1
|
Todorov H, Weißbach S, Schlichtholz L, Mueller H, Hartwich D, Gerber S, Winter J. Stage-specific expression patterns and co-targeting relationships among miRNAs in the developing mouse cerebral cortex. Commun Biol 2024; 7:1366. [PMID: 39433948 PMCID: PMC11493953 DOI: 10.1038/s42003-024-07092-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/16/2024] [Indexed: 10/23/2024] Open
Abstract
microRNAs are crucial regulators of brain development, however, miRNA regulatory networks are not sufficiently well characterized. By performing small RNA-seq of the mouse embryonic cortex at E14, E17, and P0 as well as in neural progenitor cells and neurons, here we detected clusters of miRNAs that were co-regulated at distinct developmental stages. miRNAs such as miR-92a/b acted as hubs during early, and miR-124 and miR-137 during late neurogenesis. Notably, validated targets of P0 hub miRNAs were enriched for downregulated genes related to stem cell proliferation, negative regulation of neuronal differentiation and RNA splicing, among others, suggesting that miRNAs are particularly important for modulating transcriptional programs of crucial factors that guide the switch to neuronal differentiation. As most genes contain binding sites for more than one miRNA, we furthermore constructed a co-targeting network where numerous miRNAs shared more targets than expected by chance. Using luciferase reporter assays, we demonstrated that simultaneous binding of miRNA pairs to neurodevelopmentally relevant genes exerted an enhanced transcriptional silencing effect compared to single miRNAs. Taken together, we provide a comprehensive resource of miRNA longitudinal expression changes during murine corticogenesis. Furthermore, we highlight several potential mechanisms through which miRNA regulatory networks can shape embryonic brain development.
Collapse
Affiliation(s)
- Hristo Todorov
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stephan Weißbach
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Developmental Biology and Neurobiology (iDN), Johannes Gutenberg University Mainz, Mainz, Germany
| | - Laura Schlichtholz
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Focus Program of Translational Neurosciences, University Medical Center Mainz, Mainz, Germany
| | - Hanna Mueller
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Dewi Hartwich
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Susanne Gerber
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Jennifer Winter
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
2
|
De Vincenti AP, Bonafina A, Ledda F, Paratcha G. Lrig1 regulates cell fate specification of glutamatergic neurons via FGF-driven Jak2/Stat3 signaling in cortical progenitors. Development 2024; 151:dev202879. [PMID: 39250533 DOI: 10.1242/dev.202879] [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: 03/15/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024]
Abstract
The cell-intrinsic mechanisms underlying the decision of a stem/progenitor cell to either proliferate or differentiate remain incompletely understood. Here, we identify the transmembrane protein Lrig1 as a physiological homeostatic regulator of FGF2-driven proliferation and self-renewal of neural progenitors at early-to-mid embryonic stages of cortical development. We show that Lrig1 is expressed in cortical progenitors (CPs), and its ablation caused expansion and increased proliferation of radial/apical progenitors and of neurogenic transit-amplifying Tbr2+ intermediate progenitors. Notably, our findings identify a previously unreported EGF-independent mechanism through which Lrig1 negatively regulates neural progenitor proliferation by modulating the FGF2-induced IL6/Jak2/Stat3 pathway, a molecular cascade that plays a pivotal role in the generation and maintenance of CPs. Consistently, Lrig1 knockout mice showed a significant increase in the density of pyramidal glutamatergic neurons placed in superficial layers 2 and 3 of the postnatal neocortex. Together, these results support a model in which Lrig1 regulates cortical neurogenesis by influencing the cycling activity of a set of progenitors that are temporally specified to produce upper layer glutamatergic neurons.
Collapse
Affiliation(s)
- Ana Paula De Vincenti
- Laboratorio de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina. Universidad de Buenos Aires (UBA), Buenos Aires CP1121, Argentina
| | - Antonela Bonafina
- Laboratorio de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina. Universidad de Buenos Aires (UBA), Buenos Aires CP1121, Argentina
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires, Buenos Aires C1405 BWE, Argentina
| | - Fernanda Ledda
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires, Buenos Aires C1405 BWE, Argentina
| | - Gustavo Paratcha
- Laboratorio de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina. Universidad de Buenos Aires (UBA), Buenos Aires CP1121, Argentina
| |
Collapse
|
3
|
Ma Q, Chen G, Li Y, Guo Z, Zhang X. The molecular genetics of PI3K/PTEN/AKT/mTOR pathway in the malformations of cortical development. Genes Dis 2024; 11:101021. [PMID: 39006182 PMCID: PMC11245990 DOI: 10.1016/j.gendis.2023.04.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/07/2023] [Accepted: 04/30/2023] [Indexed: 07/16/2024] Open
Abstract
Malformations of cortical development (MCD) are a group of developmental disorders characterized by abnormal cortical structures caused by genetic or harmful environmental factors. Many kinds of MCD are caused by genetic variation. MCD is the common cause of intellectual disability and intractable epilepsy. With rapid advances in imaging and sequencing technologies, the diagnostic rate of MCD has been increasing, and many potential genes causing MCD have been successively identified. However, the high genetic heterogeneity of MCD makes it challenging to understand the molecular pathogenesis of MCD and to identify effective targeted drugs. Thus, in this review, we outline important events of cortical development. Then we illustrate the progress of molecular genetic studies about MCD focusing on the PI3K/PTEN/AKT/mTOR pathway. Finally, we briefly discuss the diagnostic methods, disease models, and therapeutic strategies for MCD. The information will facilitate further research on MCD. Understanding the role of the PI3K/PTEN/AKT/mTOR pathway in MCD could lead to a novel strategy for treating MCD-related diseases.
Collapse
Affiliation(s)
- Qing Ma
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Guang Chen
- Department of Urology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Ying Li
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, Heilongjiang 150000, China
- Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Zhenming Guo
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Xue Zhang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, Heilongjiang 150000, China
| |
Collapse
|
4
|
Yang J, Li Y, Tang Y, Yang L, Guo C, Peng C. Spatial transcriptome reveals the region-specific genes and pathways regulated by Satb2 in neocortical development. BMC Genomics 2024; 25:757. [PMID: 39095712 PMCID: PMC11297773 DOI: 10.1186/s12864-024-10672-w] [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: 04/15/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND It is known that the neurodevelopmental disorder associated gene, Satb2, plays important roles in determining the upper layer neuron specification. However, it is not well known how this gene regulates other neocortical regions during the development. It is also lack of comprehensive delineation of its spatially regulatory pathways in neocortical development. RESULTS In this work, we utilized spatial transcriptomics and immuno-staining to systematically investigate the region-specific gene regulation of Satb2 by comparing the Satb2+/+ and Satb2-/- mice at embryonic stages, including the ventricle zone (VZ) or subventricle zone (SVZ), intermediate zone (IZ) and cortical plate (CP) respectively. The staining result reveals that these three regions become moderately or significantly thinner in the Satb2-/- mice. In the cellular level, the cell number increases in the VZ/SVZ, whereas the cell number decreases in the CP. The spatial transcriptomics data show that many important genes and relevant pathways are dysregulated in Satb2-/- mice in a region-specific manner. In the VZ/SVZ, the key genes involved in neural precursor cell proliferation, including the intermediate progenitor marker Tbr2 and the lactate production related gene Ldha, are up-regulated in Satb2-/- mice. In the IZ, the key genes in regulating neuronal differentiation and migration, such as Rnd2, exhibit ectopic expressions in the Satb2-/- mice. In the CP, the lineage-specific genes, Tbr1 and Bcl11b, are abnormally expressed. The neuropeptide related gene Npy is down-regulated in Satb2-/- mice. Finally, we validated the abnormal expressions of key regulators by using immunofluorescence or qPCR. CONCLUSIONS In summary, our work provides insights on the region-specific genes and pathways which are regulated by Satb2 in neocortical development.
Collapse
Affiliation(s)
- Jianfen Yang
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Yu Li
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Yiyuli Tang
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Ling Yang
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Chunming Guo
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Cheng Peng
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China.
| |
Collapse
|
5
|
Ochi S, Manabe S, Kikkawa T, Ebrahimiazar S, Kimura R, Yoshizaki K, Osumi N. A Transcriptomic Dataset of Embryonic Murine Telencephalon. Sci Data 2024; 11:586. [PMID: 38839806 PMCID: PMC11153524 DOI: 10.1038/s41597-024-03421-x] [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: 09/28/2023] [Accepted: 05/24/2024] [Indexed: 06/07/2024] Open
Abstract
Sex bias is known in the prevalence/pathology of neurodevelopmental disorders. Sex-dependent differences of the certain brain areas are known to emerge perinatally through the exposure to sex hormones, while gene expression patterns in the rodent embryonic brain does not seem to be completely the same between male and female. To investigate potential sex differences in gene expression and cortical organization during the embryonic period in mice, we conducted a comprehensive analysis of gene expression for the telencephalon at embryonic day (E) 11.5 (a peak of neural stem cell expansion) and E14.5 (a peak of neurogenesis) using bulk RNA-seq data. As a result, our data showed the existence of notable sex differences in gene expression patterns not obviously at E11.5, but clearly at E14.5 when neurogenesis has become its peak. These data can be useful for exploring potential contribution of genes exhibiting sex differences to the divergence in brain development. Additionally, our data underscore the significance of studying the embryonic period to gain a deeper understanding of sex differences in brain development.
Collapse
Affiliation(s)
- Shohei Ochi
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Shyu Manabe
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Takako Kikkawa
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Sara Ebrahimiazar
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Ryuichi Kimura
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Kaichi Yoshizaki
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
- Kobe University Graduate School of Medicine, Department of Future Medical Sciences, Division of Integrated Analysis of Bioresource and Health Care, Kobe, 650-0047, Japan
- Kobe University Hospital, Bioresource Center, Kobe, 650-0047, Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.
| |
Collapse
|
6
|
Taub DG, Jiang Q, Pietrafesa F, Su J, Carroll A, Greene C, Blanchard MR, Jain A, El-Rifai M, Callen A, Yager K, Chung C, He Z, Chen C, Woolf CJ. The secondary somatosensory cortex gates mechanical and heat sensitivity. Nat Commun 2024; 15:1289. [PMID: 38346995 PMCID: PMC10861531 DOI: 10.1038/s41467-024-45729-7] [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: 06/13/2023] [Accepted: 02/01/2024] [Indexed: 02/15/2024] Open
Abstract
The cerebral cortex is vital for the processing and perception of sensory stimuli. In the somatosensory axis, information is received primarily by two distinct regions, the primary (S1) and secondary (S2) somatosensory cortices. Top-down circuits stemming from S1 can modulate mechanical and cooling but not heat stimuli such that circuit inhibition causes blunted perception. This suggests that responsiveness to particular somatosensory stimuli occurs in a modality specific fashion and we sought to determine additional cortical substrates. In this work, we identify in a mouse model that inhibition of S2 output increases mechanical and heat, but not cooling sensitivity, in contrast to S1. Combining 2-photon anatomical reconstruction with chemogenetic inhibition of specific S2 circuits, we discover that S2 projections to the secondary motor cortex (M2) govern mechanical and heat sensitivity without affecting motor performance or anxiety. Taken together, we show that S2 is an essential cortical structure that governs mechanical and heat sensitivity.
Collapse
Affiliation(s)
- Daniel G Taub
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Qiufen Jiang
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Francesca Pietrafesa
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Junfeng Su
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Aloe Carroll
- College of Sciences, Northeastern University, Boston, MA, USA
| | - Caitlin Greene
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | | | - Aakanksha Jain
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Mahmoud El-Rifai
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Alexis Callen
- Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA, USA
| | - Katherine Yager
- Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA, USA
| | - Clara Chung
- Department of Neuroscience, Boston University, Boston, MA, USA
| | - Zhigang He
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Chinfei Chen
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Clifford J Woolf
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA.
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Mukhtar T, Taylor V. Dynamic transcriptional programs define distinct mammalian cortical lineages. Neural Regen Res 2024; 19:387-389. [PMID: 37488895 PMCID: PMC10503623 DOI: 10.4103/1673-5374.377589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/22/2023] [Accepted: 04/19/2023] [Indexed: 07/26/2023] Open
Affiliation(s)
- Tanzila Mukhtar
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
8
|
Taub DG, Jiang Q, Pietrafesa F, Su J, Greene C, Blanchard MR, Jain A, El-Rifai M, Callen A, Yager K, Chung C, He Z, Chen C, Woolf CJ. The Secondary Somatosensory Cortex Gates Mechanical and Thermal Sensitivity. RESEARCH SQUARE 2023:rs.3.rs-2976953. [PMID: 37461707 PMCID: PMC10350168 DOI: 10.21203/rs.3.rs-2976953/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
The cerebral cortex is vital for the perception and processing of sensory stimuli. In the somatosensory axis, information is received by two distinct regions, the primary (S1) and secondary (S2) somatosensory cortices. Top-down circuits stemming from S1 can modulate mechanical and cooling but not heat stimuli such that circuit inhibition causes blunted mechanical and cooling perception. Using optogenetics and chemogenetics, we find that in contrast to S1, an inhibition of S2 output increases mechanical and heat, but not cooling sensitivity. Combining 2-photon anatomical reconstruction with chemogenetic inhibition of specific S2 circuits, we discover that S2 projections to the secondary motor cortex (M2) govern mechanical and thermal sensitivity without affecting motor or cognitive function. This suggests that while S2, like S1, encodes specific sensory information, that S2 operates through quite distinct neural substrates to modulate responsiveness to particular somatosensory stimuli and that somatosensory cortical encoding occurs in a largely parallel fashion.
Collapse
Affiliation(s)
- Daniel G. Taub
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Qiufen Jiang
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Francesca Pietrafesa
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Junfeng Su
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Caitlin Greene
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | | | - Aakanksha Jain
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Mahmoud El-Rifai
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Alexis Callen
- Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA, USA
| | - Katherine Yager
- Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA, USA
| | - Clara Chung
- Department of Neuroscience, Boston University, Boston, MA, USA
| | - Zhigang He
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Chinfei Chen
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Clifford J. Woolf
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Taub DG, Jiang Q, Pietrafesa F, Su J, Greene C, Blanchard MR, Jain A, El-Rifai M, Callen A, Yager K, Chung C, He Z, Chen C, Woolf CJ. The Secondary Somatosensory Cortex Gates Mechanical and Thermal Sensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.19.541449. [PMID: 37293011 PMCID: PMC10245795 DOI: 10.1101/2023.05.19.541449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The cerebral cortex is vital for the perception and processing of sensory stimuli. In the somatosensory axis, information is received by two distinct regions, the primary (S1) and secondary (S2) somatosensory cortices. Top-down circuits stemming from S1 can modulate mechanical and cooling but not heat stimuli such that circuit inhibition causes blunted mechanical and cooling perception. Using optogenetics and chemogenetics, we find that in contrast to S1, an inhibition of S2 output increases mechanical and heat, but not cooling sensitivity. Combining 2-photon anatomical reconstruction with chemogenetic inhibition of specific S2 circuits, we discover that S2 projections to the secondary motor cortex (M2) govern mechanical and thermal sensitivity without affecting motor or cognitive function. This suggests that while S2, like S1, encodes specific sensory information, that S2 operates through quite distinct neural substrates to modulate responsiveness to particular somatosensory stimuli and that somatosensory cortical encoding occurs in a largely parallel fashion.
Collapse
|
10
|
Goo BS, Mun DJ, Kim S, Nhung TTM, Lee SB, Woo Y, Kim SJ, Suh BK, Park SJ, Lee HE, Park K, Jang H, Rah JC, Yoon KJ, Baek ST, Park SY, Park SK. Schizophrenia-associated Mitotic Arrest Deficient-1 (MAD1) regulates the polarity of migrating neurons in the developing neocortex. Mol Psychiatry 2023; 28:856-870. [PMID: 36357673 PMCID: PMC9908555 DOI: 10.1038/s41380-022-01856-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
Abstract
Although large-scale genome-wide association studies (GWAS) have identified an association between MAD1L1 (Mitotic Arrest Deficient-1 Like 1) and the pathology of schizophrenia, the molecular mechanisms underlying this association remain unclear. In the present study, we aimed to address these mechanisms by examining the role of MAD1 (the gene product of MAD1L1) in key neurodevelopmental processes in mice and human organoids. Our findings indicated that MAD1 is highly expressed during active cortical development and that MAD1 deficiency leads to impairments in neuronal migration and neurite outgrowth. We also observed that MAD1 is localized to the Golgi apparatus and regulates vesicular trafficking from the Golgi apparatus to the plasma membrane, which is required for the growth and polarity of migrating neurons. In this process, MAD1 physically interacts and collaborates with the kinesin-like protein KIFC3 (kinesin family member C3) to regulate the morphology of the Golgi apparatus and neuronal polarity, thereby ensuring proper neuronal migration and differentiation. Consequently, our findings indicate that MAD1 is an essential regulator of neuronal development and that alterations in MAD1 may underlie schizophrenia pathobiology.
Collapse
Affiliation(s)
- Bon Seong Goo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Dong Jin Mun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Seunghyun Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Truong Thi My Nhung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Su Been Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Youngsik Woo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Soo Jeong Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Bo Kyoung Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Sung Jin Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Hee-Eun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Kunyou Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Hyunsoo Jang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jong-Cheol Rah
- Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Seung Tae Baek
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Seung-Yeol Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| |
Collapse
|
11
|
Tominaga K, Sakashita E, Kasashima K, Kuroiwa K, Nagao Y, Iwamori N, Endo H. Tip60/KAT5 Histone Acetyltransferase Is Required for Maintenance and Neurogenesis of Embryonic Neural Stem Cells. Int J Mol Sci 2023; 24:ijms24032113. [PMID: 36768434 PMCID: PMC9916716 DOI: 10.3390/ijms24032113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Epigenetic regulation via epigenetic factors in collaboration with tissue-specific transcription factors is curtail for establishing functional organ systems during development. Brain development is tightly regulated by epigenetic factors, which are coordinately activated or inactivated during processes, and their dysregulation is linked to brain abnormalities and intellectual disability. However, the precise mechanism of epigenetic regulation in brain development and neurogenesis remains largely unknown. Here, we show that Tip60/KAT5 deletion in neural stem/progenitor cells (NSCs) in mice results in multiple abnormalities of brain development. Tip60-deficient embryonic brain led to microcephaly, and proliferating cells in the developing brain were reduced by Tip60 deficiency. In addition, neural differentiation and neuronal migration were severely affected in Tip60-deficient brains. Following neurogenesis in developing brains, gliogenesis started from the earlier stage of development in Tip60-deficient brains, indicating that Tip60 is involved in switching from neurogenesis to gliogenesis during brain development. It was also confirmed in vitro that poor neurosphere formation, proliferation defects, neural differentiation defects, and accelerated astrocytic differentiation in mutant NSCs are derived from Tip60-deficient embryonic brains. This study uncovers the critical role of Tip60 in brain development and NSC maintenance and function in vivo and in vitro.
Collapse
Affiliation(s)
- Kaoru Tominaga
- Division of Structural Biochemistry, Department of Biochemistry, Jichi Medical University, Tochigi 321-0498, Japan
- Division of Functional Biochemistry, Department of Biochemistry, Jichi Medical University, Tochigi 321-0498, Japan
- Correspondence: (K.T.); (N.I.)
| | - Eiji Sakashita
- Division of Functional Biochemistry, Department of Biochemistry, Jichi Medical University, Tochigi 321-0498, Japan
| | - Katsumi Kasashima
- Division of Functional Biochemistry, Department of Biochemistry, Jichi Medical University, Tochigi 321-0498, Japan
| | - Kenji Kuroiwa
- Division of Functional Biochemistry, Department of Biochemistry, Jichi Medical University, Tochigi 321-0498, Japan
| | - Yasumitsu Nagao
- Center for Experimental Medicine, Jichi Medical University, Tochigi 321-0498, Japan
| | - Naoki Iwamori
- Department of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
- Correspondence: (K.T.); (N.I.)
| | - Hitoshi Endo
- Division of Functional Biochemistry, Department of Biochemistry, Jichi Medical University, Tochigi 321-0498, Japan
| |
Collapse
|
12
|
Pärn A, Olsen D, Tuvikene J, Kaas M, Borisova E, Bilgin M, Elhauge M, Vilstrup J, Madsen P, Ambrozkiewicz MC, Goz RU, Timmusk T, Tarabykin V, Gustafsen C, Glerup S. PCSK9 deficiency alters brain lipid composition without affecting brain development and function. Front Mol Neurosci 2023; 15:1084633. [PMID: 36733269 PMCID: PMC9887304 DOI: 10.3389/fnmol.2022.1084633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/16/2022] [Indexed: 01/18/2023] Open
Abstract
PCSK9 induces lysosomal degradation of the low-density lipoprotein (LDL) receptor (LDLR) in the liver, hereby preventing removal of LDL cholesterol from the circulation. Accordingly, PCSK9 inhibitory antibodies and siRNA potently reduce LDL cholesterol to unprecedented low levels and are approved for treatment of hypercholesterolemia. In addition, PCSK9 inactivation alters the levels of several other circulating lipid classes and species. Brain function is critically influenced by cholesterol and lipid composition. However, it remains unclear how the brain is affected long-term by the reduction in circulating lipids as achieved with potent lipid lowering therapeutics such as PCSK9 inhibitors. Furthermore, it is unknown if locally expressed PCSK9 affects neuronal circuits through regulation of receptor levels. We have studied the effect of lifelong low peripheral cholesterol levels on brain lipid composition and behavior in adult PCSK9 KO mice. In addition, we studied the effect of PCSK9 on neurons in culture and in vivo in the developing cerebral cortex. We found that PCSK9 reduced LDLR and neurite complexity in cultured neurons, but neither PCSK9 KO nor overexpression affected cortical development in vivo. Interestingly, PCSK9 deficiency resulted in changes of several lipid classes in the adult cortex and cerebellum. Despite the observed changes, PCSK9 KO mice had unchanged behavior compared to WT controls. In conclusion, our findings demonstrate that altered PCSK9 levels do not compromise brain development or function in mice, and are in line with clinical trials showing that PCSK9 inhibitors have no adverse effects on cognitive function.
Collapse
Affiliation(s)
- Angela Pärn
- Department of Biomedicine, Aarhus University, Aarhus, Denmark,*Correspondence: Angela Pärn, ✉
| | - Ditte Olsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jürgen Tuvikene
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia,Protobios LLC, Tallinn, Estonia
| | - Mathias Kaas
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ekaterina Borisova
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Berlin, Germany,Tomsk National Research Medical Center of the Russian Academy of Sciences, Research Institute of Medical Genetics, Tomsk, Russia
| | - Mesut Bilgin
- Danish Cancer Society Research Center, Lipidomics Core Facility, Copenhagen, Denmark
| | - Mie Elhauge
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Joachim Vilstrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark,Draupnir Bio ApS, INCUBA Skejby, Aarhus, Denmark
| | - Peder Madsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark,Draupnir Bio ApS, INCUBA Skejby, Aarhus, Denmark
| | - Mateusz C. Ambrozkiewicz
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Roman U. Goz
- Department of Neurobiology, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Tõnis Timmusk
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia,Protobios LLC, Tallinn, Estonia
| | - Victor Tarabykin
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Berlin, Germany,Tomsk National Research Medical Center of the Russian Academy of Sciences, Research Institute of Medical Genetics, Tomsk, Russia
| | - Camilla Gustafsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark,Draupnir Bio ApS, INCUBA Skejby, Aarhus, Denmark,Camilla Gustafsen, ✉
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark,Draupnir Bio ApS, INCUBA Skejby, Aarhus, Denmark,Simon Glerup, ✉
| |
Collapse
|
13
|
Degl’Innocenti E, Dell’Anno MT. Human and mouse cortical astrocytes: a comparative view from development to morphological and functional characterization. Front Neuroanat 2023; 17:1130729. [PMID: 37139179 PMCID: PMC10150887 DOI: 10.3389/fnana.2023.1130729] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
The vision of astroglia as a bare scaffold to neuronal circuitry has been largely overturned. Astrocytes exert a neurotrophic function, but also take active part in supporting synaptic transmission and in calibrating blood circulation. Many aspects of their functioning have been unveiled from studies conducted in murine models, however evidence is showing many differences between mouse and human astrocytes starting from their development and encompassing morphological, transcriptomic and physiological variations when they achieve complete maturation. The evolutionary race toward superior cognitive abilities unique to humans has drastically impacted neocortex structure and, together with neuronal circuitry, astrocytes have also been affected with the acquisition of species-specific properties. In this review, we summarize diversities between murine and human astroglia, with a specific focus on neocortex, in a panoramic view that starts with their developmental origin to include all structural and molecular differences that mark the uniqueness of human astrocytes.
Collapse
Affiliation(s)
- Elisa Degl’Innocenti
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Maria Teresa Dell’Anno
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
- *Correspondence: Maria Teresa Dell’Anno,
| |
Collapse
|
14
|
Mukhtar T, Breda J, Adam MA, Boareto M, Grobecker P, Karimaddini Z, Grison A, Eschbach K, Chandrasekhar R, Vermeul S, Okoniewski M, Pachkov M, Harwell CC, Atanasoski S, Beisel C, Iber D, van Nimwegen E, Taylor V. Temporal and sequential transcriptional dynamics define lineage shifts in corticogenesis. EMBO J 2022; 41:e111132. [PMID: 36345783 PMCID: PMC9753470 DOI: 10.15252/embj.2022111132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 09/09/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022] Open
Abstract
The cerebral cortex contains billions of neurons, and their disorganization or misspecification leads to neurodevelopmental disorders. Understanding how the plethora of projection neuron subtypes are generated by cortical neural stem cells (NSCs) is a major challenge. Here, we focused on elucidating the transcriptional landscape of murine embryonic NSCs, basal progenitors (BPs), and newborn neurons (NBNs) throughout cortical development. We uncover dynamic shifts in transcriptional space over time and heterogeneity within each progenitor population. We identified signature hallmarks of NSC, BP, and NBN clusters and predict active transcriptional nodes and networks that contribute to neural fate specification. We find that the expression of receptors, ligands, and downstream pathway components is highly dynamic over time and throughout the lineage implying differential responsiveness to signals. Thus, we provide an expansive compendium of gene expression during cortical development that will be an invaluable resource for studying neural developmental processes and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Tanzila Mukhtar
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | - Jeremie Breda
- BiozentrumUniversity of BaselBaselSwitzerland
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
| | - Manal A Adam
- Eli and Edythe Broad Center of Regeneration Medicine and Stem cell ResearchUniversity of California, San FranciscoSan FranciscoCAUSA
- Weill Institute for NeuroscienceSan FranciscoCAUSA
- Department of NeurologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Marcelo Boareto
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
- Computational Biology Group, D‐BSSEETH ZürichBaselSwitzerland
| | - Pascal Grobecker
- BiozentrumUniversity of BaselBaselSwitzerland
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
| | - Zahra Karimaddini
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
- Computational Biology Group, D‐BSSEETH ZürichBaselSwitzerland
| | - Alice Grison
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | - Katja Eschbach
- Department of Biosystems Science and EngineeringETH ZürichBaselSwitzerland
| | | | - Swen Vermeul
- Scientific IT ServicesETH ZürichZürichSwitzerland
| | | | - Mikhail Pachkov
- BiozentrumUniversity of BaselBaselSwitzerland
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
| | - Corey C Harwell
- Eli and Edythe Broad Center of Regeneration Medicine and Stem cell ResearchUniversity of California, San FranciscoSan FranciscoCAUSA
- Weill Institute for NeuroscienceSan FranciscoCAUSA
- Department of NeurologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Suzana Atanasoski
- Department of BiomedicineUniversity of BaselBaselSwitzerland
- Faculty of MedicineUniversity of ZürichZürichSwitzerland
| | - Christian Beisel
- Department of Biosystems Science and EngineeringETH ZürichBaselSwitzerland
| | - Dagmar Iber
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
- Weill Institute for NeuroscienceSan FranciscoCAUSA
| | - Erik van Nimwegen
- BiozentrumUniversity of BaselBaselSwitzerland
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
| | - Verdon Taylor
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| |
Collapse
|
15
|
Griffin A, Mahesh A, Tiwari VK. Disruption of the gene regulatory programme in neurodevelopmental disorders. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194860. [PMID: 36007842 DOI: 10.1016/j.bbagrm.2022.194860] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 06/15/2023]
Abstract
Cortical development consists of a series of synchronised events, including fate transition of cortical progenitors, neuronal migration, specification and connectivity. It is becoming clear that gene expression programs governing these events rely on the interplay between signalling molecules, transcription factors and epigenetic mechanisms. When genetic or environmental factors disrupt expression of genes involved in important brain development processes, neurodevelopmental disorders can occur. This review aims to highlight how recent advances in technologies have helped uncover and imitate the gene regulatory mechanisms commonly disrupted in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Aoife Griffin
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queens University, Belfast BT9 7BL, United Kingdom
| | - Arun Mahesh
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queens University, Belfast BT9 7BL, United Kingdom
| | - Vijay K Tiwari
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queens University, Belfast BT9 7BL, United Kingdom.
| |
Collapse
|
16
|
Park J, Lee K, Kim K, Yi SJ. The role of histone modifications: from neurodevelopment to neurodiseases. Signal Transduct Target Ther 2022; 7:217. [PMID: 35794091 PMCID: PMC9259618 DOI: 10.1038/s41392-022-01078-9] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/11/2022] [Accepted: 06/21/2022] [Indexed: 12/24/2022] Open
Abstract
Epigenetic regulatory mechanisms, including DNA methylation, histone modification, chromatin remodeling, and microRNA expression, play critical roles in cell differentiation and organ development through spatial and temporal gene regulation. Neurogenesis is a sophisticated and complex process by which neural stem cells differentiate into specialized brain cell types at specific times and regions of the brain. A growing body of evidence suggests that epigenetic mechanisms, such as histone modifications, allow the fine-tuning and coordination of spatiotemporal gene expressions during neurogenesis. Aberrant histone modifications contribute to the development of neurodegenerative and neuropsychiatric diseases. Herein, recent progress in understanding histone modifications in regulating embryonic and adult neurogenesis is comprehensively reviewed. The histone modifications implicated in neurodegenerative and neuropsychiatric diseases are also covered, and future directions in this area are provided.
Collapse
Affiliation(s)
- Jisu Park
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Kyubin Lee
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Kyunghwan Kim
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| | - Sun-Ju Yi
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| |
Collapse
|
17
|
Iwasawa E, Brown FN, Shula C, Kahn F, Lee SH, Berta T, Ladle DR, Campbell K, Mangano FT, Goto J. The Anti-Inflammatory Agent Bindarit Attenuates the Impairment of Neural Development through Suppression of Microglial Activation in a Neonatal Hydrocephalus Mouse Model. J Neurosci 2022; 42:1820-1844. [PMID: 34992132 PMCID: PMC8896558 DOI: 10.1523/jneurosci.1160-21.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 11/30/2021] [Accepted: 12/17/2021] [Indexed: 11/21/2022] Open
Abstract
Neonatal hydrocephalus presents with various degrees of neuroinflammation and long-term neurologic deficits in surgically treated patients, provoking a need for additional medical treatment. We previously reported elevated neuroinflammation and severe periventricular white matter damage in the progressive hydrocephalus (prh) mutant which contains a point mutation in the Ccdc39 gene, causing loss of cilia-mediated unidirectional CSF flow. In this study, we identified cortical neuropil maturation defects such as impaired excitatory synapse maturation and loss of homeostatic microglia, and swimming locomotor defects in early postnatal prh mutant mice. Strikingly, systemic application of the anti-inflammatory small molecule bindarit significantly supports healthy postnatal cerebral cortical development in the prh mutant. While bindarit only mildly reduced the ventricular volume, it significantly improved the edematous appearance and myelination of the corpus callosum. Moreover, the treatment attenuated thinning in cortical Layers II-IV, excitatory synapse formation, and interneuron morphogenesis, by supporting the ramified-shaped homeostatic microglia from excessive cell death. Also, the therapeutic effect led to the alleviation of a spastic locomotor phenotype of the mutant. We found that microglia, but not peripheral monocytes, contribute to amoeboid-shaped activated myeloid cells in prh mutants' corpus callosum and the proinflammatory cytokines expression. Bindarit blocks nuclear factor (NF)-kB activation and its downstream proinflammatory cytokines, including monocyte chemoattractant protein-1, in the prh mutant. Collectively, we revealed that amelioration of neuroinflammation is crucial for white matter and neuronal maturation in neonatal hydrocephalus. Future studies of bindarit treatment combined with CSF diversion surgery may provide long-term benefits supporting neuronal development in neonatal hydrocephalus.SIGNIFICANCE STATEMENT In neonatal hydrocephalus, little is known about the signaling cascades of neuroinflammation or the impact of such inflammatory insults on neural cell development within the perinatal cerebral cortex. Here, we report that proinflammatory activation of myeloid cells, the majority of which are derived from microglia, impairs periventricular myelination and cortical neuronal maturation using the mouse prh genetic model of neonatal hydrocephalus. Administration of bindarit, an anti-inflammatory small molecule that blocks nuclear factor (NF)-kB activation, restored the cortical thinning and synaptic maturation defects in the prh mutant brain through suppression of microglial activation. These data indicate the potential therapeutic use of anti-inflammatory reagents targeting neuroinflammation in the treatment of neonatal hydrocephalus.
Collapse
Affiliation(s)
- Eri Iwasawa
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45242
| | - Farrah N Brown
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45242
| | - Crystal Shula
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45242
| | - Fatima Kahn
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45242
| | - Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, 45242
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, 45242
| | - David R Ladle
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, Ohio, 45435
| | - Kenneth Campbell
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45242
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45242
| | - Francesco T Mangano
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45242
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45242
| | - June Goto
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45242
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45242
| |
Collapse
|
18
|
New Strategies for the Treatment of Neuropsychiatric Disorders Based on Reelin Dysfunction. Int J Mol Sci 2022; 23:ijms23031829. [PMID: 35163751 PMCID: PMC8836358 DOI: 10.3390/ijms23031829] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 12/16/2022] Open
Abstract
Reelin is an extracellular matrix protein that is mainly produced in Cajal-Retzius cells and controls neuronal migration, which is important for the proper formation of cortical layers in the developmental stage of the brain. In the adult brain, Reelin plays a crucial role in the regulation of N-methyl-D-aspartate receptor-dependent synaptic function, and its expression decreases postnatally. Clinical studies showed reductions in Reelin protein and mRNA expression levels in patients with psychiatric disorders; however, the causal relationship remains unclear. Reelin-deficient mice exhibit an abnormal neuronal morphology and behavior, while Reelin supplementation ameliorates learning deficits, synaptic dysfunctions, and spine loss in animal models with Reelin deficiency. These findings suggest that the neuronal deficits and brain dysfunctions associated with the down-regulated expression of Reelin are attenuated by enhancements in its expression and functions in the brain. In this review, we summarize findings on the role of Reelin in neuropsychiatric disorders and discuss potential therapeutic approaches for neuropsychiatric disorders associated with Reelin dysfunctions.
Collapse
|
19
|
Yildiz CB, Zimmer-Bensch G. Role of DNMTs in the Brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1389:363-394. [DOI: 10.1007/978-3-031-11454-0_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
20
|
Reichard J, Zimmer-Bensch G. The Epigenome in Neurodevelopmental Disorders. Front Neurosci 2021; 15:776809. [PMID: 34803599 PMCID: PMC8595945 DOI: 10.3389/fnins.2021.776809] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/04/2021] [Indexed: 12/26/2022] Open
Abstract
Neurodevelopmental diseases (NDDs), such as autism spectrum disorders, epilepsy, and schizophrenia, are characterized by diverse facets of neurological and psychiatric symptoms, differing in etiology, onset and severity. Such symptoms include mental delay, cognitive and language impairments, or restrictions to adaptive and social behavior. Nevertheless, all have in common that critical milestones of brain development are disrupted, leading to functional deficits of the central nervous system and clinical manifestation in child- or adulthood. To approach how the different development-associated neuropathologies can occur and which risk factors or critical processes are involved in provoking higher susceptibility for such diseases, a detailed understanding of the mechanisms underlying proper brain formation is required. NDDs rely on deficits in neuronal identity, proportion or function, whereby a defective development of the cerebral cortex, the seat of higher cognitive functions, is implicated in numerous disorders. Such deficits can be provoked by genetic and environmental factors during corticogenesis. Thereby, epigenetic mechanisms can act as an interface between external stimuli and the genome, since they are known to be responsive to external stimuli also in cortical neurons. In line with that, DNA methylation, histone modifications/variants, ATP-dependent chromatin remodeling, as well as regulatory non-coding RNAs regulate diverse aspects of neuronal development, and alterations in epigenomic marks have been associated with NDDs of varying phenotypes. Here, we provide an overview of essential steps of mammalian corticogenesis, and discuss the role of epigenetic mechanisms assumed to contribute to pathophysiological aspects of NDDs, when being disrupted.
Collapse
Affiliation(s)
- Julia Reichard
- Functional Epigenetics in the Animal Model, Institute for Biology II, RWTH Aachen University, Aachen, Germany
- Research Training Group 2416 MultiSenses-MultiScales, Institute for Biology II, RWTH Aachen University, Aachen, Germany
| | - Geraldine Zimmer-Bensch
- Functional Epigenetics in the Animal Model, Institute for Biology II, RWTH Aachen University, Aachen, Germany
- Research Training Group 2416 MultiSenses-MultiScales, Institute for Biology II, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
21
|
Islam KUS, Meli N, Blaess S. The Development of the Mesoprefrontal Dopaminergic System in Health and Disease. Front Neural Circuits 2021; 15:746582. [PMID: 34712123 PMCID: PMC8546303 DOI: 10.3389/fncir.2021.746582] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/10/2021] [Indexed: 12/18/2022] Open
Abstract
Midbrain dopaminergic neurons located in the substantia nigra and the ventral tegmental area are the main source of dopamine in the brain. They send out projections to a variety of forebrain structures, including dorsal striatum, nucleus accumbens, and prefrontal cortex (PFC), establishing the nigrostriatal, mesolimbic, and mesoprefrontal pathways, respectively. The dopaminergic input to the PFC is essential for the performance of higher cognitive functions such as working memory, attention, planning, and decision making. The gradual maturation of these cognitive skills during postnatal development correlates with the maturation of PFC local circuits, which undergo a lengthy functional remodeling process during the neonatal and adolescence stage. During this period, the mesoprefrontal dopaminergic innervation also matures: the fibers are rather sparse at prenatal stages and slowly increase in density during postnatal development to finally reach a stable pattern in early adulthood. Despite the prominent role of dopamine in the regulation of PFC function, relatively little is known about how the dopaminergic innervation is established in the PFC, whether and how it influences the maturation of local circuits and how exactly it facilitates cognitive functions in the PFC. In this review, we provide an overview of the development of the mesoprefrontal dopaminergic system in rodents and primates and discuss the role of altered dopaminergic signaling in neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- K Ushna S Islam
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Norisa Meli
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany.,Institute of Neuropathology, Section for Translational Epilepsy Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
22
|
King EL, Irigoyen N. Zika Virus and Neuropathogenesis: The Unanswered Question of Which Strain Is More Prone to Causing Microcephaly and Other Neurological Defects. Front Cell Neurosci 2021; 15:695106. [PMID: 34658789 PMCID: PMC8514627 DOI: 10.3389/fncel.2021.695106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/25/2021] [Indexed: 12/14/2022] Open
Abstract
Despite being perceived to be a relatively innocuous pathogen during its circulation in Africa in the 20th century, consequent outbreaks in French Polynesia and Latin America revealed the Zika virus (ZIKV) to be capable of causing severe neurological defects. Foetuses infected with the virus during pregnancy developed a range of pathologies including microcephaly, cerebral calcifications and macular scarring. These are now collectively known as Congenital Zika syndrome (CZS). It has been established that the neuropathogenesis of ZIKV results from infection of neural progenitor cells in the developing cerebral cortex. Following this, two main hypotheses have emerged: the virus causes either apoptosis or premature differentiation of neural progenitor cells, reducing the final number of mature neurons in the cerebral cortex. This review describes the cellular processes which could potentially cause virus induced apoptosis or premature differentiation, leading to speculation that a combination of the two may be responsible for the pathologies associated with ZIKV. The review also discusses which specific lineages of the ZIKV can employ these mechanisms. It has been unclear in the past whether the virus evolved its neurotropic capability following circulation in Africa, or if the virus has always caused microcephaly but public health surveillance in Africa had failed to detect it. Understanding the true neuropathogenesis of ZIKV is key to being prepared for further outbreaks in the future, and it will also provide insight into how neurotropic viruses can cause profound and life-long neurological defects.
Collapse
Affiliation(s)
- Emily Louise King
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Nerea Irigoyen
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
23
|
Guy B, Zhang JS, Duncan LH, Johnston RJ. Human neural organoids: Models for developmental neurobiology and disease. Dev Biol 2021; 478:102-121. [PMID: 34181916 PMCID: PMC8364509 DOI: 10.1016/j.ydbio.2021.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/08/2021] [Accepted: 06/24/2021] [Indexed: 12/25/2022]
Abstract
Human organoids stand at the forefront of basic and translational research, providing experimentally tractable systems to study human development and disease. These stem cell-derived, in vitro cultures can generate a multitude of tissue and organ types, including distinct brain regions and sensory systems. Neural organoid systems have provided fundamental insights into molecular mechanisms governing cell fate specification and neural circuit assembly and serve as promising tools for drug discovery and understanding disease pathogenesis. In this review, we discuss several human neural organoid systems, how they are generated, advances in 3D imaging and bioengineering, and the impact of organoid studies on our understanding of the human nervous system.
Collapse
Affiliation(s)
- Brian Guy
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, 21218, USA
| | - Jingliang Simon Zhang
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, 21218, USA
| | - Leighton H Duncan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Robert J Johnston
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, 21218, USA.
| |
Collapse
|
24
|
Kuroda R, Tominaga K, Kasashima K, Kuroiwa K, Sakashita E, Hayakawa H, Kouki T, Ohno N, Kawai K, Endo H. Loss of mitochondrial transcription factor A in neural stem cells leads to immature brain development and triggers the activation of the integral stress response in vivo. PLoS One 2021; 16:e0255355. [PMID: 34320035 PMCID: PMC8318236 DOI: 10.1371/journal.pone.0255355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 07/14/2021] [Indexed: 11/18/2022] Open
Abstract
Mitochondrial dysfunction is significantly associated with neurological deficits and age-related neurological diseases. While mitochondria are dynamically regulated and properly maintained during neurogenesis, the manner in which mitochondrial activities are controlled and contribute to these processes is not fully understood. Mitochondrial transcription factor A (TFAM) contributes to mitochondrial function by maintaining mitochondrial DNA (mtDNA). To clarify how mitochondrial dysfunction affects neurogenesis, we induced mitochondrial dysfunction specifically in murine neural stem cells (NSCs) by inactivating Tfam. Tfam inactivation in NSCs resulted in mitochondrial dysfunction by reducing respiratory chain activities and causing a severe deficit in neural differentiation and maturation both in vivo and in vitro. Brain tissue from Tfam-deficient mice exhibited neuronal cell death primarily at layer V and microglia were activated prior to cell death. Cultured Tfam-deficient NSCs showed a reduction in reactive oxygen species produced by the mitochondria. Tfam inactivation during neurogenesis resulted in the accumulation of ATF4 and activation of target gene expression. Therefore, we propose that the integrated stress response (ISR) induced by mitochondrial dysfunction in neurogenesis is activated to protect the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Rintaro Kuroda
- Department of Biochemistry, Jichi Medical University, Shimotsuke, Tochigi, Japan
- Department of Neurosurgery, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Kaoru Tominaga
- Department of Biochemistry, Jichi Medical University, Shimotsuke, Tochigi, Japan
- * E-mail: (KT); (HE)
| | - Katsumi Kasashima
- Department of Biochemistry, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Kenji Kuroiwa
- Department of Biochemistry, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Eiji Sakashita
- Department of Biochemistry, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Hiroko Hayakawa
- Core Center of Research Apparatus, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Tom Kouki
- Department of Anatomy, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Jichi Medical University, Shimotsuke, Tochigi, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Kensuke Kawai
- Department of Neurosurgery, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Hitoshi Endo
- Department of Biochemistry, Jichi Medical University, Shimotsuke, Tochigi, Japan
- * E-mail: (KT); (HE)
| |
Collapse
|
25
|
D'Souza L, Channakkar AS, Muralidharan B. Chromatin remodelling complexes in cerebral cortex development and neurodevelopmental disorders. Neurochem Int 2021; 147:105055. [PMID: 33964373 PMCID: PMC7611358 DOI: 10.1016/j.neuint.2021.105055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/11/2021] [Accepted: 04/24/2021] [Indexed: 12/19/2022]
Abstract
The diverse number of neurons in the cerebral cortex are generated during development by neural stem cells lining the ventricle, and they continue maturing postnatally. Dynamic chromatin regulation in these neural stem cells is a fundamental determinant of the emerging property of the functional neural network, and the chromatin remodellers are critical determinants of this process. Chromatin remodellers participate in several steps of this process from proliferation, differentiation, migration leading to complex network formation which forms the basis of higher-order functions of cognition and behaviour. Here we review the role of these ATP-dependent chromatin remodellers in cortical development in health and disease and highlight several key mouse mutants of the subunits of the complexes which have revealed how the remodelling mechanisms control the cortical stem cell chromatin landscape for expression of stage-specific transcripts. Consistent with their role in cortical development, several putative risk variants in the subunits of the remodelling complexes have been identified as the underlying causes of several neurodevelopmental disorders. A basic understanding of the detailed molecular mechanism of their action is key to understating how mutations in the same networks lead to disease pathologies and perhaps pave the way for therapeutic development for these complex multifactorial disorders.
Collapse
Affiliation(s)
- Leora D'Souza
- Brain Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore Life Science Cluster, Bangalore, India
| | - Asha S Channakkar
- Brain Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore Life Science Cluster, Bangalore, India
| | - Bhavana Muralidharan
- Brain Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore Life Science Cluster, Bangalore, India.
| |
Collapse
|
26
|
Jagomäe T, Singh K, Philips MA, Jayaram M, Seppa K, Tekko T, Gilbert SF, Vasar E, Lilleväli K. Alternative Promoter Use Governs the Expression of IgLON Cell Adhesion Molecules in Histogenetic Fields of the Embryonic Mouse Brain. Int J Mol Sci 2021; 22:6955. [PMID: 34203377 PMCID: PMC8268470 DOI: 10.3390/ijms22136955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/19/2021] [Accepted: 06/23/2021] [Indexed: 01/17/2023] Open
Abstract
The members of the IgLON superfamily of cell adhesion molecules facilitate fundamental cellular communication during brain development, maintain functional brain circuitry, and are associated with several neuropsychiatric disorders such as depression, autism, schizophrenia, and intellectual disabilities. Usage of alternative promoter-specific 1a and 1b mRNA isoforms in Lsamp, Opcml, Ntm, and the single promoter of Negr1 in the mouse and human brain has been previously described. To determine the precise spatiotemporal expression dynamics of Lsamp, Opcml, Ntm isoforms, and Negr1, in the developing brain, we generated isoform-specific RNA probes and carried out in situ hybridization in the developing (embryonic, E10.5, E11.5, 13.5, 17; postnatal, P0) and adult mouse brains. We show that promoter-specific expression of IgLONs is established early during pallial development (at E10.5), where it remains throughout its differentiation through adulthood. In the diencephalon, midbrain, and hindbrain, strong expression patterns are initiated a few days later and begin fading after birth, being only faintly expressed during adulthood. Thus, the expression of specific IgLONs in the developing brain may provide the means for regionally specific functionality as well as for specific regional vulnerabilities. The current study will therefore improve the understanding of how IgLON genes are implicated in the development of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Toomas Jagomäe
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (T.J.); (M.-A.P.); (M.J.); (K.S.); (E.V.); (K.L.)
- Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 50090 Tartu, Estonia
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia
| | - Katyayani Singh
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (T.J.); (M.-A.P.); (M.J.); (K.S.); (E.V.); (K.L.)
- Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 50090 Tartu, Estonia
| | - Mari-Anne Philips
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (T.J.); (M.-A.P.); (M.J.); (K.S.); (E.V.); (K.L.)
- Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 50090 Tartu, Estonia
| | - Mohan Jayaram
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (T.J.); (M.-A.P.); (M.J.); (K.S.); (E.V.); (K.L.)
- Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 50090 Tartu, Estonia
| | - Kadri Seppa
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (T.J.); (M.-A.P.); (M.J.); (K.S.); (E.V.); (K.L.)
- Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 50090 Tartu, Estonia
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia
| | - Triin Tekko
- The Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal;
| | - Scott F. Gilbert
- Department of Biology, Swarthmore College, Swarthmore, PA 19081, USA;
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (T.J.); (M.-A.P.); (M.J.); (K.S.); (E.V.); (K.L.)
- Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 50090 Tartu, Estonia
| | - Kersti Lilleväli
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (T.J.); (M.-A.P.); (M.J.); (K.S.); (E.V.); (K.L.)
- Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 50090 Tartu, Estonia
| |
Collapse
|
27
|
Antón IM, Wandosell F. WIP, YAP/TAZ and Actin Connections Orchestrate Development and Transformation in the Central Nervous System. Front Cell Dev Biol 2021; 9:673986. [PMID: 34195190 PMCID: PMC8237755 DOI: 10.3389/fcell.2021.673986] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/12/2021] [Indexed: 01/01/2023] Open
Abstract
YAP (Yes-associated protein) and TAZ (transcriptional coactivator with PDZ-binding motif) are transcription co-regulators that make up the terminal components of the Hippo signaling pathway, which plays a role in organ size control and derived tissue homeostasis through regulation of the proliferation, differentiation and apoptosis of a wide variety of differentiated and stem cells. Hippo/YAP signaling contributes to normal development of the nervous system, as it participates in self-renewal of neural stem cells, proliferation of neural progenitor cells and differentiation, activation and myelination of glial cells. Not surprisingly, alterations in this pathway underlie the development of severe neurological diseases. In glioblastomas, YAP and TAZ levels directly correlate with the amount of the actin-binding molecule WIP (WASP interacting protein), which regulates stemness and invasiveness. In neurons, WIP modulates cytoskeleton dynamics through actin polymerization/depolymerization and acts as a negative regulator of neuritogenesis, dendrite branching and dendritic spine formation. Our working hypothesis is that WIP regulates the YAP/TAZ pools using a Hippo-independent pathway. Thus, in this review we will present some of the data that links WIP, YAP and TAZ, with a focus on their function in cells from the central and peripheral nervous systems. It is hoped that a better understanding of the mechanisms involved in brain and nervous development and the pathologies that arise due to their alteration will reveal novel therapeutic targets for neurologic diseases.
Collapse
Affiliation(s)
- Inés M Antón
- Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Francisco Wandosell
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Departamento de Neuropatología Molecular, Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
28
|
Sokpor G, Rosenbusch J, Kunwar AJ, Rickmann M, Tuoc T, Rizzoli SO, Tarabykin V, von Mollard GF, Krieglstein K, Staiger JF. Ablation of Vti1a/1b Triggers Neural Progenitor Pool Depletion and Cortical Layer 5 Malformation in Late-embryonic Mouse Cortex. Neuroscience 2021; 463:303-316. [PMID: 33774122 DOI: 10.1016/j.neuroscience.2021.03.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 10/21/2022]
Abstract
Cortical morphogenesis entails several neurobiological events, including proliferation and differentiation of progenitors, migration of neuroblasts, and neuronal maturation leading to functional neural circuitry. These neurodevelopmental processes are delicately regulated by many factors. Endosomal SNAREs have emerged as formidable modulators of neuronal growth, aside their well-known function in membrane/vesicular trafficking. However, our understanding of their influence on cortex formation is limited. Here, we report that the SNAREs Vti1a and Vti1b (Vti1a/1b) are critical for proper cortical development. Following null mutation of Vti1a/1b in mouse, the late-embryonic mutant cortex appeared dysgenic, and the cortical progenitors therein were depleted beyond normal. Notably, cortical layer 5 (L5) is distinctively disorganized in the absence of Vti1a/1b. The latter defect, coupled with an overt apoptosis of Ctip2-expressing L5 neurons, likely contributed to the substantial loss of corticospinal and callosal projections in the Vti1a/1b-deficient mouse brain. These findings suggest that Vti1a/1b serve key neurodevelopmental functions during cortical histogenesis, which when mechanistically elucidated, can lend clarity to how endosomal SNAREs regulate brain development, or how their dysfunction may have implications for neurological disorders.
Collapse
Affiliation(s)
- Godwin Sokpor
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany; Institute for Human Genetics, Ruhr University of Bochum, Bochum, Germany
| | - Joachim Rosenbusch
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Ajaya J Kunwar
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany; Department of Anatomy, Nepalese Army Institute of Health Sciences, College of Medicine, Kathmandu, Nepal; Kathmandu Center for Genomics and Research Laboratory, Kathmandu, Nepal
| | - Michael Rickmann
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Tran Tuoc
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany; Institute for Human Genetics, Ruhr University of Bochum, Bochum, Germany
| | - Silvio O Rizzoli
- Institute of Neuro- and Sensory Physiology, University of Göttingen Medical Centre, Germany
| | - Victor Tarabykin
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany; Institute of Neuroscience, Lobachevsky State University of Nizhni Novogorod, 23 Prospekt Gagarina, 603950 Nizhny Novgorod, Russia
| | | | - Kerstin Krieglstein
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany; Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany.
| |
Collapse
|
29
|
Harkins D, Cooper HM, Piper M. The role of lipids in ependymal development and the modulation of adult neural stem cell function during aging and disease. Semin Cell Dev Biol 2020; 112:61-68. [PMID: 32771376 DOI: 10.1016/j.semcdb.2020.07.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/24/2020] [Accepted: 07/29/2020] [Indexed: 01/10/2023]
Abstract
Within the adult mammalian central nervous system, the ventricular-subventricular zone (V-SVZ) lining the lateral ventricles houses neural stem cells (NSCs) that continue to produce neurons throughout life. Developmentally, the V-SVZ neurogenic niche arises during corticogenesis following the terminal differentiation of telencephalic radial glial cells (RGCs) into either adult neural stem cells (aNSCs) or ependymal cells. In mice, these two cellular populations form rosettes during the late embryonic and early postnatal period, with ependymal cells surrounding aNSCs. These aNSCs and ependymal cells serve a number of key purposes, including the generation of neurons throughout life (aNSCs), and acting as a barrier between the CSF and the parenchyma and promoting CSF bulk flow (ependymal cells). Interestingly, the development of this neurogenic niche, as well as its ongoing function, has been shown to be reliant on different aspects of lipid biology. In this review we discuss the developmental origins of the rodent V-SVZ neurogenic niche, and highlight research which has implicated a role for lipids in the physiology of this part of the brain. We also discuss the role of lipids in the maintenance of the V-SVZ niche, and discuss new research which has suggested that alterations to lipid biology could contribute to ependymal cell dysfunction in aging and disease.
Collapse
Affiliation(s)
- Danyon Harkins
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Helen M Cooper
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia
| | - Michael Piper
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia.
| |
Collapse
|
30
|
Jossin Y. Reelin Functions, Mechanisms of Action and Signaling Pathways During Brain Development and Maturation. Biomolecules 2020; 10:biom10060964. [PMID: 32604886 PMCID: PMC7355739 DOI: 10.3390/biom10060964] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022] Open
Abstract
During embryonic development and adulthood, Reelin exerts several important functions in the brain including the regulation of neuronal migration, dendritic growth and branching, dendritic spine formation, synaptogenesis and synaptic plasticity. As a consequence, the Reelin signaling pathway has been associated with several human brain disorders such as lissencephaly, autism, schizophrenia, bipolar disorder, depression, mental retardation, Alzheimer’s disease and epilepsy. Several elements of the signaling pathway are known. Core components, such as the Reelin receptors very low-density lipoprotein receptor (VLDLR) and Apolipoprotein E receptor 2 (ApoER2), Src family kinases Src and Fyn, and the intracellular adaptor Disabled-1 (Dab1), are common to most but not all Reelin functions. Other downstream effectors are, on the other hand, more specific to defined tasks. Reelin is a large extracellular protein, and some aspects of the signal are regulated by its processing into smaller fragments. Rather than being inhibitory, the processing at two major sites seems to be fulfilling important physiological functions. In this review, I describe the various cellular events regulated by Reelin and attempt to explain the current knowledge on the mechanisms of action. After discussing the shared and distinct elements of the Reelin signaling pathway involved in neuronal migration, dendritic growth, spine development and synaptic plasticity, I briefly outline the data revealing the importance of Reelin in human brain disorders.
Collapse
Affiliation(s)
- Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium
| |
Collapse
|
31
|
Brock S, Vanderhasselt T, Vermaning S, Keymolen K, Régal L, Romaniello R, Wieczorek D, Storm TM, Schaeferhoff K, Hehr U, Kuechler A, Krägeloh-Mann I, Haack TB, Kasteleijn E, Schot R, Mancini GMS, Webster R, Mohammad S, Leventer RJ, Mirzaa G, Dobyns WB, Bahi-Buisson N, Meuwissen M, Jansen AC, Stouffs K. Defining the phenotypical spectrum associated with variants in TUBB2A. J Med Genet 2020; 58:33-40. [PMID: 32571897 PMCID: PMC7803914 DOI: 10.1136/jmedgenet-2019-106740] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/05/2020] [Accepted: 03/05/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND Variants in genes belonging to the tubulin superfamily account for a heterogeneous spectrum of brain malformations referred to as tubulinopathies. Variants in TUBB2A have been reported in 10 patients with a broad spectrum of brain imaging features, ranging from a normal cortex to polymicrogyria, while one patient has been reported with progressive atrophy of the cerebellar vermis. METHODS In order to further refine the phenotypical spectrum associated with TUBB2A, clinical and imaging features of 12 patients with pathogenic TUBB2A variants, recruited via the international network of the authors, were reviewed. RESULTS We report 12 patients with eight novel and one recurrent variants spread throughout the TUBB2A gene but encoding for amino acids clustering at the protein surface. Eleven patients (91.7%) developed seizures in early life. All patients suffered from intellectual disability, and 11 patients had severe motor developmental delay, with 4 patients (36.4 %) being non-ambulatory. The cerebral cortex was normal in five individuals and showed dysgyria of variable severity in seven patients. Associated brain malformations were less frequent in TUBB2A patients compared with other tubulinopathies. None of the patients had progressive cerebellar atrophy. CONCLUSION The imaging phenotype associated with pathogenic variants in TUBB2A is highly variable, ranging from a normal cortex to extensive dysgyria with associated brain malformations. For recurrent variants, no clear genotype-phenotype correlations could be established, suggesting the role of additional modifiers.
Collapse
Affiliation(s)
- Stefanie Brock
- Department of Pathology, Universitair Ziekenhuis Brussel, Brussels, Belgium .,Neurogenetics Research Group, Reproduction Genetics and Regenerative Medicine Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Tim Vanderhasselt
- Department of Radiology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Sietske Vermaning
- Belgium Center for Reproduction and Genetics, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Kathelijn Keymolen
- Belgium Center for Reproduction and Genetics, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Luc Régal
- Pediatric Neurology Unit, Department of Pediatrics, Universitair Ziekenhuis, Brussels, Belgium
| | - Romina Romaniello
- Neuropsychiatry and Neurorehabilitation Unit, Scientific Institute, IRCCS Eugenio Medea, Lecco, Italy
| | - Dagmar Wieczorek
- Institut fuer Humangenetik, Universitaetsklininikum Essen, Essen, Germany.,Institute of Human Genetics, Heinrich Heine University Düsseldorf, Dusseldorf, Nordrhein-Westfalen, Germany
| | - Tim Matthias Storm
- Institut für Humangenetik, Technische Universität München, Munchen, Bayern, Germany
| | - Karin Schaeferhoff
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Ute Hehr
- Zentrum für Humangenetik Regensburg, Universitätsklinikum Regensburg, Regensburg, Bayern, Germany
| | - Alma Kuechler
- Institut fuer Humangenetik, Universitaetsklininikum Essen, Essen, Germany
| | - Ingeborg Krägeloh-Mann
- Department of Pediatric Neurology and Developmental Medicine, University Children's Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Tobias B Haack
- Institute of Medical Genetics and Applied Genomics, Eberhard-Karls-Universitat Tubingen Medizinische Fakultat, Tübingen, Baden-Württemberg, Germany
| | - Esmee Kasteleijn
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Zuid-Holland, The Netherlands
| | - Rachel Schot
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Zuid-Holland, The Netherlands
| | - Grazia Maria Simonetta Mancini
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Zuid-Holland, The Netherlands.,ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Zuid-Holland, The Netherlands
| | - Richard Webster
- Department of Neurology, Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Shekeeb Mohammad
- Department of Neurology, Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Richard J Leventer
- Department of Neurology, Murdoch Childrens Research Institute, Melbourne, Victoria, Australia
| | - Ghayda Mirzaa
- Division of Genetic Medicine, Department of Pediatrics, Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - William B Dobyns
- Division of Genetic Medicine, Department of Pediatrics, Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Nadia Bahi-Buisson
- Embryology and Genetics of Congenital Malformations, INSERM, Paris, Île-de-France, France
| | - Marije Meuwissen
- Center of Human Genetics, Universiteit Antwerpen, Antwerpen, Belgium
| | - Anna C Jansen
- Neurogenetics Research Group, Reproduction Genetics and Regenerative Medicine Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium.,Pediatric Neurology Unit, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Katrien Stouffs
- Neurogenetics Research Group, Reproduction Genetics and Regenerative Medicine Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, Universitair Ziekenhuis Brussel, Brussels, Belgium, Brussels, Belgium
| |
Collapse
|
32
|
Jossin Y. Molecular mechanisms of cell polarity in a range of model systems and in migrating neurons. Mol Cell Neurosci 2020; 106:103503. [PMID: 32485296 DOI: 10.1016/j.mcn.2020.103503] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/20/2020] [Accepted: 05/23/2020] [Indexed: 01/09/2023] Open
Abstract
Cell polarity is defined as the asymmetric distribution of cellular components along an axis. Most cells, from the simplest single-cell organisms to highly specialized mammalian cells, are polarized and use similar mechanisms to generate and maintain polarity. Cell polarity is important for cells to migrate, form tissues, and coordinate activities. During development of the mammalian cerebral cortex, cell polarity is essential for neurogenesis and for the migration of newborn but as-yet undifferentiated neurons. These oriented migrations include both the radial migration of excitatory projection neurons and the tangential migration of inhibitory interneurons. In this review, I will first describe the development of the cerebral cortex, as revealed at the cellular level. I will then define the core molecular mechanisms - the Par/Crb/Scrib polarity complexes, small GTPases, the actin and microtubule cytoskeletons, and phosphoinositides/PI3K signaling - that are required for asymmetric cell division, apico-basal and front-rear polarity in model systems, including C elegans zygote, Drosophila embryos and cultured mammalian cells. As I go through each core mechanism I will explain what is known about its importance in radial and tangential migration in the developing mammalian cerebral cortex.
Collapse
Affiliation(s)
- Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
33
|
Tead transcription factors differentially regulate cortical development. Sci Rep 2020; 10:4625. [PMID: 32170161 PMCID: PMC7070074 DOI: 10.1038/s41598-020-61490-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/27/2020] [Indexed: 12/16/2022] Open
Abstract
Neural stem cells (NSCs) generate neurons of the cerebral cortex with distinct morphologies and functions. How specific neuron production, differentiation and migration are orchestrated is unclear. Hippo signaling regulates gene expression through Tead transcription factors (TFs). We show that Hippo transcriptional coactivators Yap1/Taz and the Teads have distinct functions during cortical development. Yap1/Taz promote NSC maintenance and Satb2+ neuron production at the expense of Tbr1+ neuron generation. However, Teads have moderate effects on NSC maintenance and do not affect Satb2+ neuron differentiation. Conversely, whereas Tead2 blocks Tbr1+ neuron formation, Tead1 and Tead3 promote this early fate. In addition, we found that Hippo effectors regulate neuronal migration to the cortical plate (CP) in a reciprocal fashion, that ApoE, Dab2 and Cyr61 are Tead targets, and these contribute to neuronal fate determination and migration. Our results indicate that multifaceted Hippo signaling is pivotal in different aspects of cortical development.
Collapse
|