed that primary brain tumors recapitulate many steps of neurodevelopment. Interestingly, neuronal characteristics, including the ability to extend neurite-like protrusions, neuronal gene expression signatures and interactions with neurons, have now been found not only in brain and neuroendocrine tumors but also in some cancers of epithelial origin. In this review, we will provide an overview about neurite-like protrusions as well as neurodevelopmental origins, hierarchies and gene expression signatures in cancer. We will also discuss how "Cancer Neuroscience" might provide a framework for the development of novel therapies.
Collapse
Affiliation(s)
- Erik Jung
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julieta Alfonso
- Department of Clinical Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hannah Monyer
- Department of Clinical Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Clinical Neurobiology, Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Wolfgang Wick
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
14
|
Jane EP, Premkumar DR, Thambireddy S, Golbourn B, Agnihotri S, Bertrand KC, Mack SC, Myers MI, Chattopadhyay A, Taylor DL, Schurdak ME, Stern AM, Pollack IF. Targeting NAD + Biosynthesis Overcomes Panobinostat and Bortezomib-Induced Malignant Glioma Resistance. Mol Cancer Res 2020; 18:1004-1017. [PMID: 32238439 DOI: 10.1158/1541-7786.mcr-19-0669] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 12/17/2019] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
To improve therapeutic responses in patients with glioma, new combination therapies that exploit a mechanistic understanding of the inevitable emergence of drug resistance are needed. Intratumoral heterogeneity enables a low barrier to resistance in individual patients with glioma. We reasoned that targeting two or more fundamental processes that gliomas are particularly dependent upon could result in pleiotropic effects that would reduce the diversity of resistant subpopulations allowing convergence to a more robust therapeutic strategy. In contrast to the cytostatic responses observed with each drug alone, the combination of the histone deacetylase inhibitor panobinostat and the proteasome inhibitor bortezomib synergistically induced apoptosis of adult and pediatric glioma cell lines at clinically achievable doses. Resistance that developed was examined using RNA-sequencing and pharmacologic screening of resistant versus drug-naïve cells. Quinolinic acid phosphoribosyltransferase (QPRT), the rate-determining enzyme for de novo synthesis of NAD+ from tryptophan, exhibited particularly high differential gene expression in resistant U87 cells and protein expression in all resistant lines tested. Reducing QPRT expression reversed resistance, suggesting that QPRT is a selective and targetable dependency for the panobinostat-bortezomib resistance phenotype. Pharmacologic inhibition of either NAD+ biosynthesis or processes such as DNA repair that consume NAD+ or their simultaneous inhibition with drug combinations, specifically enhanced apoptosis in treatment-resistant cells. Concomitantly, de novo vulnerabilities to known drugs were observed. IMPLICATIONS: These data provide new insights into mechanisms of treatment resistance in gliomas, hold promise for targeting recurrent disease, and provide a potential strategy for further exploration of next-generation inhibitors.
Collapse
Affiliation(s)
- Esther P Jane
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Daniel R Premkumar
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania. .,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute Brain Tumor Center, Pittsburgh, Pennsylvania
| | - Swetha Thambireddy
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brian Golbourn
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sameer Agnihotri
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute Brain Tumor Center, Pittsburgh, Pennsylvania
| | - Kelsey C Bertrand
- Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Stephen C Mack
- Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Max I Myers
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ansuman Chattopadhyay
- Molecular Biology Information Service, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - D Lansing Taylor
- Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania.,Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Mark E Schurdak
- Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania.,Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Andrew M Stern
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ian F Pollack
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania. .,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute Brain Tumor Center, Pittsburgh, Pennsylvania
| |
Collapse
|
15
|
Meel MH, de Gooijer MC, Metselaar DS, Sewing ACP, Zwaan K, Waranecki P, Breur M, Buil LCM, Lagerweij T, Wedekind LE, Twisk JWR, Koster J, Hashizume R, Raabe EH, Montero Carcaboso Á, Bugiani M, Phoenix TN, van Tellingen O, van Vuurden DG, Kaspers GJL, Hulleman E. Combined Therapy of AXL and HDAC Inhibition Reverses Mesenchymal Transition in Diffuse Intrinsic Pontine Glioma. Clin Cancer Res 2020; 26:3319-3332. [PMID: 32165429 DOI: 10.1158/1078-0432.ccr-19-3538] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/04/2020] [Accepted: 03/06/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Diffuse intrinsic pontine glioma (DIPG) is an incurable type of pediatric brain cancer, which in the majority of cases is driven by mutations in genes encoding histone 3 (H3K27M). We here determined the preclinical therapeutic potential of combined AXL and HDAC inhibition in these tumors to reverse their mesenchymal, therapy-resistant, phenotype. EXPERIMENTAL DESIGN We used public databases and patient-derived DIPG cells to identify putative drivers of the mesenchymal transition in these tumors. Patient-derived neurospheres, xenografts, and allografts were used to determine the therapeutic potential of combined AXL/HDAC inhibition for the treatment of DIPG. RESULTS We identified AXL as a therapeutic target and regulator of the mesenchymal transition in DIPG. Combined AXL and HDAC inhibition had a synergistic and selective antitumor effect on H3K27M DIPG cells. Treatment of DIPG cells with the AXL inhibitor BGB324 and the HDAC inhibitor panobinostat resulted in a decreased expression of mesenchymal and stem cell genes. Moreover, this combination treatment decreased expression of DNA damage repair genes in DIPG cells, strongly sensitizing them to radiation. Pharmacokinetic studies showed that BGB324, like panobinostat, crosses the blood-brain barrier. Consequently, treatment of patient-derived DIPG xenograft and murine DIPG allograft-bearing mice with BGB324 and panobinostat resulted in a synergistic antitumor effect and prolonged survival. CONCLUSIONS Combined inhibition of AXL and HDACs in DIPG cells results in a synergistic antitumor effect by reversing their mesenchymal, stem cell-like, therapy-resistant phenotype. As such, this treatment combination may serve as part of a future multimodal therapeutic strategy for DIPG.
Collapse
Affiliation(s)
- Michaël H Meel
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Mark C de Gooijer
- Division of Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Dennis S Metselaar
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - A Charlotte P Sewing
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Kenn Zwaan
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Piotr Waranecki
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Marjolein Breur
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Levi C M Buil
- Division of Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Tonny Lagerweij
- Department of Neurosurgery, Neuro-oncology Research Group, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Laurine E Wedekind
- Department of Neurosurgery, Neuro-oncology Research Group, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Jos W R Twisk
- Department of Epidemiology and Biostatistics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Jan Koster
- Department of Oncogenomics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Rintaro Hashizume
- Departments of Neurological Surgery and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Eric H Raabe
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ángel Montero Carcaboso
- Preclinical Therapeutics and Drug Delivery Research Program, Department of Oncology, Hospital Sant Joan de Déu Barcelona, Spain
| | - Marianna Bugiani
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Timothy N Phoenix
- Division of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati/Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Olaf van Tellingen
- Division of Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Dannis G van Vuurden
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Gertjan J L Kaspers
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Esther Hulleman
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands. .,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
16
|
Celastrol-induced degradation of FANCD2 sensitizes pediatric high-grade gliomas to the DNA-crosslinking agent carboplatin. EBioMedicine 2019; 50:81-92. [PMID: 31735550 PMCID: PMC6921187 DOI: 10.1016/j.ebiom.2019.10.062] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/26/2019] [Accepted: 10/31/2019] [Indexed: 12/25/2022] Open
Abstract
Background Pediatric high-grade gliomas (pHGG) are the leading cause of cancer-related death during childhood. Due to their diffuse growth characteristics, chemoresistance and location behind the blood-brain barrier (BBB), the prognosis of pHGG has barely improved in the past decades. As such, there is a dire need for new therapies that circumvent those difficulties. Since aberrant expression of DNA damage-response associated Fanconi anemia proteins play a central role in the onset and therapy resistance of many cancers, we here investigated if FANCD2 depletion could sensitize pHGG to additional DNA damage. Methods We determined the capacity of celastrol, a BBB-penetrable compound that degrades FANCD2, to sensitize glioma cells to the archetypical DNA-crosslinking agent carboplatin in vitro in seven patient-derived pHGG models. In addition, we tested this drug combination in vivo in a patient-derived orthotopic pHGG xenograft model. Underlying mechanisms to drug response were investigated using mRNA expression profiling, western blotting, immunofluorescence, FANCD2 knockdown and DNA fiber assays. Findings FANCD2 is overexpressed in HGGs and depletion of FANCD2 by celastrol synergises with carboplatin to induce cytotoxicity. Combination therapy prolongs survival of pHGG-bearing mice over monotherapy and control groups in vivo (P<0.05). In addition, our results suggest that celastrol treatment stalls ongoing replication forks, causing sensitivity to DNA-crosslinking in FANCD2-dependent glioma cells. Interpretation Our results show that depletion of FANCD2 acts as a chemo-sensitizing strategy in pHGG. Combination therapy using celastrol and carboplatin might serve as a clinically relevant strategy for the treatment of pHGG. Funding This study was funded by a grant from the Children Cancer-Free Foundation (KIKA, project 210). The disclosed funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Collapse
|
17
|
Petanjek Z, Sedmak D, Džaja D, Hladnik A, Rašin MR, Jovanov-Milosevic N. The Protracted Maturation of Associative Layer IIIC Pyramidal Neurons in the Human Prefrontal Cortex During Childhood: A Major Role in Cognitive Development and Selective Alteration in Autism. Front Psychiatry 2019; 10:122. [PMID: 30923504 PMCID: PMC6426783 DOI: 10.3389/fpsyt.2019.00122] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 02/18/2019] [Indexed: 12/12/2022] Open
Abstract
The human specific cognitive shift starts around the age of 2 years with the onset of self-awareness, and continues with extraordinary increase in cognitive capacities during early childhood. Diffuse changes in functional connectivity in children aged 2-6 years indicate an increase in the capacity of cortical network. Interestingly, structural network complexity does not increase during this time and, thus, it is likely to be induced by selective maturation of a specific neuronal subclass. Here, we provide an overview of a subclass of cortico-cortical neurons, the associative layer IIIC pyramids of the human prefrontal cortex. Their local axonal collaterals are in control of the prefrontal cortico-cortical output, while their long projections modulate inter-areal processing. In this way, layer IIIC pyramids are the major integrative element of cortical processing, and changes in their connectivity patterns will affect global cortical functioning. Layer IIIC neurons have a unique pattern of dendritic maturation. In contrast to other classes of principal neurons, they undergo an additional phase of extensive dendritic growth during early childhood, and show characteristic molecular changes. Taken together, circuits associated with layer IIIC neurons have the most protracted period of developmental plasticity. This unique feature is advanced but also provides a window of opportunity for pathological events to disrupt normal formation of cognitive circuits involving layer IIIC neurons. In this manuscript, we discuss how disrupted dendritic and axonal maturation of layer IIIC neurons may lead into global cortical disconnectivity, affecting development of complex communication and social abilities. We also propose a model that developmentally dictated incorporation of layer IIIC neurons into maturing cortico-cortical circuits between 2 to 6 years will reveal a previous (perinatal) lesion affecting other classes of principal neurons. This "disclosure" of pre-existing functionally silent lesions of other neuronal classes induced by development of layer IIIC associative neurons, or their direct alteration, could be found in different forms of autism spectrum disorders. Understanding the gene-environment interaction in shaping cognitive microcircuitries may be fundamental for developing rehabilitation and prevention strategies in autism spectrum and other cognitive disorders.
Collapse
Affiliation(s)
- Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dora Sedmak
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Domagoj Džaja
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ana Hladnik
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mladen Roko Rašin
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Nataša Jovanov-Milosevic
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Medical Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
18
|
Schulmann A, Ryu E, Goncalves V, Rollins B, Christiansen M, Frye MA, Biernacka J, Vawter MP. Novel Complex Interactions between Mitochondrial and Nuclear DNA in Schizophrenia and Bipolar Disorder. MOLECULAR NEUROPSYCHIATRY 2019; 5:13-27. [PMID: 31019915 PMCID: PMC6465701 DOI: 10.1159/000495658] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 11/20/2018] [Indexed: 12/21/2022]
Abstract
Mitochondrial dysfunction has been associated with schizophrenia (SZ) and bipolar disorder (BD). This review examines recent publications and novel associations between mitochondrial genes and SZ and BD. Associations of nuclear-encoded mitochondrial variants with SZ were found using gene- and pathway-based approaches. Two control region mitochondrial DNA (mtDNA) SNPs, T16519C and T195C, both showed an association with SZ and BD. A review of 4 studies of A15218G located in the cytochrome B oxidase gene (CYTB, SZ = 11,311, control = 35,735) shows a moderate association with SZ (p = 2.15E-03). Another mtDNA allele A12308G was nominally associated with psychosis in BD type I subjects and SZ. The first published study testing the epistatic interaction between nuclear-encoded and mitochondria-encoded genes demonstrated evidence for potential interactions between mtDNA and the nuclear genome for BD. A similar analysis for the risk of SZ revealed significant joint effects (34 nuclear-mitochondria SNP pairs with joint effect p ≤ 5E-07) and significant enrichment of projection neurons. The mitochondria-encoded gene CYTB was found in both the epistatic interactions for SZ and BD and the single SNP association of SZ. Future efforts considering population stratification and polygenic risk scores will test the role of mitochondrial variants in psychiatric disorders.
Collapse
Affiliation(s)
- Anton Schulmann
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Euijung Ryu
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Vanessa Goncalves
- Molecular Brain Science Department, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Brandi Rollins
- Functional Genomics Laboratory, Department of Psychiatry and Human Behavior, School of Medicine, University of California, Irvine, California, USA
| | - Michael Christiansen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Department of Biomedical Science, University of Copenhagen, Copenhagen, Denmark
| | - Mark A. Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Joanna Biernacka
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Marquis P. Vawter
- Functional Genomics Laboratory, Department of Psychiatry and Human Behavior, School of Medicine, University of California, Irvine, California, USA
| |
Collapse
|
19
|
Robson JP, Remke M, Kool M, Julian E, Korshunov A, Pfister SM, Osborne GW, Taylor MD, Wainwright B, Reynolds BA. Identification of CD24 as a marker of Patched1 deleted medulloblastoma-initiating neural progenitor cells. PLoS One 2019; 14:e0210665. [PMID: 30657775 PMCID: PMC6338368 DOI: 10.1371/journal.pone.0210665] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/28/2018] [Indexed: 12/15/2022] Open
Abstract
High morbidity and mortality are common traits of malignant tumours and identification of the cells responsible is a focus of on-going research. Many studies are now reporting the use of antibodies specific to Clusters of Differentiation (CD) cell surface antigens to identify tumour-initiating cell (TIC) populations in neural tumours. Medulloblastoma is one of the most common malignant brain tumours in children and despite a considerable amount of research investigating this tumour, the identity of the TICs, and the means by which such cells can be targeted remain largely unknown. Current prognostication and stratification of medulloblastoma using clinical factors, histology and genetic profiling have classified this tumour into four main subgroups: WNT, Sonic hedgehog (SHH), Group 3 and Group 4. Of these subgroups, SHH remains one of the most studied tumour groups due to the ability to model medulloblastoma formation through targeted deletion of the Shh pathway inhibitor Patched1 (Ptch1). Here we sought to utilise CD antibody expression to identify and isolate TIC populations in Ptch1 deleted medulloblastoma, and determine if these antibodies can help classify the identity of human medulloblastoma subgroups. Using a fluorescence-activated cell sorted (FACS) CD antibody panel, we identified CD24 as a marker of TICs in Ptch1 deleted medulloblastoma. CD24 expression was not correlated with markers of astrocytes or oligodendrocytes, but co-labelled with markers of neural progenitor cells. In conjunction with CD15, proliferating CD24+/CD15+ granule cell precursors (GCPs) were identified as a TIC population in Ptch1 deleted medulloblastoma. On human medulloblastoma, CD24 was found to be highly expressed on Group 3, Group 4 and SHH subgroups compared with the WNT subgroup, which was predominantly positive for CD15, suggesting CD24 is an important marker of non-WNT medulloblastoma initiating cells and a potential therapeutic target in human medulloblastoma. This study reports the use of CD24 and CD15 to isolate a GCP-like TIC population in Ptch1 deleted medulloblastoma, and suggests CD24 expression as a marker to help stratify human WNT tumours from other medulloblastoma subgroups.
Collapse
Affiliation(s)
- Jonathan P. Robson
- Division of Molecular Genetics and Development, Institute for Molecular Biosciences, University of Queensland, Brisbane, Queensland, Australia
- * E-mail:
| | - Marc Remke
- Department of Pediatric Neuro-Oncogenomics, German Cancer Research Centre and the German Cancer Consortium, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Marcel Kool
- Hopp Children´s Cancer Center at the National Center for Tumor Diseases, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center, Heidelberg, Germany
| | - Elaine Julian
- Division of Molecular Genetics and Development, Institute for Molecular Biosciences, University of Queensland, Brisbane, Queensland, Australia
| | - Andrey Korshunov
- Division of Clinical Cooperation Unit Neuropathology, German Cancer Research Centre, University of Heidelberg, Heidelberg, Germany
| | - Stefan M. Pfister
- Hopp Children´s Cancer Center at the National Center for Tumor Diseases, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center, Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Geoffrey W. Osborne
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- The Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Michael D. Taylor
- Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Brandon Wainwright
- Division of Molecular Genetics and Development, Institute for Molecular Biosciences, University of Queensland, Brisbane, Queensland, Australia
| | - Brent A. Reynolds
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
20
|
Breen MS, Ozcan S, Ramsey JM, Wang Z, Ma’ayan A, Rustogi N, Gottschalk MG, Webster MJ, Weickert CS, Buxbaum JD, Bahn S. Temporal proteomic profiling of postnatal human cortical development. Transl Psychiatry 2018; 8:267. [PMID: 30518843 PMCID: PMC6281671 DOI: 10.1038/s41398-018-0306-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 09/28/2018] [Accepted: 11/08/2018] [Indexed: 01/18/2023] Open
Abstract
Healthy cortical development depends on precise regulation of transcription and translation. However, the dynamics of how proteins are expressed, function and interact across postnatal human cortical development remain poorly understood. We surveyed the proteomic landscape of 69 dorsolateral prefrontal cortex samples across seven stages of postnatal life and integrated these data with paired transcriptome data. We detected 911 proteins by liquid chromatography-mass spectrometry, and 83 were significantly associated with postnatal age (FDR < 5%). Network analysis identified three modules of co-regulated proteins correlated with age, including two modules with increasing expression involved in gliogenesis and NADH metabolism and one neurogenesis-related module with decreasing expression throughout development. Integration with paired transcriptome data revealed that these age-related protein modules overlapped with RNA modules and displayed collinear developmental trajectories. Importantly, RNA expression profiles that are dynamically regulated throughout cortical development display tighter correlations with their respective translated protein expression compared to those RNA profiles that are not. Moreover, the correspondence between RNA and protein expression significantly decreases as a function of cortical aging, especially for genes involved in myelination and cytoskeleton organization. Finally, we used this data resource to elucidate the functional impact of genetic risk loci for intellectual disability, converging on gliogenesis, myelination and ATP-metabolism modules in the proteome and transcriptome. We share all data in an interactive, searchable companion website. Collectively, our findings reveal dynamic aspects of protein regulation and provide new insights into brain development, maturation, and disease.
Collapse
Affiliation(s)
- Michael S. Breen
- 0000 0001 0670 2351grid.59734.3cDepartment of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cDepartment of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cSeaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Sureyya Ozcan
- 0000000121885934grid.5335.0Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK
| | - Jordan M. Ramsey
- 0000000121885934grid.5335.0Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK
| | - Zichen Wang
- 0000 0001 0670 2351grid.59734.3cDepartment of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, BD2K-LINCS Data Coordination and Integration Center, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Avi Ma’ayan
- 0000 0001 0670 2351grid.59734.3cDepartment of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, BD2K-LINCS Data Coordination and Integration Center, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Nitin Rustogi
- 0000000121885934grid.5335.0Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK
| | - Michael G. Gottschalk
- 0000000121885934grid.5335.0Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK ,grid.5963.9Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg Germany, Freiburg, Germany
| | - Maree J. Webster
- Stanley Medical Research Institute, Laboratory of Brain Research, Rockville, MD 20850 USA
| | - Cynthia Shannon Weickert
- 0000 0000 8900 8842grid.250407.4Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW 2031 Australia ,0000 0004 4902 0432grid.1005.4School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052 Australia ,0000 0000 9159 4457grid.411023.5Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY 13210 USA
| | - Joseph D. Buxbaum
- 0000 0001 0670 2351grid.59734.3cDepartment of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cDepartment of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cSeaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Sabine Bahn
- 0000000121885934grid.5335.0Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK
| |
Collapse
|
21
|
Barfield ET, Gourley SL. Prefrontal cortical trkB, glucocorticoids, and their interactions in stress and developmental contexts. Neurosci Biobehav Rev 2018; 95:535-558. [PMID: 30477984 PMCID: PMC6392187 DOI: 10.1016/j.neubiorev.2018.10.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/14/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023]
Abstract
The tropomyosin/tyrosine receptor kinase B (trkB) and glucocorticoid receptor (GR) regulate neuron structure and function and the hormonal stress response. Meanwhile, disruption of trkB and GR activity (e.g., by chronic stress) can perturb neuronal morphology in cortico-limbic regions implicated in stressor-related illnesses like depression. Further, several of the short- and long-term neurobehavioral consequences of stress depend on the developmental timing and context of stressor exposure. We review how the levels and activities of trkB and GR in the prefrontal cortex (PFC) change during development, interact, are modulated by stress, and are implicated in depression. We review evidence that trkB- and GR-mediated signaling events impact the density and morphology of dendritic spines, the primary sites of excitatory synapses in the brain, highlighting effects in adolescents when possible. Finally, we review the role of neurotrophin and glucocorticoid systems in stress-related metaplasticity. We argue that better understanding the long-term effects of developmental stressors on PFC trkB, GR, and related factors may yield insights into risk for chronic, remitting depression and related neuropsychiatric illnesses.
Collapse
Affiliation(s)
- Elizabeth T Barfield
- Department of Pediatrics, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Graduate Program in Neuroscience, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Department of Psychiatry and Behavioral Sciences, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA.
| | - Shannon L Gourley
- Department of Pediatrics, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Graduate Program in Neuroscience, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Department of Psychiatry and Behavioral Sciences, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Molecular and Systems Pharmacology Program, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA.
| |
Collapse
|
22
|
Toker L, Mancarci BO, Tripathy S, Pavlidis P. Transcriptomic Evidence for Alterations in Astrocytes and Parvalbumin Interneurons in Subjects With Bipolar Disorder and Schizophrenia. Biol Psychiatry 2018; 84:787-796. [PMID: 30177255 PMCID: PMC6226343 DOI: 10.1016/j.biopsych.2018.07.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 07/05/2018] [Accepted: 07/06/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND High-throughput expression analyses of postmortem brain tissue have been widely used to study bipolar disorder and schizophrenia. However, despite the extensive efforts, no consensus has emerged as to the functional interpretation of the findings. We hypothesized that incorporating information on cell type-specific expression would provide new insights. METHODS We reanalyzed 15 publicly available bulk tissue expression datasets on schizophrenia and bipolar disorder, representing various brain regions from eight different cohorts of subjects (unique subjects: 332 control, 129 bipolar disorder, 341 schizophrenia). We studied changes in the expression profiles of cell type marker genes and evaluated whether these expression profiles could serve as surrogates for relative abundance of their corresponding cells. RESULTS In both bipolar disorder and schizophrenia, we consistently observed an increase in the expression profiles of cortical astrocytes and a decrease in the expression profiles of fast-spiking parvalbumin interneurons. No changes in astrocyte expression profiles were observed in subcortical regions. Furthermore, we found that many of the genes previously identified as differentially expressed in schizophrenia are highly correlated with the expression profiles of astrocytes or fast-spiking parvalbumin interneurons. CONCLUSIONS Our results indicate convergence of transcriptome studies of schizophrenia and bipolar disorder on changes in cortical astrocytes and fast-spiking parvalbumin interneurons, providing a unified interpretation of numerous studies. We suggest that these changes can be attributed to alterations in the relative abundance of the cells and are important for understanding the pathophysiology of the disorders.
Collapse
Affiliation(s)
- Lilah Toker
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Burak Ogan Mancarci
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Graduate Program in Bioinformatics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shreejoy Tripathy
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Paul Pavlidis
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
23
|
Malgulwar PB, Sharma V, Tomar AS, Verma C, Nambirajan A, Singh M, Suri V, Sarkar C, Sharma MC. Transcriptional co-expression regulatory network analysis for Snail and Slug identifies IL1R1, an inflammatory cytokine receptor, to be preferentially expressed in ST-EPN- RELA and PF-EPN-A molecular subgroups of intracranial ependymomas. Oncotarget 2018; 9:35480-35492. [PMID: 30464804 PMCID: PMC6231457 DOI: 10.18632/oncotarget.26211] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/08/2018] [Indexed: 12/14/2022] Open
Abstract
Recent molecular subgrouping of ependymomas (EPN) by DNA methylation profiling has identified ST-EPN-RELA and PF-EPN-A subgroups to be associated with poor outcome. Snail/Slug are cardinal epithelial-to-mesenchymal transcription factors (EMT-TFs) and are overexpressed in several CNS tumors, including EPNs. A systematic analysis of gene-sets/modules co-expressed with Snail and Slug genes using published expression microarray dataset (GSE27279)identified 634 genes for Snail with enriched TGF-β, PPAR and PI3K signaling pathways, and 757 genes for Slug with enriched focal adhesion, ECM-receptor interaction and regulation of actin cytoskeleton related pathways. Of 37 genes commonly expressed with both Snail and Slug, IL1R1, a cytokine receptor of interleukin-1 receptor family, was positively correlated with Snail (r=0.43) and Slug (r=0.51), preferentially expressed in ST-EPN-RELA and PF-EPN-A molecular groups, and enriched for pathways related to inflammation, angiogenesis and glycolysis. IL1R1 expression was fairly specific to EPNs among various CNS tumors analyzed. It also showed significant positive correlation with EMT, stemness and MDSC (myeloid derived suppressor cell) markers. Our study reports IL1R1 as a poor prognostic marker associated with EMT-like phenotype and stemness in EPNs. Our findings emphasize the need to further examine and validate IL1R1 as a novel therapeutic target in aggressive subsets of intracranial EPNs.
Collapse
Affiliation(s)
- Prit Benny Malgulwar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Vikas Sharma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Ashutosh Singh Tomar
- Center for Cellular and Molecular Biology-Council of Scientific and Industrial Research (CCMB-CSIR), Hyderabad, Telangana-500007, India
| | - Chaitenya Verma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Aruna Nambirajan
- Department of Pathology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Manmohan Singh
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Vaishali Suri
- Department of Pathology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Chitra Sarkar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Mehar Chand Sharma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi-110029, India
| |
Collapse
|
24
|
Sun L, Min L, Li M, Shao F, Wang W. Transcriptomic analysis reveals oxidative phosphorylation activation in an adolescent social isolation rat model. Brain Res Bull 2018; 142:304-312. [PMID: 30142370 DOI: 10.1016/j.brainresbull.2018.08.013] [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: 04/02/2018] [Revised: 07/23/2018] [Accepted: 08/18/2018] [Indexed: 12/29/2022]
Abstract
Complex interactions between genetic and environmental factors exert a sustained influence on the pathogenesis of schizophrenia (SCZ). Adolescent social isolation is regarded as a typical paradigm for SCZ. However, the underlying pathological mechanisms are not fully understood. In this study, adolescent Sprague-Dawley (SD) rats were placed in isolation rearing (IR) or social rearing (SR) conditions from postnatal day (PND) 21 to 34 to establish a SCZ disease model and a control model, respectively. Prepulse inhibition (PPI) assays and elevated plus maze tests were performed on PND 56. Next, prefrontal cortex (PFC) tissues were isolated for transcriptomic sequencing and RT-qPCR analyses. The results indicated that adolescent social isolation induced anxious behaviors and disrupted PPIs as well as specific PFC gene expression patterns in adult SD rats. A total of 196 genes were identified as upregulated, and 748 genes were identified as down-regulated in the IR group compared with those in the SR group. Differentially expressed genes (DEGs) were highly enriched in the KEGG pathways associated with the comorbidity of neurological disorder and oxidative phosphorylation (OXPHOS); 26 out of 27 comorbid neurological disorder-associated DEGs overlapped with 31 OXPHOS-associated DEGs. Those 26 overlapping DEGs were all upregulated in the IR group and could easily distinguish the IR group from the SR group; 6 of these DEGs (COX7C, NDUFB11, NDUFA2, NDUFC2, ATP5C1, and COX6A1) were verified by RT-qPCR. Here, we provide a systematic overview of gene expression alterations in adolescent-social-isolation-induced SCZ (ASI-SCZ), which suggests that genes that are associated with the comorbidity of neurological disorders, especially OXPHOS-related genes, contribute to the pathogenesis of ASI-SCZ.
Collapse
Affiliation(s)
- Lan Sun
- School of Psychological and Cognitive Sciences, Beijing Key Laboratory of Behavior and Mental Health, Peking University, Beijing, 100871, China
| | - Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Man Li
- Department of Psychology, School of Educational Science, Tianjin Normal University, Tianjin, 300387, China
| | - Feng Shao
- School of Psychological and Cognitive Sciences, Beijing Key Laboratory of Behavior and Mental Health, Peking University, Beijing, 100871, China.
| | - Weiwen Wang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
25
|
Erben L, He MX, Laeremans A, Park E, Buonanno A. A Novel Ultrasensitive In Situ Hybridization Approach to Detect Short Sequences and Splice Variants with Cellular Resolution. Mol Neurobiol 2018; 55:6169-6181. [PMID: 29264769 PMCID: PMC5994223 DOI: 10.1007/s12035-017-0834-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 12/08/2017] [Indexed: 01/30/2023]
Abstract
Investigating the expression of RNAs that differ by short or single nucleotide sequences at a single-cell level in tissue has been limited by the sensitivity and specificity of in situ hybridization (ISH) techniques. Detection of short isoform-specific sequences requires RNA isolation for PCR analysis-an approach that loses the regional and cell-type-specific distribution of isoforms. Having the capability to distinguish the differential expression of RNA variants in tissue is critical because alterations in mRNA splicing and editing, as well as coding single nucleotide polymorphisms, have been associated with numerous cancers, neurological and psychiatric disorders. Here we introduce a novel highly sensitive single-probe colorimetric/fluorescent ISH approach that targets short exon/exon RNA splice junctions using single-pair oligonucleotide probes (~ 50 bp). We use this approach to investigate, with single-cell resolution, the expression of four transcripts encoding the neuregulin (NRG) receptor ErbB4 that differ by alternative splicing of exons encoding two juxtamembrane (JMa/JMb) and two cytoplasmic (CYT-1/CYT-2) domains that alter receptor stability and signaling modes, respectively. By comparing ErbB4 hybridization on sections from wild-type and ErbB4 knockout mice (missing exon 2), we initially demonstrate that single-pair probes provide the sensitivity and specificity to visualize and quantify the differential expression of ErbB4 isoforms. Using cell-type-specific GFP reporter mice, we go on to demonstrate that expression of ErbB4 isoforms differs between neurons and oligodendrocytes, and that this differential expression of ErbB4 isoforms is evolutionarily conserved to humans. This single-pair probe ISH approach, known as BaseScope, could serve as an invaluable diagnostic tool to detect alternative spliced isoforms, and potentially single base polymorphisms, associated with disease.
Collapse
Affiliation(s)
- Larissa Erben
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bldg. 35, Room 2C-1000, Bethesda, MD, 20892, USA
- Institute of Molecular Psychiatry, University Bonn, 53127, Bonn, Germany
| | - Ming-Xiao He
- Advanced Cell Diagnostics, Newark, CA, 94560, USA
| | | | - Emily Park
- Advanced Cell Diagnostics, Newark, CA, 94560, USA
| | - Andres Buonanno
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bldg. 35, Room 2C-1000, Bethesda, MD, 20892, USA.
| |
Collapse
|
26
|
Meel MH, Schaper SA, Kaspers GJL, Hulleman E. Signaling pathways and mesenchymal transition in pediatric high-grade glioma. Cell Mol Life Sci 2018; 75:871-887. [PMID: 29164272 PMCID: PMC5809527 DOI: 10.1007/s00018-017-2714-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/24/2017] [Accepted: 11/14/2017] [Indexed: 12/16/2022]
Abstract
Pediatric high-grade gliomas (pHGG), including diffuse intrinsic pontine gliomas (DIPG), are the most lethal types of cancer in children. In recent years, it has become evident that these tumors are driven by epigenetic events, mainly mutations involving genes encoding Histone 3, setting them apart from their adult counterparts. These tumors are exceptionally resistant to chemotherapy and respond only temporarily to radiotherapy. Moreover, their delicate location and diffuse growth pattern make complete surgical resection impossible. In many other forms of cancer, chemo- and radioresistance, in combination with a diffuse, invasive phenotype, are associated with a transcriptional program termed the epithelial-to-mesenchymal transition (EMT). Activation of this program allows cancer cells to survive individually, invade surrounding tissues and metastasize. It also enables them to survive exposure to cytotoxic therapy, including chemotherapeutic drugs and radiation. We here suggest that EMT plays an important, yet poorly understood role in the biology and therapy resistance of pHGG and DIPG. This review summarizes the current knowledge on the major signal transduction pathways and transcription factors involved in the epithelial-to-mesenchymal transition in cancer in general and in pediatric HGG and DIPG in particular. Despite the fact that the mesenchymal transition has not yet been specifically studied in pHGG and DIPG, activation of pathways and high levels of transcription factors involved in EMT have been described. We conclude that the mesenchymal transition is likely to be an important element of the biology of pHGG and DIPG and warrants further investigation for the development of novel therapeutics.
Collapse
Affiliation(s)
- Michaël H Meel
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Sophie A Schaper
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Gertjan J L Kaspers
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Uppsalalaan 8, 3584CT, Utrecht, The Netherlands
| | - Esther Hulleman
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| |
Collapse
|
27
|
Hartung H, Cichon N, De Feo V, Riemann S, Schildt S, Lindemann C, Mulert C, Gogos JA, Hanganu-Opatz IL. From Shortage to Surge: A Developmental Switch in Hippocampal-Prefrontal Coupling in a Gene-Environment Model of Neuropsychiatric Disorders. Cereb Cortex 2018; 26:4265-4281. [PMID: 27613435 PMCID: PMC5066837 DOI: 10.1093/cercor/bhw274] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 08/10/2016] [Indexed: 12/21/2022] Open
Abstract
Cognitive deficits represent a major burden of neuropsychiatric disorders and result in part from abnormal communication within hippocampal–prefrontal circuits. While it has been hypothesized that this network dysfunction arises during development, long before the first clinical symptoms, experimental evidence is still missing. Here, we show that pre-juvenile mice mimicking genetic and environmental risk factors of disease (dual-hit GE mice) have poorer recognition memory that correlates with augmented coupling by synchrony and stronger directed interactions between prefrontal cortex and hippocampus. The network dysfunction emerges already during neonatal development, yet it initially consists in a diminished hippocampal theta drive and consequently, a weaker and disorganized entrainment of local prefrontal circuits in discontinuous oscillatory activity in dual-hit GE mice when compared with controls. Thus, impaired maturation of functional communication within hippocampal–prefrontal networks switching from hypo- to hyper-coupling may represent a mechanism underlying the pathophysiology of cognitive deficits in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Henrike Hartung
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Laboratory of Neurobiology, Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland
| | - Nicole Cichon
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Vito De Feo
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Laboratory of Neural Computation, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy
| | - Stephanie Riemann
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Current address: German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Sandra Schildt
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Christoph Lindemann
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Christoph Mulert
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Joseph A Gogos
- Department of Neuroscience, Columbia University, New York, NY 10032, USA.,Department of Physiology, Columbia University, New York, NY 10032, USA
| | - Ileana L Hanganu-Opatz
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
28
|
Zbukvic IC, Hyun Kim J. Divergent prefrontal dopaminergic mechanisms mediate drug- and fear-associated cue extinction during adolescence versus adulthood. Eur Neuropsychopharmacol 2018; 28:1-12. [PMID: 29174948 DOI: 10.1016/j.euroneuro.2017.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/08/2017] [Accepted: 11/03/2017] [Indexed: 01/21/2023]
Abstract
Cue-associated learning is vital to guiding behaviour for survival. Adolescence represents a key developmental stage for perturbations in cue-related learning, including a characteristic deficit in cue extinction learning. The present review summarizes evidence from animal and human literature that cue extinction is critically mediated by prefrontal dopamine, a system that undergoes dramatic reorganization during adolescence. We propose that extinction learning and memory is governed by a developmentally dynamic balance of dopamine receptors in the prefrontal cortex, which changes across adolescence into adulthood. This is contrary to the previous idea that extinction deficits during adolescence reflect inefficiency in the same neural circuitry as adults. This leads to proposal of the novel theory that cue extinction involves divergent prefrontal dopaminergic mechanisms depending on the age of extinction.
Collapse
Affiliation(s)
- Isabel C Zbukvic
- Black Dog Institute, Randwick 2031, NSW, Australia; Behavioral Neuroscience Division, The Florey Institute of Neuroscience & Mental Health, Parkville 3051, VIC, Australia.
| | - Jee Hyun Kim
- Behavioral Neuroscience Division, The Florey Institute of Neuroscience & Mental Health, Parkville 3051, VIC, Australia; The Florey Department of Neuroscience & Mental Health, University of Melbourne, Parkville 3010, VIC, Australia
| |
Collapse
|
29
|
Hrvoj-Mihic B, Hanson KL, Lew CH, Stefanacci L, Jacobs B, Bellugi U, Semendeferi K. Basal Dendritic Morphology of Cortical Pyramidal Neurons in Williams Syndrome: Prefrontal Cortex and Beyond. Front Neurosci 2017; 11:419. [PMID: 28848376 PMCID: PMC5554499 DOI: 10.3389/fnins.2017.00419] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/05/2017] [Indexed: 12/26/2022] Open
Abstract
Williams syndrome (WS) is a unique neurodevelopmental disorder with a specific behavioral and cognitive profile, which includes hyperaffiliative behavior, poor social judgment, and lack of social inhibition. Here we examined the morphology of basal dendrites on pyramidal neurons in the cortex of two rare adult subjects with WS. Specifically, we examined two areas in the prefrontal cortex (PFC)-the frontal pole (Brodmann area 10) and the orbitofrontal cortex (Brodmann area 11)-and three areas in the motor, sensory, and visual cortex (BA 4, BA 3-1-2, BA 18). The findings suggest that the morphology of basal dendrites on the pyramidal neurons is altered in the cortex of WS, with differences that were layer-specific, more prominent in PFC areas, and displayed an overall pattern of dendritic organization that differentiates WS from other disorders. In particular, and unlike what was expected based on typically developing brains, basal dendrites in the two PFC areas did not display longer and more branched dendrites compared to motor, sensory and visual areas. Moreover, dendritic branching, dendritic length, and the number of dendritic spines differed little within PFC and between the central executive region (BA 10) and BA 11 that is part of the orbitofrontal region involved into emotional processing. In contrast, the relationship between the degree of neuronal branching in supra- versus infra-granular layers was spared in WS. Although this study utilized tissue held in formalin for a prolonged period of time and the number of neurons available for analysis was limited, our findings indicate that WS cortex, similar to that in other neurodevelopmental disorders such as Down syndrome, Rett syndrome, Fragile X, and idiopathic autism, has altered morphology of basal dendrites on pyramidal neurons, which appears more prominent in selected areas of the PFC. Results were examined from developmental perspectives and discussed in the context of other neurodevelopmental disorders. We have proposed hypotheses for further investigations of morphological changes on basal dendrites in WS, a syndrome of particular interest given its unique social and cognitive phenotype.
Collapse
Affiliation(s)
- Branka Hrvoj-Mihic
- Department of Anthropology, University of California, San DiegoSan Diego, La Jolla, CA, United States
| | - Kari L Hanson
- Department of Anthropology, University of California, San DiegoSan Diego, La Jolla, CA, United States
| | - Caroline H Lew
- Department of Anthropology, University of California, San DiegoSan Diego, La Jolla, CA, United States
| | - Lisa Stefanacci
- Department of Anthropology, University of California, San DiegoSan Diego, La Jolla, CA, United States
| | - Bob Jacobs
- Neuroscience Program, Colorado CollegeColorado Springs, CO, United States
| | - Ursula Bellugi
- Laboratory for Cognitive Neuroscience, The Salk Institute for Biological StudiesLa Jolla, CA, United States
| | - Katerina Semendeferi
- Department of Anthropology, University of California, San DiegoSan Diego, La Jolla, CA, United States.,Kavli Institute for Brain and Mind, University of California, San DiegoSan Diego, La Jolla, CA, United States
| |
Collapse
|
30
|
Oxidative stress, prefrontal cortex hypomyelination and cognitive symptoms in schizophrenia. Transl Psychiatry 2017; 7:e1171. [PMID: 28934193 PMCID: PMC5538118 DOI: 10.1038/tp.2017.138] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/12/2017] [Accepted: 05/06/2017] [Indexed: 12/13/2022] Open
Abstract
Schizophrenia (SZ) is a neurodevelopmental disorder with a broad symptomatology, including cognitive symptoms that are thought to arise from the prefrontal cortex (PFC). The neurobiological aetiology of these symptoms remains elusive, yet both impaired redox control and PFC dysconnectivity have been recently implicated. PFC dysconnectivity has been linked to white matter, oligodendrocyte (OL) and myelin abnormalities in SZ patients. Myelin is produced by mature OLs, and OL precursor cells (OPCs) are exceptionally susceptible to oxidative stress. Here we propose a hypothesis for the aetiology of cognitive symptomatology in SZ: the redox-induced prefrontal OPC-dysfunctioning hypothesis. We pose that the combination of genetic and environmental factors causes oxidative stress marked by a build-up of reactive oxygen species that, during late adolescence, impair OPC signal transduction processes that are necessary for OPC proliferation and differentiation, and involve AMP-activated protein kinase, Akt-mTOR-P70S6K and peroxisome proliferator receptor alpha signalling. OPC dysfunctioning coincides with the relatively late onset of PFC myelination, causing hypomyelination and disruption of connectivity in this brain area. The resulting cognitive deficits arise in parallel with SZ onset. Hence, our hypothesis provides a novel neurobiological framework for the aetiology of SZ cognitive symptoms. Future research addressing our hypothesis could have important implications for the development of new (combined) antioxidant- and promyelination-based strategies to treat the cognitive symptoms in SZ.
Collapse
|
31
|
Agoglia AE, Holstein SE, Small AT, Spanos M, Burrus BM, Hodge CW. Comparison of the adolescent and adult mouse prefrontal cortex proteome. PLoS One 2017; 12:e0178391. [PMID: 28570644 PMCID: PMC5453624 DOI: 10.1371/journal.pone.0178391] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 05/14/2017] [Indexed: 12/28/2022] Open
Abstract
Adolescence is a developmental period characterized by unique behavioral phenotypes (increased novelty seeking, risk taking, sociability and impulsivity) and increased risk for destructive behaviors, impaired decision making and psychiatric illness. Adaptive and maladaptive adolescent traits have been associated with development of the medial prefrontal cortex (mPFC), a brain region that mediates regulatory control of behavior. However, the molecular changes that underlie brain development and behavioral vulnerability have not been fully characterized. Using high-throughput 2D DIGE spot profiling with identification by MALDI-TOF mass spectrometry, we identified 62 spots in the PFC that exhibited age-dependent differences in expression. Identified proteins were associated with diverse cellular functions, including intracellular signaling, synaptic plasticity, cellular organization and metabolism. Separate Western blot analyses confirmed age-related changes in DPYSL2, DNM1, STXBP1 and CFL1 in the mPFC and expanded these findings to the dorsal striatum, nucleus accumbens, motor cortex, amygdala and ventral tegmental area. Ingenuity Pathway Analysis (IPA) identified functional interaction networks enriched with proteins identified in the proteomics screen, linking age-related alterations in protein expression to cellular assembly and development, cell signaling and behavior, and psychiatric illness. These results provide insight into potential molecular components of adolescent cortical development, implicating structural processes that begin during embryonic development as well as plastic adaptations in signaling that may work in concert to bring the cortex, and other brain regions, into maturity.
Collapse
Affiliation(s)
- Abigail E. Agoglia
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Sarah E. Holstein
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Amanda T. Small
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Marina Spanos
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Brainard M. Burrus
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Clyde W. Hodge
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
32
|
Sewing ACP, Lagerweij T, van Vuurden DG, Meel MH, Veringa SJE, Carcaboso AM, Gaillard PJ, Peter Vandertop W, Wesseling P, Noske D, Kaspers GJL, Hulleman E. Preclinical evaluation of convection-enhanced delivery of liposomal doxorubicin to treat pediatric diffuse intrinsic pontine glioma and thalamic high-grade glioma. J Neurosurg Pediatr 2017; 19:518-530. [PMID: 28291423 DOI: 10.3171/2016.9.peds16152] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Pediatric high-grade gliomas (pHGGs) including diffuse intrinsic pontine gliomas (DIPGs) are primary brain tumors with high mortality and morbidity. Because of their poor brain penetrance, systemic chemotherapy regimens have failed to deliver satisfactory results; however, convection-enhanced delivery (CED) may be an alternative mode of drug delivery. Anthracyclines are potent chemotherapeutics that have been successfully delivered via CED in preclinical supratentorial glioma models. This study aims to assess the potency of anthracyclines against DIPG and pHGG cell lines in vitro and to evaluate the efficacy of CED with anthracyclines in orthotopic pontine and thalamic tumor models. METHODS The sensitivity of primary pHGG cell lines to a range of anthracyclines was tested in vitro. Preclinical CED of free doxorubicin and pegylated liposomal doxorubicin (PLD) to the brainstem and thalamus of naïve nude mice was performed. The maximum tolerated dose (MTD) was determined based on the observation of clinical symptoms, and brains were analyzed after H & E staining. Efficacy of the MTD was tested in adult glioma E98-FM-DIPG and E98-FM-thalamus models and in the HSJD-DIPG-007-Fluc primary DIPG model. RESULTS Both pHGG and DIPG cells were sensitive to anthracyclines in vitro. Doxorubicin was selected for further preclinical evaluation. Convection-enhanced delivery of the MTD of free doxorubicin and PLD in the pons was 0.02 mg/ml, and the dose tolerated in the thalamus was 10 times higher (0.2 mg/ml). Free doxorubicin or PLD via CED was ineffective against E98-FM-DIPG or HSJD-DIPG-007-Fluc in the brainstem; however, when applied in the thalamus, 0.2 mg/ml of PLD slowed down tumor growth and increased survival in a subset of animals with small tumors. CONCLUSIONS Local delivery of doxorubicin to the brainstem causes severe toxicity, even at doxorubicin concentrations that are safe in the thalamus. As a consequence, the authors could not establish a therapeutic window for treating orthotopic brainstem tumors in mice. For tumors in the thalamus, therapeutic concentrations to slow down tumor growth could be reached. These data suggest that anatomical location determines the severity of toxicity after local delivery of therapeutic agents and that caution should be used when translating data from supratentorial CED studies to treat infratentorial tumors.
Collapse
Affiliation(s)
- A Charlotte P Sewing
- Departments of 1 Pediatric Oncology.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Tonny Lagerweij
- Neurosurgery, and.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Dannis G van Vuurden
- Departments of 1 Pediatric Oncology.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Michaël H Meel
- Departments of 1 Pediatric Oncology.,Neurosurgery, and.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Susanna J E Veringa
- Departments of 1 Pediatric Oncology.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Angel M Carcaboso
- Preclinical Therapeutics and Drug Delivery Research Program, Department of Oncology, Hospital Sant Joan de Déu Barcelona, Spain
| | | | - W Peter Vandertop
- Neurosurgery, and.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Pieter Wesseling
- Pathology.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam.,2-BBB Medicines, Leiden.,Department of Pathology, RadboudUMC, Nijmegen
| | - David Noske
- Neurosurgery, and.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| | - Gertjan J L Kaspers
- Neuro-Oncology Research Group.,Academy of Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands ; and
| | - Esther Hulleman
- Departments of 1 Pediatric Oncology.,Neuro-Oncology Research Group.,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam
| |
Collapse
|
33
|
Zbukvic IC, Park CHJ, Ganella DE, Lawrence AJ, Kim JH. Prefrontal Dopaminergic Mechanisms of Extinction in Adolescence Compared to Adulthood in Rats. Front Behav Neurosci 2017; 11:32. [PMID: 28275342 PMCID: PMC5319962 DOI: 10.3389/fnbeh.2017.00032] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 02/10/2017] [Indexed: 01/11/2023] Open
Abstract
Adolescents with anxiety disorders attain poorer outcomes following extinction-based treatment compared to adults. Extinction deficit during adolescence has been identified to involve immaturity in the medial prefrontal cortex (mPFC). Findings from adult rodents suggest extinction involves dopamine signaling in the mPFC. This system changes dramatically during adolescence, but its role in adolescent extinction is unknown. Therefore, we investigated the role of prefrontal dopamine in extinction using Pavlovian fear conditioning in adolescent and adult rats. Using quantitative PCR (qPCR) analyses, we measured changes in dopamine receptor gene expression in the mPFC before and after extinction. We then enhanced dopamine 1 receptor (D1R) or dopamine 2 receptor (D2R) signaling in the infralimbic cortex (IL) of the mPFC using agonists at the time of extinction. Adolescent rats displayed a deficit in extinction retention compared to adults. Extinction induced a reduction in D1R compared to D2R gene expression in adolescent rats, whereas an increase of D1R compared to D2R gene expression was observed in adult rats. Acutely enhancing IL D1R signaling using SKF-81297 had no effect on extinction at either age. In contrast, acutely enhancing IL D2R signaling with quinpirole significantly enhanced long-term extinction in adolescents, and impaired within-session extinction in adults. Our results suggest a dissociated role for prefrontal dopamine in fear extinction during adolescence compared to adulthood. Findings highlight the dopamine system as a potential pharmacological target to improve extinction-based treatments for adolescents.
Collapse
Affiliation(s)
- Isabel C Zbukvic
- Developmental Psychobiology Laboratory, Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, VIC, Australia; Developmental Psychobiology Laboratory, The Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| | - Chun Hui J Park
- Developmental Psychobiology Laboratory, Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, VIC, Australia; Developmental Psychobiology Laboratory, The Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| | - Despina E Ganella
- Developmental Psychobiology Laboratory, Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, VIC, Australia; Developmental Psychobiology Laboratory, The Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| | - Andrew J Lawrence
- Developmental Psychobiology Laboratory, Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, VIC, Australia; Developmental Psychobiology Laboratory, The Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| | - Jee Hyun Kim
- Developmental Psychobiology Laboratory, Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, VIC, Australia; Developmental Psychobiology Laboratory, The Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| |
Collapse
|
34
|
Howes OD, McCutcheon R. Inflammation and the neural diathesis-stress hypothesis of schizophrenia: a reconceptualization. Transl Psychiatry 2017; 7:e1024. [PMID: 28170004 PMCID: PMC5438023 DOI: 10.1038/tp.2016.278] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/27/2016] [Indexed: 12/12/2022] Open
Abstract
An interaction between external stressors and intrinsic vulnerability is one of the longest standing pathoaetiological explanations for schizophrenia. However, novel lines of evidence from genetics, preclinical studies, epidemiology and imaging have shed new light on the mechanisms that may underlie this, implicating microglia as a key potential mediator. Microglia are the primary immune cells of the central nervous system. They have a central role in the inflammatory response, and are also involved in synaptic pruning and neuronal remodeling. In addition to immune and traumatic stimuli, microglial activation occurs in response to psychosocial stress. Activation of microglia perinatally may make them vulnerable to subsequent overactivation by stressors experienced in later life. Recent advances in genetics have shown that variations in the complement system are associated with schizophrenia, and this system has been shown to regulate microglial synaptic pruning. This suggests a mechanism via which genetic and environmental influences may act synergistically and lead to pathological microglial activation. Microglial overactivation may lead to excessive synaptic pruning and loss of cortical gray matter. Microglial mediated damage to stress-sensitive regions such as the prefrontal cortex and hippocampus may lead directly to cognitive and negative symptoms, and account for a number of the structural brain changes associated with the disorder. Loss of cortical control may also lead to disinhibition of subcortical dopamine-thereby leading to positive psychotic symptoms. We review the preclinical and in vivo evidence for this model and consider the implications this has for treatment, and future directions.
Collapse
Affiliation(s)
- O D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK,MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK,PET Imaging Group, MRC Clinical Sciences Centre, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK. E-mail:
| | - R McCutcheon
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK,MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
35
|
Stedehouder J, Kushner SA. Myelination of parvalbumin interneurons: a parsimonious locus of pathophysiological convergence in schizophrenia. Mol Psychiatry 2017; 22:4-12. [PMID: 27646261 PMCID: PMC5414080 DOI: 10.1038/mp.2016.147] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 07/09/2016] [Accepted: 07/13/2016] [Indexed: 12/11/2022]
Abstract
Schizophrenia is a debilitating psychiatric disorder characterized by positive, negative and cognitive symptoms. Despite more than a century of research, the neurobiological mechanism underlying schizophrenia remains elusive. White matter abnormalities and interneuron dysfunction are the most widely replicated cellular neuropathological alterations in patients with schizophrenia. However, a unifying model incorporating these findings has not yet been established. Here, we propose that myelination of fast-spiking parvalbumin (PV) interneurons could be an important locus of pathophysiological convergence in schizophrenia. Myelination of interneurons has been demonstrated across a wide diversity of brain regions and appears highly specific for the PV interneuron subclass. Given the critical influence of fast-spiking PV interneurons for mediating oscillations in the gamma frequency range (~30-120 Hz), PV myelination is well positioned to optimize action potential fidelity and metabolic homeostasis. We discuss this hypothesis with consideration of data from human postmortem studies, in vivo brain imaging and electrophysiology, and molecular genetics, as well as fundamental and translational studies in rodent models. Together, the parvalbumin interneuron myelination hypothesis provides a falsifiable model for guiding future studies of schizophrenia pathophysiology.
Collapse
Affiliation(s)
- J Stedehouder
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - S A Kushner
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
36
|
Ueno H, Suemitsu S, Okamoto M, Matsumoto Y, Ishihara T. Parvalbumin neurons and perineuronal nets in the mouse prefrontal cortex. Neuroscience 2016; 343:115-127. [PMID: 27923740 DOI: 10.1016/j.neuroscience.2016.11.035] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/22/2016] [Accepted: 11/23/2016] [Indexed: 11/30/2022]
Abstract
The prefrontal cortex (PFC) plays a key role in cognitive functions, memory, and attention. Alterations in parvalbumin interneurons (PV neurons) and perineuronal nets (PNNs) within the PFC have been implicated in schizophrenia and autism spectrum disorder pathology. However, it remains unclear why PV neurons and PNNs in the PFC are selectively impaired. Here we aimed to clarify if PV neurons and PNNs in the PFC have region-specific features. We found that PV neurons and PNNs were increased in a region-specific manner in the PFC during postnatal development. In the mature PFC, the expression of PV protein is lower than in other parts of the cortex. Furthermore, PNNs in the mature PFC are not typical lattice-like structures and do not have the major components of PNNs and tenascin-R. The present study indicates that PV neurons and PNNs have region-specific features, and our results suggest that PV neurons and PNNs have structural vulnerability within the PFC.
Collapse
Affiliation(s)
- Hiroshi Ueno
- Department of Medical Technology, Kawasaki College of Allied Health Professions, Okayama 701-0194, Japan; Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700-8558, Japan.
| | - Shunsuke Suemitsu
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Motoi Okamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700-8558, Japan
| | - Yosuke Matsumoto
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Takeshi Ishihara
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan
| |
Collapse
|
37
|
Rao YS, Pak TR. microRNAs and the adolescent brain: Filling the knowledge gap. Neurosci Biobehav Rev 2016; 70:313-322. [PMID: 27328787 PMCID: PMC5074866 DOI: 10.1016/j.neubiorev.2016.06.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 06/09/2016] [Accepted: 06/11/2016] [Indexed: 12/14/2022]
Abstract
Over two decades ago the discovery of microRNAs (miRNA) broadened our understanding of the diverse molecular pathways mediating post-transcriptional control over gene expression. These small non-coding RNAs dynamically fluctuate, temporally and spatially, throughout the lifespan of all organisms. The fundamental role that miRNAs have in shaping embryonic neurodevelopment provides strong evidence that adolescent brain remodeling could be rooted in the changing miRNA landscape of the cell. Few studies have directly measured miRNA gene expression changes in the brain across pubertal development, and even less is known about the functional impact of those miRNAs on the maturational processes that occur in the developing adolescent brain. This review summarizes miRNA biogenesis and function in the brain in the context of normal (i.e. not diseased) physiology. These landmark studies can guide predictions about the role of miRNAs in facilitating maturation of the adolescent brain. However, there are clear indicators that adolescence/puberty is a unique life stage, suggesting miRNA function during adolescence is distinct from those in any other previously described system.
Collapse
Affiliation(s)
- Yathindar S Rao
- Loyola University Chicago, Stritch School of Medicine, Department of Cell and Molecular Physiology, United States
| | - Toni R Pak
- Loyola University Chicago, Stritch School of Medicine, Department of Cell and Molecular Physiology, United States.
| |
Collapse
|
38
|
Mostaid MS, Lloyd D, Liberg B, Sundram S, Pereira A, Pantelis C, Karl T, Weickert CS, Everall IP, Bousman CA. Neuregulin-1 and schizophrenia in the genome-wide association study era. Neurosci Biobehav Rev 2016; 68:387-409. [DOI: 10.1016/j.neubiorev.2016.06.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 05/30/2016] [Accepted: 06/03/2016] [Indexed: 12/22/2022]
|
39
|
Steullet P, Cabungcal JH, Monin A, Dwir D, O'Donnell P, Cuenod M, Do KQ. Redox dysregulation, neuroinflammation, and NMDA receptor hypofunction: A "central hub" in schizophrenia pathophysiology? Schizophr Res 2016; 176:41-51. [PMID: 25000913 PMCID: PMC4282982 DOI: 10.1016/j.schres.2014.06.021] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 06/06/2014] [Accepted: 06/08/2014] [Indexed: 12/18/2022]
Abstract
Accumulating evidence points to altered GABAergic parvalbumin-expressing interneurons and impaired myelin/axonal integrity in schizophrenia. Both findings could be due to abnormal neurodevelopmental trajectories, affecting local neuronal networks and long-range synchrony and leading to cognitive deficits. In this review, we present data from animal models demonstrating that redox dysregulation, neuroinflammation and/or NMDAR hypofunction (as observed in patients) impairs the normal development of both parvalbumin interneurons and oligodendrocytes. These observations suggest that a dysregulation of the redox, neuroimmune, and glutamatergic systems due to genetic and early-life environmental risk factors could contribute to the anomalies of parvalbumin interneurons and white matter in schizophrenia, ultimately impacting cognition, social competence, and affective behavior via abnormal function of micro- and macrocircuits. Moreover, we propose that the redox, neuroimmune, and glutamatergic systems form a "central hub" where an imbalance within any of these "hub" systems leads to similar anomalies of parvalbumin interneurons and oligodendrocytes due to the tight and reciprocal interactions that exist among these systems. A combination of vulnerabilities for a dysregulation within more than one of these systems may be particularly deleterious. For these reasons, molecules, such as N-acetylcysteine, that possess antioxidant and anti-inflammatory properties and can also regulate glutamatergic transmission are promising tools for prevention in ultra-high risk patients or for early intervention therapy during the first stages of the disease.
Collapse
Affiliation(s)
- P Steullet
- Center for Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Site de Cery, 1008 Prilly-Lausanne, Switzerland
| | - J H Cabungcal
- Center for Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Site de Cery, 1008 Prilly-Lausanne, Switzerland
| | - A Monin
- Center for Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Site de Cery, 1008 Prilly-Lausanne, Switzerland
| | - D Dwir
- Center for Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Site de Cery, 1008 Prilly-Lausanne, Switzerland
| | - P O'Donnell
- Neuroscience Research Unit, Pfizer, Inc., 700 Main Street, Cambridge, MA 02139, USA
| | - M Cuenod
- Center for Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Site de Cery, 1008 Prilly-Lausanne, Switzerland
| | - K Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Site de Cery, 1008 Prilly-Lausanne, Switzerland.
| |
Collapse
|
40
|
Nyima T, Müller M, Hooiveld GJEJ, Morine MJ, Scotti M. Nonlinear transcriptomic response to dietary fat intake in the small intestine of C57BL/6J mice. BMC Genomics 2016; 17:106. [PMID: 26861690 PMCID: PMC4748552 DOI: 10.1186/s12864-016-2424-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 02/02/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND A high caloric diet, in conjunction with low levels of physical activity, promotes obesity. Many studies are available regarding the relation between dietary saturated fats and the etiology of obesity, but most focus on liver, muscle and white adipose tissue. Furthermore, the majority of transcriptomic studies seek to identify linear effects of an external stimulus on gene expression, although such an assumption does not necessarily hold. Our work assesses the dose-dependent effects of dietary fat intake on differential gene expression in the proximal, middle and distal sections of the small intestine in C57BL/6J mice. Gene expression is analyzed in terms of either linear or nonlinear responses to fat intake. RESULTS The highest number of differentially expressed genes was observed in the middle section. In all intestine sections, most of the identified processes exhibited a linear response to increasing fat intake. The relative importance of logarithmic and exponential responses was higher in the proximal and distal sections, respectively. Functional enrichment analysis highlighted a constantly linear regulation of acute-phase response along the whole small intestine, with up-regulation of Serpina1b. The study of gene expression showed that exponential down-regulation of cholesterol transport in the middle section is coupled with logarithmic up-regulation of cholesterol homeostasis. A shift from linear to exponential response was observed in genes involved in the negative regulation of caspase activity, from middle to distal section (e.g., Birc5, up-regulated). CONCLUSIONS The transcriptomic signature associated with inflammatory processes preserved a linear response in the whole small intestine (e.g., up-regulation of Serpina1b). Processes related to cholesterol homeostasis were particularly active in the middle small intestine and only the highest fat intake down-regulated cholesterol transport and efflux (with a key role played by the down-regulation of ATP binding cassette transporters). Characterization of nonlinear patterns of gene expression triggered by different levels of dietary fat is an absolute novelty in intestinal studies. This approach helps identifying which processes are overloaded (i.e., positive, logarithmic regulation) or arrested (i.e., negative, exponential regulation) in response to excessive fat intake, and can shed light on the relationships linking lipid intake to obesity and its associated molecular disturbances.
Collapse
Affiliation(s)
- Tenzin Nyima
- The Microsoft Research - University of Trento Centre for Computational and Systems Biology, Rovereto, Italy.
| | - Michael Müller
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands. .,Norwich Medical School, University of East Anglia, Norwich, UK.
| | - Guido J E J Hooiveld
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands.
| | - Melissa J Morine
- The Microsoft Research - University of Trento Centre for Computational and Systems Biology, Rovereto, Italy.
| | - Marco Scotti
- The Microsoft Research - University of Trento Centre for Computational and Systems Biology, Rovereto, Italy. .,GEOMAR Helmholtz Centre for Ocean Research, Kiel, Germany.
| |
Collapse
|
41
|
Monin A, Fournier M, Baumann PS, Cuénod M, Do KQ. Role of Redox Dysregulation in White Matter Anomalies Associated with Schizophrenia. HANDBOOK OF BEHAVIORAL NEUROSCIENCE 2016. [DOI: 10.1016/b978-0-12-800981-9.00028-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
42
|
Hjelm BE, Rollins B, Mamdani F, Lauterborn JC, Kirov G, Lynch G, Gall CM, Sequeira A, Vawter MP. Evidence of Mitochondrial Dysfunction within the Complex Genetic Etiology of Schizophrenia. MOLECULAR NEUROPSYCHIATRY 2015; 1:201-19. [PMID: 26550561 DOI: 10.1159/000441252] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 09/22/2015] [Indexed: 01/19/2023]
Abstract
Genetic evidence has supported the hypothesis that schizophrenia (SZ) is a polygenic disorder caused by the disruption in function of several or many genes. The most common and reproducible cellular phenotype associated with SZ is a reduction in dendritic spines within the neocortex, suggesting alterations in dendritic architecture may cause aberrant cortical circuitry and SZ symptoms. Here, we review evidence supporting a multifactorial model of mitochondrial dysfunction in SZ etiology and discuss how these multiple paths to mitochondrial dysfunction may contribute to dendritic spine loss and/or underdevelopment in some SZ subjects. The pathophysiological role of mitochondrial dysfunction in SZ is based upon genomic analyses of both the mitochondrial genome and nuclear genes involved in mitochondrial function. Previous studies and preliminary data suggest SZ is associated with specific alleles and haplogroups of the mitochondrial genome, and also correlates with a reduction in mitochondrial copy number and an increase in synonymous and nonsynonymous substitutions of mitochondrial DNA. Mitochondrial dysfunction has also been widely implicated in SZ by genome-wide association, exome sequencing, altered gene expression, proteomics, microscopy analyses, and induced pluripotent stem cell studies. Together, these data support the hypothesis that SZ is a polygenic disorder with an enrichment of mitochondrial targets.
Collapse
Affiliation(s)
- Brooke E Hjelm
- Departments of Psychiatry & Human Behavior, University of California, Irvine, Calif., USA
| | - Brandi Rollins
- Departments of Psychiatry & Human Behavior, University of California, Irvine, Calif., USA
| | - Firoza Mamdani
- Departments of Psychiatry & Human Behavior, University of California, Irvine, Calif., USA
| | - Julie C Lauterborn
- Departments of Anatomy & Neurobiology, University of California, Irvine, Calif., USA
| | - George Kirov
- MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Gary Lynch
- Departments of Psychiatry & Human Behavior, University of California, Irvine, Calif., USA; Departments of Anatomy & Neurobiology, University of California, Irvine, Calif., USA
| | - Christine M Gall
- Departments of Anatomy & Neurobiology, University of California, Irvine, Calif., USA; Departments of Neurobiology & Behavior, University of California, Irvine, Calif., USA
| | - Adolfo Sequeira
- Departments of Psychiatry & Human Behavior, University of California, Irvine, Calif., USA
| | - Marquis P Vawter
- Departments of Psychiatry & Human Behavior, University of California, Irvine, Calif., USA
| |
Collapse
|
43
|
GDH-Dependent Glutamate Oxidation in the Brain Dictates Peripheral Energy Substrate Distribution. Cell Rep 2015; 13:365-75. [PMID: 26440896 DOI: 10.1016/j.celrep.2015.09.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 08/17/2015] [Accepted: 09/01/2015] [Indexed: 12/27/2022] Open
Abstract
Glucose, the main energy substrate used in the CNS, is continuously supplied by the periphery. Glutamate, the major excitatory neurotransmitter, is foreseen as a complementary energy contributor in the brain. In particular, astrocytes actively take up glutamate and may use it through oxidative glutamate dehydrogenase (GDH) activity. Here, we investigated the significance of glutamate as energy substrate for the brain. Upon glutamate exposure, astrocytes generated ATP in a GDH-dependent way. The observed lack of glutamate oxidation in brain-specific GDH null CnsGlud1(-/-) mice resulted in a central energy-deprivation state with increased ADP/ATP ratios and phospho-AMPK in the hypothalamus. This induced changes in the autonomous nervous system balance, with increased sympathetic activity promoting hepatic glucose production and mobilization of substrates reshaping peripheral energy stores. Our data reveal the importance of glutamate as necessary energy substrate for the brain and the role of central GDH in the regulation of whole-body energy homeostasis.
Collapse
|
44
|
van Vuurden DG, Aronica E, Hulleman E, Wedekind LE, Biesmans D, Malekzadeh A, Bugiani M, Geerts D, Noske DP, Vandertop WP, Kaspers GJL, Cloos J, Würdinger T, van der Stoop PPM. Pre-B-cell leukemia homeobox interacting protein 1 is overexpressed in astrocytoma and promotes tumor cell growth and migration. Neuro Oncol 2015; 16:946-59. [PMID: 24470547 DOI: 10.1093/neuonc/not308] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Glial brain tumors cause considerable mortality and morbidity in children and adults. Innovative targets for therapy are needed to improve survival and reduce long-term sequelae. The aim of this study was to find a candidate tumor-promoting protein, abundantly expressed in tumor cells but not in normal brain tissues, as a potential target for therapy. METHODS In silico proteomics and genomics, immunohistochemistry, and immunofluorescence microscopy validation were performed. RNA interference was used to ascertain the functional role of the overexpressed candidate target protein. RESULTS In silico proteomics and genomics revealed pre-B-cell leukemia homeobox (PBX) interacting protein 1 (PBXIP1) overexpression in adult and childhood high-grade glioma and ependymoma compared with normal brain. PBXIP1 is a PBX-family interacting microtubule-binding protein with a putative role in migration and proliferation of cancer cells. Immunohistochemical studies in glial tumors validated PBXIP1 expression in astrocytoma and ependymoma but not in oligodendroglioma. RNAi-mediated PBXIP1-knockdown in glioblastoma cell lines strongly reduced proliferation and migration and induced morphological changes, indicating that PBXIP1 knockdown decreases glioma cell viability and motility through rearrangements of the actin cytoskeleton. Furthermore, expression of PBXIP1 was observed in radial glia and astrocytic progenitor cells in human fetal tissues, suggesting that PBXIP1 is an astroglial progenitor cell marker during human embryonic development. CONCLUSION PBXIP1 is a novel protein overexpressed in astrocytoma and ependymoma, involved in tumor cell proliferation and migration, that warrants further exploration as a novel therapeutic target in these tumors.
Collapse
|
45
|
Monin A, Baumann PS, Griffa A, Xin L, Mekle R, Fournier M, Butticaz C, Klaey M, Cabungcal JH, Steullet P, Ferrari C, Cuenod M, Gruetter R, Thiran JP, Hagmann P, Conus P, Do KQ. Glutathione deficit impairs myelin maturation: relevance for white matter integrity in schizophrenia patients. Mol Psychiatry 2015; 20:827-38. [PMID: 25155877 DOI: 10.1038/mp.2014.88] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/30/2014] [Accepted: 06/23/2014] [Indexed: 12/18/2022]
Abstract
Schizophrenia pathophysiology implies both abnormal redox control and dysconnectivity of the prefrontal cortex, partly related to oligodendrocyte and myelin impairments. As oligodendrocytes are highly vulnerable to altered redox state, we investigated the interplay between glutathione and myelin. In control subjects, multimodal brain imaging revealed a positive association between medial prefrontal glutathione levels and both white matter integrity and resting-state functional connectivity along the cingulum bundle. In early psychosis patients, only white matter integrity was correlated with glutathione levels. On the other side, in the prefrontal cortex of peripubertal mice with genetically impaired glutathione synthesis, mature oligodendrocyte numbers, as well as myelin markers, were decreased. At the molecular levels, under glutathione-deficit conditions induced by short hairpin RNA targeting the key glutathione synthesis enzyme, oligodendrocyte progenitors showed a decreased proliferation mediated by an upregulation of Fyn kinase activity, reversed by either the antioxidant N-acetylcysteine or Fyn kinase inhibitors. In addition, oligodendrocyte maturation was impaired. Interestingly, the regulation of Fyn mRNA and protein expression was also impaired in fibroblasts of patients deficient in glutathione synthesis. Thus, glutathione and redox regulation have a critical role in myelination processes and white matter maturation in the prefrontal cortex of rodent and human, a mechanism potentially disrupted in schizophrenia.
Collapse
Affiliation(s)
- A Monin
- 1] Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland [2] Department of Psychiatry, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland
| | - P S Baumann
- 1] Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland [2] Department of Psychiatry, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland [3] Service of General Psychiatry, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland
| | - A Griffa
- 1] Signal Processing Laboratory, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland [2] Department of Radiology, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Lausanne, Switzerland
| | - L Xin
- Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - R Mekle
- Physikalisch-Technische Bundesanstalt, Berlin, Germany
| | - M Fournier
- 1] Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland [2] Department of Psychiatry, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland
| | - C Butticaz
- 1] Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland [2] Department of Psychiatry, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland
| | - M Klaey
- 1] Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland [2] Department of Psychiatry, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland
| | - J H Cabungcal
- 1] Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland [2] Department of Psychiatry, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland
| | - P Steullet
- 1] Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland [2] Department of Psychiatry, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland
| | - C Ferrari
- 1] Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland [2] Department of Psychiatry, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland
| | - M Cuenod
- 1] Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland [2] Department of Psychiatry, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland
| | - R Gruetter
- 1] Department of Radiology, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Lausanne, Switzerland [2] Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - J P Thiran
- 1] Signal Processing Laboratory, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland [2] Department of Radiology, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Lausanne, Switzerland
| | - P Hagmann
- 1] Signal Processing Laboratory, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland [2] Department of Radiology, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Lausanne, Switzerland
| | - P Conus
- 1] Department of Psychiatry, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland [2] Service of General Psychiatry, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland
| | - K Q Do
- 1] Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland [2] Department of Psychiatry, Centre Hospitalier Universitaire Vaudois and University of Lausanne (CHUV-UNIL), Prilly-Lausanne, Switzerland
| |
Collapse
|
46
|
Raherison ESM, Giguère I, Caron S, Lamara M, MacKay JJ. Modular organization of the white spruce (Picea glauca) transcriptome reveals functional organization and evolutionary signatures. THE NEW PHYTOLOGIST 2015; 207:172-187. [PMID: 25728802 PMCID: PMC5024012 DOI: 10.1111/nph.13343] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 01/18/2015] [Indexed: 05/13/2023]
Abstract
Transcript profiling has shown the molecular bases of several biological processes in plants but few studies have developed an understanding of overall transcriptome variation. We investigated transcriptome structure in white spruce (Picea glauca), aiming to delineate its modular organization and associated functional and evolutionary attributes. Microarray analyses were used to: identify and functionally characterize groups of co-expressed genes; investigate expressional and functional diversity of vascular tissue preferential genes which were conserved among Picea species, and identify expression networks underlying wood formation. We classified 22 857 genes as variable (79%; 22 coexpression groups) or invariant (21%) by profiling across several vegetative tissues. Modular organization and complex transcriptome restructuring among vascular tissue preferential genes was revealed by their assignment to coexpression groups with partially overlapping profiles and partially distinct functions. Integrated analyses of tissue-based and temporally variable profiles identified secondary xylem gene networks, showed their remodelling over a growing season and identified PgNAC-7 (no apical meristerm (NAM), Arabidopsis transcription activation factor (ATAF) and cup-shaped cotyledon (CUC) transcription factor 007 in Picea glauca) as a major hub gene specific to earlywood formation. Reference profiling identified comprehensive, statistically robust coexpressed groups, revealing that modular organization underpins the evolutionary conservation of the transcriptome structure.
Collapse
Affiliation(s)
- Elie S M Raherison
- Center for Forest Research and Institute for Integrative and Systems Biology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Isabelle Giguère
- Center for Forest Research and Institute for Integrative and Systems Biology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Sébastien Caron
- Center for Forest Research and Institute for Integrative and Systems Biology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Mebarek Lamara
- Center for Forest Research and Institute for Integrative and Systems Biology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - John J MacKay
- Department of Plant Sciences, University of Oxford, OX1 3RB, Oxford, UK
| |
Collapse
|
47
|
Sensory Deprivation during Early Postnatal Period Alters the Density of Interneurons in the Mouse Prefrontal Cortex. Neural Plast 2015; 2015:753179. [PMID: 26161272 PMCID: PMC4487934 DOI: 10.1155/2015/753179] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 04/14/2015] [Accepted: 06/04/2015] [Indexed: 11/17/2022] Open
Abstract
Early loss of one sensory system can cause improved function of other sensory systems. However, both the time course and neuronal mechanism of cross-modal plasticity remain elusive. Recent study using functional MRI in humans suggests a role of the prefrontal cortex (PFC) in cross-modal plasticity. Since this phenomenon is assumed to be associated with altered GABAergic inhibition in the PFC, we have tested the hypothesis that early postnatal sensory deprivation causes the changes of inhibitory neuronal circuit in different regions of the PFC of the mice. We determined the effects of sensory deprivation from birth to postnatal day 28 (P28) or P58 on the density of parvalbumin (PV), calbindin (CB), and calretinin (CR) neurons in the prelimbic, infralimbic, and dorsal anterior cingulate cortices. The density of PV and CB neurons was significantly increased in layer 5/6 (L5/6). Moreover, the density of CR neurons was higher in L2/3 in sensory deprived mice compared to intact mice. These changes were more prominent at P56 than at P28. These results suggest that long-term sensory deprivation causes the changes of intracortical inhibitory networks in the PFC and the changes of inhibitory networks in the PFC may contribute to cross-modal plasticity.
Collapse
|
48
|
Hass J, Walton E, Wright C, Beyer A, Scholz M, Turner J, Liu J, Smolka MN, Roessner V, Sponheim SR, Gollub RL, Calhoun VD, Ehrlich S. Associations between DNA methylation and schizophrenia-related intermediate phenotypes - a gene set enrichment analysis. Prog Neuropsychopharmacol Biol Psychiatry 2015; 59:31-39. [PMID: 25598502 PMCID: PMC4346504 DOI: 10.1016/j.pnpbp.2015.01.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 01/06/2015] [Accepted: 01/13/2015] [Indexed: 12/18/2022]
Abstract
Multiple genetic approaches have identified microRNAs as key effectors in psychiatric disorders as they post-transcriptionally regulate expression of thousands of target genes. However, their role in specific psychiatric diseases remains poorly understood. In addition, epigenetic mechanisms such as DNA methylation, which affect the expression of both microRNAs and coding genes, are critical for our understanding of molecular mechanisms in schizophrenia. Using clinical, imaging, genetic, and epigenetic data of 103 patients with schizophrenia and 111 healthy controls of the Mind Clinical Imaging Consortium (MCIC) study of schizophrenia, we conducted gene set enrichment analysis to identify markers for schizophrenia-associated intermediate phenotypes. Genes were ranked based on the correlation between DNA methylation patterns and each phenotype, and then searched for enrichment in 221 predicted microRNA target gene sets. We found the predicted hsa-miR-219a-5p target gene set to be significantly enriched for genes (EPHA4, PKNOX1, ESR1, among others) whose methylation status is correlated with hippocampal volume independent of disease status. Our results were strengthened by significant associations between hsa-miR-219a-5p target gene methylation patterns and hippocampus-related neuropsychological variables. IPA pathway analysis of the respective predicted hsa-miR-219a-5p target genes revealed associated network functions in behavior and developmental disorders. Altered methylation patterns of predicted hsa-miR-219a-5p target genes are associated with a structural aberration of the brain that has been proposed as a possible biomarker for schizophrenia. The (dys)regulation of microRNA target genes by epigenetic mechanisms may confer additional risk for developing psychiatric symptoms. Further study is needed to understand possible interactions between microRNAs and epigenetic changes and their impact on risk for brain-based disorders such as schizophrenia.
Collapse
Affiliation(s)
- Johanna Hass
- Translational Developmental Neuroscience Section, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Esther Walton
- Translational Developmental Neuroscience Section, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Carrie Wright
- Department of Neurosciences, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA,The Mind Research Network, Albuquerque, NM USA
| | - Andreas Beyer
- Cellular Networks and Systems Biology, Biotechnology Center, TU Dresden, Dresden, Germany,University of Cologne, CECAD, Cologne, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany,LIFE (Leipzig Interdisciplinary Research Cluster of Genetic Factors, Phenotypes and Environment), University of Leipzig, Leipzig, Germany
| | - Jessica Turner
- The Mind Research Network, Albuquerque, NM USA,Psychology Department, University of New Mexico, Albuquerque, NM, USA
| | - Jingyu Liu
- The Mind Research Network, Albuquerque, NM USA,Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM USA
| | - Michael N. Smolka
- Department of Psychiatry, Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Veit Roessner
- Translational Developmental Neuroscience Section, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Scott R. Sponheim
- Department of Psychiatry and the Center for magnetic Resonance Research, University of Minnesota, Minneapolis, MN USA
| | - Randy L. Gollub
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA USA,MGH/MIT/HMS Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA USA
| | - Vince D. Calhoun
- The Mind Research Network, Albuquerque, NM USA,Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM USA
| | - Stefan Ehrlich
- Translational Developmental Neuroscience Section, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany; Department of Psychiatry, Massachusetts General Hospital, Boston, MA USA; MGH/MIT/HMS Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA USA.
| |
Collapse
|
49
|
Long LE, Anderson P, Frank E, Shaw A, Liu S, Huang XF, Pinault D, Karl T, O’Brien TJ, Shannon Weickert C, Jones NC. Neuregulin 1 expression and electrophysiological abnormalities in the Neuregulin 1 transmembrane domain heterozygous mutant mouse. PLoS One 2015; 10:e0124114. [PMID: 25992564 PMCID: PMC4437646 DOI: 10.1371/journal.pone.0124114] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 03/10/2015] [Indexed: 01/29/2023] Open
Abstract
Background The Neuregulin 1 transmembrane domain heterozygous mutant (Nrg1 TM HET) mouse is used to investigate the role of Nrg1 in brain function and schizophrenia-like behavioural phenotypes. However, the molecular alterations in brain Nrg1 expression that underpin the behavioural observations have been assumed, but not directly determined. Here we comprehensively characterise mRNA Nrg1 transcripts throughout development of the Nrg1 TM HET mouse. In addition, we investigate the regulation of high-frequency (gamma) electrophysiological oscillations in this mutant mouse to associate molecular changes in Nrg1 with a schizophrenia-relevant neurophysiological profile. Methods Using exonic probes spanning the cysteine-rich, epidermal growth factor (EGF)-like, transmembrane and intracellular domain encoding regions of Nrg1, mRNA levels were measured using qPCR in hippocampus and frontal cortex from male and female Nrg1 TM HET and wild type-like (WT) mice throughout development. We also performed electrophysiological recordings in adult mice and analysed gamma oscillatory at baseline, in responses to auditory stimuli and to ketamine. Results In both hippocampus and cortex, Nrg1 TM HET mice show significantly reduced expression of the exon encoding the transmembrane domain of Nrg1 compared with WT, but unaltered mRNA expression encoding the extracellular bioactive EGF-like and the cysteine-rich (type III) domains, and development-specific and region-specific reductions in the mRNA encoding the intracellular domain. Hippocampal Nrg1 protein expression was not altered, but NMDA receptor NR2B subunit phosphorylation was lower in Nrg1 TM HET mice. We identified elevated ongoing and reduced sensory-evoked gamma power in Nrg1 TM HET mice. Interpretation We found no evidence to support the claim that the Nrg1 TM HET mouse represents a simple haploinsufficient model. Further research is required to explore the possibility that mutation results in a gain of Nrg1 function.
Collapse
Affiliation(s)
- Leonora E. Long
- Schizophrenia Research Institute, Sydney, New South Wales, Australia
- Neuroscience Research Australia, Randwick, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Paul Anderson
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Elisabeth Frank
- Schizophrenia Research Institute, Sydney, New South Wales, Australia
- University of Wollongong, Wollongong, New South Wales, Australia
| | - Alex Shaw
- Schizophrenia Research Institute, Sydney, New South Wales, Australia
- Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - Shijie Liu
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Xu-Feng Huang
- Schizophrenia Research Institute, Sydney, New South Wales, Australia
- University of Wollongong, Wollongong, New South Wales, Australia
| | - Didier Pinault
- INSERM U1114, psychopathologie cognitive et physiopathologie de la schizophrénie, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Tim Karl
- Schizophrenia Research Institute, Sydney, New South Wales, Australia
- Neuroscience Research Australia, Randwick, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Terence J. O’Brien
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Institute, Sydney, New South Wales, Australia
- Neuroscience Research Australia, Randwick, New South Wales, Australia
- School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
- * E-mail: (CSW); (NCJ)
| | - Nigel C. Jones
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
- * E-mail: (CSW); (NCJ)
| |
Collapse
|
50
|
Hiddingh L, Tannous BA, Teng J, Tops B, Jeuken J, Hulleman E, Boots-Sprenger SH, Vandertop WP, Noske DP, Kaspers GJL, Wesseling P, Wurdinger T. EFEMP1 induces γ-secretase/Notch-mediated temozolomide resistance in glioblastoma. Oncotarget 2015; 5:363-74. [PMID: 24495907 PMCID: PMC3964213 DOI: 10.18632/oncotarget.1620] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor. Temozolomide (TMZ) is the standard chemotherapeutic agent for this disease. However, intrinsic and acquired TMZ-resistance represents a major obstacle for this therapy. In order to identify factors involved in TMZ-resistance, we engineered different TMZ-resistant glioblastoma cell lines. Gene expression analysis demonstrated that EFEMP1, an extracellular matrix protein, is associated with TMZ-resistant phenotype. Silencing of EFEMP1 in glioblastoma cells resulted in decreased cell survival following TMZ treatment, whereas overexpression caused TMZ-resistance. EFEMP1 acts via multiple signaling pathways, including γ-secretase-mediated activation of the Notch pathway. We show that inhibition of γ-secretase by RO4929097 causes at least partial sensitization of glioblastoma cells to temozolomide in vitro and in vivo. In addition, we show that EFEMP1 expression levels correlate with survival in TMZ-treated glioblastoma patients. Altogether our results suggest EFEMP1 as a potential therapeutic target to overcome TMZ-resistance in glioblastoma.
Collapse
Affiliation(s)
- Lotte Hiddingh
- Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|