42 results on '"Waclaw RR"'
Search Results
2. Distinct requirements for Tcf3 and Tcf12 during oligodendrocyte development in the mouse telencephalon.
- Author
-
Talley MJ, Nardini D, Ehrman LA, Lu QR, and Waclaw RR
- Subjects
- Animals, Mice, Cell Differentiation, Embryonic Stem Cells, Hysteria, Basic Helix-Loop-Helix Transcription Factors genetics, Transcription Factors genetics, Brain
- Abstract
Background: E-proteins encoded by Tcf3, Tcf4, and Tcf12 are class I basic helix-loop-helix (bHLH) transcription factors (TFs) that are thought to be widely expressed during development. However, their function in the developing brain, specifically in the telencephalon remains an active area of research. Our study examines for the first time if combined loss of two E-proteins (Tcf3 and Tcf12) influence distinct cell fates and oligodendrocyte development in the mouse telencephalon., Methods: We generated Tcf3/12 double conditional knockouts (dcKOs) using Olig2
Cre/+ or Olig1Cre/+ to overcome compensatory mechanisms between E-proteins and to understand the specific requirement for Tcf3 and Tcf12 in the ventral telencephalon and during oligodendrogenesis. We utilized a combination of in situ hybridization, immunohistochemistry, and immunofluorescence to address development of the telencephalon and oligodendrogenesis at embryonic and postnatal stages in Tcf3/12 dcKOs., Results: We show that the E-proteins Tcf3 and Tcf12 are expressed in progenitors of the embryonic telencephalon and throughout the oligodendrocyte lineage in the postnatal brain. Tcf3/12 dcKOs showed transient defects in progenitor cells with an enlarged medial ganglionic eminence (MGE) region which correlated with reduced generation of embryonic oligodendrocyte progenitor cells (OPCs) and increased expression of MGE interneuron genes. Postnatal Tcf3/12 dcKOs showed a recovery of OPCs but displayed a sustained reduction in mature oligodendrocytes (OLs). Interestingly, Tcf4 remained expressed in the dcKOs suggesting that it cannot compensate for the loss of Tcf3 and Tcf12. Generation of Tcf3/12 dcKOs with Olig1Cre/+ avoided the MGE morphology defect caused by Olig2Cre/+ but dcKOs still exhibited reduced embryonic OPCs and subsequent reduction in postnatal OLs., Conclusion: Our data reveal that Tcf3 and Tcf12 play a role in controlling OPC versus cortical interneuron cell fate decisions in MGE progenitors in addition to playing roles in the generation of embryonic OPCs and differentiation of postnatal OLs in the oligodendrocyte lineage., (© 2023. BioMed Central Ltd., part of Springer Nature.)- Published
- 2023
- Full Text
- View/download PDF
3. Conserved and Distinct Functions of the Autism-Related Chromatin Remodeler CHD8 in Embryonic and Adult Forebrain Neurogenesis.
- Author
-
Dong C, Zhao C, Chen X, Berry K, Wang J, Zhang F, Liao Y, Han R, Ogurek S, Xu L, Zhang L, Lin Y, Zhou W, Xin M, Lim DA, Campbell K, Nakafuku M, Waclaw RR, and Lu QR
- Subjects
- Animals, Female, Male, Mice, Fluoxetine, Hippocampus metabolism, Mice, Knockout, Tumor Suppressor Protein p53, Prosencephalon, Autistic Disorder, Chromatin, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Neurogenesis physiology
- Abstract
The chromatin remodeler CHD8 represents a high-confidence risk factor in autism, a multistage progressive neurologic disorder, however the underlying stage-specific functions remain elusive. In this study, by analyzing Chd8 conditional knock-out mice (male and female), we find that CHD8 controls cortical neural stem/progenitor cell (NSC) proliferation and survival in a stage-dependent manner. Strikingly, inducible genetic deletion reveals that CHD8 is required for the production and fitness of transit-amplifying intermediate progenitors (IPCs) essential for upper-layer neuron expansion in the embryonic cortex. p53 loss of function partially rescues apoptosis and neurogenesis defects in the Chd8 -deficient brain. Further, transcriptomic and epigenomic profiling indicates that CHD8 regulates the chromatin accessibility landscape to activate neurogenesis-promoting factors including TBR2, a key regulator of IPC neurogenesis, while repressing DNA damage- and p53-induced apoptotic programs. In the adult brain, CHD8 depletion impairs forebrain neurogenesis by impeding IPC differentiation from NSCs in both subventricular and subgranular zones; however, unlike in embryos, it does not affect NSC proliferation and survival. Treatment with an antidepressant approved by the Federal Drug Administration (FDA), fluoxetine, partially restores adult hippocampal neurogenesis in Chd8 -ablated mice. Together, our multistage functional studies identify temporally specific roles for CHD8 in developmental and adult neurogenesis, pointing to a potential strategy to enhance neurogenesis in the CHD8-deficient brain. SIGNIFICANCE STATEMENT The role of the high-confidence autism gene CHD8 in neurogenesis remains incompletely understood. Here, we identify a stage-specific function of CHD8 in development of NSCs in developing and adult brains by conserved, yet spatiotemporally distinct, mechanisms. In embryonic cortex, CHD8 is critical for the proliferation, survival, and differentiation of both NSC and IPCs during cortical neurogenesis. In adult brain, CHD8 is required for IPC generation but not the proliferation and survival of adult NSCs. Treatment with FDA-approved antidepressant fluoxetine partially rescues the adult neurogenesis defects in CHD8 mutants. Thus, our findings help resolve CHD8 functions throughout life during embryonic and adult neurogenesis and point to a potential avenue to promote neurogenesis in CHD8 deficiency., (Copyright © 2022 the authors.)
- Published
- 2022
- Full Text
- View/download PDF
4. Formation of the Mouse Internal Capsule and Cerebral Peduncle: A Pioneering Role for Striatonigral Axons as Revealed in Isl1 Conditional Mutants.
- Author
-
Ehrman JM, Merchan-Sala P, Ehrman LA, Chen B, Lim HW, Waclaw RR, and Campbell K
- Subjects
- Animals, Axons physiology, Cerebral Cortex metabolism, Internal Capsule, Mice, Mice, Knockout, Mice, Transgenic, Neural Pathways physiology, Thalamus, Autism Spectrum Disorder metabolism, Cerebral Peduncle
- Abstract
The projection neurons of the striatum, the principal nucleus of the basal ganglia, belong to one of the following two major pathways: the striatopallidal (indirect) pathway or the striatonigral (direct) pathway. Striatonigral axons project long distances and encounter ascending tracts (thalamocortical) while coursing alongside descending tracts (corticofugal) as they extend through the internal capsule and cerebral peduncle. These observations suggest that striatal circuitry may help to guide their trajectories. To investigate the developmental contributions of striatonigral axons to internal capsule formation, we have made use of Sox8-EGFP (striatal direct pathway) and Fezf2-TdTomato (corticofugal pathway) BAC transgenic reporter mice in combination with immunohistochemical markers to trace these axonal pathways throughout development. We show that striatonigral axons pioneer the internal capsule and cerebral peduncle and are temporally and spatially well positioned to provide guidance for corticofugal and thalamocortical axons. Using Isl1 conditional knock-out (cKO) mice, which exhibit disrupted striatonigral axon outgrowth, we observe both corticofugal and thalamocortical axon defects with either ventral forebrain- or telencephalon-specific Isl1 inactivation, despite Isl1 not being expressed in either cortical or thalamic projection neurons. Striatonigral axon defects can thus disrupt internal capsule formation. Our genome-wide transcriptomic analysis in Isl1 cKOs reveals changes in gene expression relevant to cell adhesion, growth cone dynamics, and extracellular matrix composition, suggesting potential mechanisms by which the striatonigral pathway exerts this guidance role. Together, our data support a novel pioneering role for the striatal direct pathway in the correct assembly of the ascending and descending axon tracts within the internal capsule and cerebral peduncle. SIGNIFICANCE STATEMENT The basal ganglia are a group of subcortical nuclei with established roles in the coordination of voluntary motor programs, aspects of cognition, and the selection of appropriate social behaviors. Hence, disruptions in basal ganglia connectivity have been implicated in the motor, cognitive, and social dysfunction characterizing common neurodevelopmental disorders such as attention-deficit/hyperactivity disorder, autism spectrum disorder, obsessive-compulsive disorder, and tic disorder. Here, we identified a novel role for the striatonigral (direct) pathway in pioneering the internal capsule and cerebral peduncle, and in guiding axons extending to and from the cortex. Our findings suggest that the abnormal development of basal ganglia circuits can drive secondary internal capsule defects and thereby may contribute to the pathology of these disorders., (Copyright © 2022 the authors.)
- Published
- 2022
- Full Text
- View/download PDF
5. A role for sustained MAPK activity in the mouse ventral telencephalon.
- Author
-
Talley MJ, Nardini D, Qin S, Prada CE, Ehrman LA, and Waclaw RR
- Subjects
- Animals, DNA-Binding Proteins metabolism, Embryo, Mammalian metabolism, Ganglia metabolism, Gene Expression genetics, Gene Expression Regulation, Developmental genetics, Homeodomain Proteins metabolism, MAP Kinase Kinase 1 metabolism, Mice, Mice, Transgenic, Neural Stem Cells cytology, Neurogenesis physiology, Neuroglia metabolism, Neurons metabolism, SOXE Transcription Factors genetics, Telencephalon embryology, Telencephalon physiology, Transcription Factors metabolism, Cell Differentiation physiology, MAP Kinase Signaling System physiology, Telencephalon metabolism
- Abstract
The MAPK pathway is a major growth signal that has been implicated during the development of progenitors, neurons, and glia in the embryonic brain. Here, we show that the MAPK pathway plays an important role in the generation of distinct cell types from progenitors in the ventral telencephalon. Our data reveal that phospho-p44/42 (called p-ERK1/2) and the ETS transcription factor Etv5, both downstream effectors in the MAPK pathway, show a regional bias in expression during ventral telencephalic development, with enriched expression in the dorsal region of the LGE and ventral region of the MGE at E13.5 and E15.5. Interestingly, expression of both factors becomes more uniform in ventricular zone (VZ) progenitors by E18.5. To gain insight into the role of MAPK activity during progenitor cell development, we used a cre inducible constitutively active MEK1 allele (Rosa
MEK1DD/+ ) in combination with a ventral telencephalon enriched cre (Gsx2e-cre) or a dorsal telencephalon enriched cre (Emx1cre/+ ). Sustained MEK/MAPK activity in the ventral telencephalon (Gsx2e-cre; RosaMEK1DD/+ ) expanded dorsal lateral ganglionic eminence (dLGE) enriched genes (Gsx2 and Sp8) and oligodendrocyte progenitor cell (OPC) markers (Olig2, Pdgfrα, and Sox10), and also reduced markers in the ventral (v) LGE domain (Isl1 and Foxp1). Activation of MEK/MAPK activity in the dorsal telencephalon (Emx1cre/+ ; RosaMEK1DD/+ ) did not initially activate the expression of dLGE or OPC genes at E15.5 but ectopic expression of Gsx2 and OPC markers were observed at E18.5. These results support the idea that MAPK activity as readout by p-ERK1/2 and Etv5 expression is enriched in distinct subdomains of ventral telencephalic progenitors during development. In addition, sustained activation of the MEK/MAPK pathway in the ventral or dorsal telencephalon influences dLGE and OPC identity from progenitors., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
6. A Novel Mutation in Cse1l Disrupts Brain and Eye Development with Specific Effects on Pax6 Expression.
- Author
-
Blizzard LE, Menke C, Patel SD, Waclaw RR, Lachke SA, and Stottmann RW
- Abstract
Forward genetics in the mouse continues to be a useful and unbiased approach to identifying new genes and alleles with previously unappreciated roles in mammalian development and disease. Here, we report a new mouse allele of Cse1l that was recovered from an ENU mutagenesis screen. Embryos homozygous for the anteater allele of Cse1l display a number of variable phenotypes, with craniofacial and ocular malformations being the most obvious. We provide evidence that Cse1l is the causal gene through complementation with a novel null allele of Cse1l generated by CRISPR-Cas9 editing. While the variability in the anteater phenotype was high enough to preclude a detailed molecular analysis, we demonstrate a very penetrant reduction in Pax6 levels in the developing eye along with significant ocular developmental phenotypes. The eye gene discovery tool iSyTE shows Cse1l to be significantly expressed in the lens from early eye development stages in embryos through adulthood. Cse1l has not previously been shown to be required for organogenesis as homozygosity for a null allele results in very early lethality. Future detailed studies of Cse1l function in craniofacial and neural development will be best served with a conditional allele to circumvent the variable phenotypes we report here. We suggest that human next-generation (whole genome or exome) sequencing studies yielding variants of unknown significance in CSE1L could consider these findings as part of variant analysis.
- Published
- 2021
- Full Text
- View/download PDF
7. Generation of a Mouse Model to Study the Noonan Syndrome Gene Lztr1 in the Telencephalon.
- Author
-
Talley MJ, Nardini D, Shabbir N, Ehrman LA, Prada CE, and Waclaw RR
- Abstract
The leucine zipper-like transcriptional regulator 1 ( Lztr1 ) is a BTB-Kelch domain protein involved in RAS/MAPK pathway regulation. Mutations in LZTR1 are associated with cancers and Noonan syndrome, the most common RASopathy. The expression and function of Lztr1 in the developing brain remains poorly understood. Here we show that Lztr1 is expressed in distinct regions of the telencephalon, the most anterior region of the forebrain. Lztr1 expression was robust in the cortex, amygdala, hippocampus, and oligodendrocytes in the white matter. To gain insight into the impact of Lztr1 deficiency, we generated a conditional knockout (cKO) restricted to the telencephalon using Foxg1
IREScre/ + . Lztr1 cKOs are viable to postnatal stages and show reduced Lztr1 expression in the telencephalon. Interestingly, Lztr1 cKOs exhibit an increase in MAPK pathway activation in white matter regions and subsequently show an altered expression of stage-specific markers in the oligodendrocyte lineage with increased oligodendrocyte progenitor cells (OPCs) and decreased markers of oligodendrocyte differentiation. Moreover, Lztr1 cKOs also exhibit an increased expression of the astrocyte marker GFAP. These results highlight the generation of a new mouse model to study Lztr1 deficiency in the brain and reveal a novel role for Lztr1 in normal oligodendrocyte and astrocyte development in the telencephalon., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Talley, Nardini, Shabbir, Ehrman, Prada and Waclaw.)- Published
- 2021
- Full Text
- View/download PDF
8. Analysis of reactive astrogliosis in mouse brain using in situ hybridization combined with immunohistochemistry.
- Author
-
Muraleedharan R, Nardini D, Waclaw RR, and Dasgupta B
- Subjects
- Animals, Astrocytes metabolism, Brain metabolism, Brain physiology, Cells, Cultured, Central Nervous System metabolism, Gliosis metabolism, Gliosis physiopathology, Inflammation, Mice, Neurons metabolism, Gliosis diagnostic imaging, Immunohistochemistry methods, In Situ Hybridization methods
- Abstract
Reactive astrogliosis is characterized by a profound change in astrocyte phenotype in response to all CNS injuries. Here, we present a revised in situ hybridization and immunohistochemistry (IHC) protocol to label the reactive astrocytes in the mouse brain. Several approaches for quantifying astrocyte reactivity lacked sensitivity to discriminate across the spectrum. We optimized in situ hybridization followed by IHC. We provide a staining protocol for quantitative measures of astrocyte reactivity as an independent confirmation of the magnitude of reactive gliosis. For complete details on the use and execution of this protocol, please refer to Muraleedharan et al. (2020)., Competing Interests: The authors declare no competing interests., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
9. Temporally Distinct Roles for the Zinc Finger Transcription Factor Sp8 in the Generation and Migration of Dorsal Lateral Ganglionic Eminence (dLGE)-Derived Neuronal Subtypes in the Mouse.
- Author
-
Kuerbitz J, Madhavan M, Ehrman LA, Kohli V, Waclaw RR, and Campbell K
- Subjects
- Animals, Globus Pallidus embryology, Mice, Neural Stem Cells metabolism, Cell Movement physiology, DNA-Binding Proteins metabolism, Globus Pallidus cytology, Neural Stem Cells cytology, Neurogenesis physiology, Transcription Factors metabolism
- Abstract
Progenitors in the dorsal lateral ganglionic eminence (dLGE) are known to give rise to olfactory bulb (OB) interneurons and intercalated cells (ITCs) of the amygdala. The dLGE enriched transcription factor Sp8 is required for the normal generation of ITCs as well as OB interneurons, particularly the calretinin (CR)-expressing subtype. In this study, we used a genetic gain-of-function approach in mice to examine the roles Sp8 plays in controlling the development of dLGE-derived neuronal subtypes. Misexpression of Sp8 throughout the ventral telencephalic subventricular zone (SVZ) from early embryonic stages, led to an increased generation of ITCs which was dependent on Tshz1 gene dosage. Additionally, Sp8 misexpression impaired rostral migration of OB interneurons with clusters of CR interneurons seen in the SVZ along with decreased differentiation of calbindin OB interneurons. Sp8 misexpression throughout the ventral telencephalon also reduced ventral LGE neuronal subtypes including striatal projection neurons. Delaying Sp8 misexpression until E14-15 rescued the striatal and amygdala phenotypes but only partially rescued OB interneuron reductions, consistent with an early window of striatal and amygdala neurogenesis and ongoing OB interneuron generation at this late stage. Our results demonstrate critical roles for the timing and neuronal cell-type specificity of Sp8 expression in mouse LGE neurogenesis., (© The Author(s) 2020. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permission@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
10. Enhanced MAPK1 Function Causes a Neurodevelopmental Disorder within the RASopathy Clinical Spectrum.
- Author
-
Motta M, Pannone L, Pantaleoni F, Bocchinfuso G, Radio FC, Cecchetti S, Ciolfi A, Di Rocco M, Elting MW, Brilstra EH, Boni S, Mazzanti L, Tamburrino F, Walsh L, Payne K, Fernández-Jaén A, Ganapathi M, Chung WK, Grange DK, Dave-Wala A, Reshmi SC, Bartholomew DW, Mouhlas D, Carpentieri G, Bruselles A, Pizzi S, Bellacchio E, Piceci-Sparascio F, Lißewski C, Brinkmann J, Waclaw RR, Waisfisz Q, van Gassen K, Wentzensen IM, Morrow MM, Álvarez S, Martínez-García M, De Luca A, Memo L, Zampino G, Rossi C, Seri M, Gelb BD, Zenker M, Dallapiccola B, Stella L, Prada CE, Martinelli S, Flex E, and Tartaglia M
- Subjects
- Child, Preschool, Female, Humans, MAP Kinase Signaling System genetics, Male, Mutation, Missense genetics, Neurodevelopmental Disorders pathology, Noonan Syndrome physiopathology, Phenotype, Protein Tyrosine Phosphatase, Non-Receptor Type 11 genetics, Signal Transduction, Exome Sequencing, ras Proteins genetics, Carcinogenesis genetics, Mitogen-Activated Protein Kinase 1 genetics, Neurodevelopmental Disorders genetics, Noonan Syndrome genetics
- Abstract
Signal transduction through the RAF-MEK-ERK pathway, the first described mitogen-associated protein kinase (MAPK) cascade, mediates multiple cellular processes and participates in early and late developmental programs. Aberrant signaling through this cascade contributes to oncogenesis and underlies the RASopathies, a family of cancer-prone disorders. Here, we report that de novo missense variants in MAPK1, encoding the mitogen-activated protein kinase 1 (i.e., extracellular signal-regulated protein kinase 2, ERK2), cause a neurodevelopmental disease within the RASopathy phenotypic spectrum, reminiscent of Noonan syndrome in some subjects. Pathogenic variants promote increased phosphorylation of the kinase, which enhances translocation to the nucleus and boosts MAPK signaling in vitro and in vivo. Two variant classes are identified, one of which directly disrupts binding to MKP3, a dual-specificity protein phosphatase negatively regulating ERK function. Importantly, signal dysregulation driven by pathogenic MAPK1 variants is stimulus reliant and retains dependence on MEK activity. Our data support a model in which the identified pathogenic variants operate with counteracting effects on MAPK1 function by differentially impacting the ability of the kinase to interact with regulators and substrates, which likely explains the minor role of these variants as driver events contributing to oncogenesis. After nearly 20 years from the discovery of the first gene implicated in Noonan syndrome, PTPN11, the last tier of the MAPK cascade joins the group of genes mutated in RASopathies., (Copyright © 2020 American Society of Human Genetics. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
11. Single-Cell Transcriptomics Uncovers Glial Progenitor Diversity and Cell Fate Determinants during Development and Gliomagenesis.
- Author
-
Weng Q, Wang J, Wang J, He D, Cheng Z, Zhang F, Verma R, Xu L, Dong X, Liao Y, He X, Potter A, Zhang L, Zhao C, Xin M, Zhou Q, Aronow BJ, Blackshear PJ, Rich JN, He Q, Zhou W, Suvà ML, Waclaw RR, Potter SS, Yu G, and Lu QR
- Subjects
- Animals, Biodiversity, Butyrate Response Factor 1 genetics, Carcinogenesis, Cell Differentiation, Cellular Reprogramming, Fetal Development, Gene Expression Regulation, Humans, Mice, Mice, Knockout, Glioma genetics, Neoplastic Stem Cells physiology, Neuroglia physiology, Sequence Analysis, RNA methods, Single-Cell Analysis methods, Stem Cells physiology, Transcriptome genetics
- Abstract
The identity and degree of heterogeneity of glial progenitors and their contributions to brain tumor malignancy remain elusive. By applying lineage-targeted single-cell transcriptomics, we uncover an unanticipated diversity of glial progenitor pools with unique molecular identities in developing brain. Our analysis identifies distinct transitional intermediate states and their divergent developmental trajectories in astroglial and oligodendroglial lineages. Moreover, intersectional analysis uncovers analogous intermediate progenitors during brain tumorigenesis, wherein oligodendrocyte-progenitor intermediates are abundant, hyper-proliferative, and progressively reprogrammed toward a stem-like state susceptible to further malignant transformation. Similar actively cycling intermediate progenitors are prominent components in human gliomas with distinct driver mutations. We further unveil lineage-driving networks underlying glial fate specification and identify Zfp36l1 as necessary for oligodendrocyte-astrocyte lineage transition and glioma growth. Together, our results resolve the dynamic repertoire of common and divergent glial progenitors during development and tumorigenesis and highlight Zfp36l1 as a molecular nexus for balancing glial cell-fate decision and controlling gliomagenesis., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
12. Gsx transcription factors control neuronal versus glial specification in ventricular zone progenitors of the mouse lateral ganglionic eminence.
- Author
-
Chapman H, Riesenberg A, Ehrman LA, Kohli V, Nardini D, Nakafuku M, Campbell K, and Waclaw RR
- Subjects
- Animals, Cell Differentiation genetics, Cell Differentiation physiology, Cell Lineage, Embryo, Mammalian metabolism, Ganglia metabolism, Ganglia physiology, Homeodomain Proteins metabolism, Mice, Mice, Transgenic, Neural Stem Cells cytology, Neurogenesis physiology, Neuroglia metabolism, Neuroglia physiology, Neurons metabolism, Neurons physiology, Oligodendrocyte Transcription Factor 2, Oligodendroglia cytology, Oligodendroglia physiology, Stem Cells metabolism, Stem Cells physiology, Telencephalon metabolism, Transcription Factors, Homeodomain Proteins genetics
- Abstract
The homeobox gene Gsx2 has previously been shown to inhibit oligodendroglial specification in dorsal lateral ganglionic eminence (dLGE) progenitors of the ventral telencephalon. The precocious specification of oligodendrocyte progenitor cells (OPCs) observed in Gsx2 mutants, however, is transient and begins to normalize by late stages of embryogenesis. Interestingly, this normalization correlates with the expansion of Gsx1, a close homolog of Gsx2, in a subset of progenitors in the Gsx2 mutant LGE. Here, we interrogated the mechanisms underlying oligodendroglial specification in Gsx2 mutants in relation to Gsx1. We found that Gsx1/2 double mutant embryos exhibit a more robust expansion of Olig2
+ cells (i.e. OPCs) in the subventricular zone (SVZ) of the dLGE than Gsx2 mutants. Moreover, misexpression of Gsx1 throughout telencephalic VZ progenitors from E15 and onward resulted in a significant reduction of cortical OPCs. These results demonstrate redundant roles of Gsx1 and Gsx2 in suppressing early OPC specification in LGE VZ progenitors. However, Gsx1/2 mutants did not show a significant increase in adjacent cortical OPCs at later stages compared to Gsx2 mutants. This is likely due to reduced proliferation of OPCs within the SVZ of the Gsx1/2 double mutant LGE, suggesting a novel role for Gsx1 in expansion of migrating OPCs in the ventral telencephalon. We further investigated the glial specification mechanisms downstream of Gsx2 by generating Olig2/Gsx2 double mutants. Consistent with the known essential role for Olig2 in OPC specification, ectopic production of cortical OPCs observed in Gsx2 mutants disappeared in Olig2/Gsx2 double mutants. These mutants, however, maintained the expanded expression of gliogenic markers Zbtb20 and Bcan in the VZ of the LGE similarly to Gsx2 single mutants, suggesting that Gsx2 suppresses gliogenesis via Olig2-dependent and -independent mechanisms., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
13. Gain-of-function mutations in the gene encoding the tyrosine phosphatase SHP2 induce hydrocephalus in a catalytically dependent manner.
- Author
-
Zheng H, Yu WM, Waclaw RR, Kontaridis MI, Neel BG, and Qu CK
- Subjects
- Animals, Biocatalysis, Ependyma cytology, Ependyma metabolism, Genetic Predisposition to Disease genetics, Humans, Hydrocephalus metabolism, LEOPARD Syndrome metabolism, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Neural Stem Cells metabolism, Noonan Syndrome metabolism, Protein Tyrosine Phosphatase, Non-Receptor Type 11 metabolism, Gain of Function Mutation, Hydrocephalus genetics, LEOPARD Syndrome genetics, Noonan Syndrome genetics, Protein Tyrosine Phosphatase, Non-Receptor Type 11 genetics
- Abstract
Catalytically activating mutations in Ptpn11 , which encodes the protein tyrosine phosphatase SHP2, cause 50% of Noonan syndrome (NS) cases, whereas inactivating mutations in Ptpn11 are responsible for nearly all cases of the similar, but distinct, developmental disorder Noonan syndrome with multiple lentigines (NSML; formerly called LEOPARD syndrome). However, both types of disease mutations are gain-of-function mutations because they cause SHP2 to constitutively adopt an open conformation. We found that the catalytic activity of SHP2 was required for the pathogenic effects of gain-of-function, disease-associated mutations on the development of hydrocephalus in the mouse. Targeted pan-neuronal knockin of a Ptpn11 allele encoding the active SHP2 E76K mutant resulted in hydrocephalus due to aberrant development of ependymal cells and their cilia. These pathogenic effects of the E76K mutation were suppressed by the additional mutation C459S, which abolished the catalytic activity of SHP2. Moreover, ependymal cells in NSML mice bearing the inactive SHP2 mutant Y279C were also unaffected. Mechanistically, the SHP2 E76K mutant induced developmental defects in ependymal cells by enhancing dephosphorylation and inhibition of the transcription activator STAT3. Whereas STAT3 activity was reduced in Ptpn11
E76K/+ cells, the activities of the kinases ERK and AKT were enhanced, and neural cell-specific Stat3 knockout mice also manifested developmental defects in ependymal cells and cilia. These genetic and biochemical data demonstrate a catalytic-dependent role of SHP2 gain-of-function disease mutants in the pathogenesis of hydrocephalus., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2018
- Full Text
- View/download PDF
14. Dek overexpression in murine epithelia increases overt esophageal squamous cell carcinoma incidence.
- Author
-
Matrka MC, Cimperman KA, Haas SR, Guasch G, Ehrman LA, Waclaw RR, Komurov K, Lane A, Wikenheiser-Brokamp KA, and Wells SI
- Subjects
- 4-Nitroquinoline-1-oxide toxicity, Animals, Carcinoma, Squamous Cell chemically induced, DNA-Binding Proteins metabolism, Epithelium pathology, Esophageal Neoplasms chemically induced, Esophageal Squamous Cell Carcinoma, Gene Expression Regulation, Neoplastic, Keratinocytes pathology, Mice, Transgenic, Oncogene Proteins metabolism, Poly-ADP-Ribose Binding Proteins metabolism, Response Elements drug effects, Response Elements genetics, Tetracycline pharmacology, Tongue drug effects, Tongue pathology, Transgenes, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell pathology, DNA-Binding Proteins genetics, Esophageal Neoplasms genetics, Esophageal Neoplasms pathology, Oncogene Proteins genetics, Poly-ADP-Ribose Binding Proteins genetics
- Abstract
Esophageal cancer occurs as either squamous cell carcinoma (ESCC) or adenocarcinoma. ESCCs comprise almost 90% of cases worldwide, and recur with a less than 15% five-year survival rate despite available treatments. The identification of new ESCC drivers and therapeutic targets is critical for improving outcomes. Here we report that expression of the human DEK oncogene is strongly upregulated in esophageal SCC based on data in the cancer genome atlas (TCGA). DEK is a chromatin-associated protein with important roles in several nuclear processes including gene transcription, epigenetics, and DNA repair. Our previous data have utilized a murine knockout model to demonstrate that Dek expression is required for oral and esophageal SCC growth. Also, DEK overexpression in human keratinocytes, the cell of origin for SCC, was sufficient to cause hyperplasia in 3D organotypic raft cultures that mimic human skin, thus linking high DEK expression in keratinocytes to oncogenic phenotypes. However, the role of DEK over-expression in ESCC development remains unknown in human cells or genetic mouse models. To define the consequences of Dek overexpression in vivo, we generated and validated a tetracycline responsive Dek transgenic mouse model referred to as Bi-L-Dek. Dek overexpression was induced in the basal keratinocytes of stratified squamous epithelium by crossing Bi-L-Dek mice to keratin 5 tetracycline transactivator (K5-tTA) mice. Conditional transgene expression was validated in the resulting Bi-L-Dek_K5-tTA mice and was suppressed with doxycycline treatment in the tetracycline-off system. The mice were subjected to an established HNSCC and esophageal carcinogenesis protocol using the chemical carcinogen 4-nitroquinoline 1-oxide (4NQO). Dek overexpression stimulated gross esophageal tumor development, when compared to doxycycline treated control mice. Furthermore, high Dek expression caused a trend toward esophageal hyperplasia in 4NQO treated mice. Taken together, these data demonstrate that Dek overexpression in the cell of origin for SCC is sufficient to promote esophageal SCC development in vivo.
- Published
- 2018
- Full Text
- View/download PDF
15. A histone deacetylase 3-dependent pathway delimits peripheral myelin growth and functional regeneration.
- Author
-
He X, Zhang L, Queme LF, Liu X, Lu A, Waclaw RR, Dong X, Zhou W, Kidd G, Yoon SO, Buonanno A, Rubin JB, Xin M, Nave KA, Trapp BD, Jankowski MP, and Lu QR
- Subjects
- Animals, Genome, Histone Deacetylases, Humans, Mice, Transgenic, Myelin Sheath genetics, Myelin Sheath metabolism, Nerve Degeneration genetics, Nerve Degeneration physiopathology, Peripheral Nerve Injuries physiopathology, Peripheral Nerve Injuries rehabilitation, Recovery of Function genetics, Schwann Cells metabolism, Schwann Cells pathology, Sciatic Nerve growth & development, Sciatic Nerve injuries, Sciatic Nerve metabolism, Signal Transduction, TEA Domain Transcription Factors, DNA-Binding Proteins genetics, E1A-Associated p300 Protein genetics, Muscle Proteins genetics, Nerve Regeneration genetics, Peripheral Nerve Injuries genetics, Remyelination genetics, Transcription Factors genetics
- Abstract
Deficits in Schwann cell-mediated remyelination impair functional restoration after nerve damage, contributing to peripheral neuropathies. The mechanisms mediating block of remyelination remain elusive. Here, through small-molecule screening focusing on epigenetic modulators, we identified histone deacetylase 3 (HDAC3; a histone-modifying enzyme) as a potent inhibitor of peripheral myelinogenesis. Inhibition of HDAC3 enhanced myelin growth and regeneration and improved functional recovery after peripheral nerve injury in mice. HDAC3 antagonizes the myelinogenic neuregulin-PI3K-AKT signaling axis. Moreover, genome-wide profiling analyses revealed that HDAC3 represses promyelinating programs through epigenetic silencing while coordinating with p300 histone acetyltransferase to activate myelination-inhibitory programs that include the HIPPO signaling effector TEAD4 to inhibit myelin growth. Schwann cell-specific deletion of either Hdac3 or Tead4 in mice resulted in an elevation of myelin thickness in sciatic nerves. Thus, our findings identify the HDAC3-TEAD4 network as a dual-function switch of cell-intrinsic inhibitory machinery that counters myelinogenic signals and maintains peripheral myelin homeostasis, highlighting the therapeutic potential of transient HDAC3 inhibition for improving peripheral myelin repair.
- Published
- 2018
- Full Text
- View/download PDF
16. Loss of Intercalated Cells (ITCs) in the Mouse Amygdala of Tshz1 Mutants Correlates with Fear, Depression, and Social Interaction Phenotypes.
- Author
-
Kuerbitz J, Arnett M, Ehrman S, Williams MT, Vorhees CV, Fisher SE, Garratt AN, Muglia LJ, Waclaw RR, and Campbell K
- Subjects
- Amygdala growth & development, Animals, Behavior, Animal, Extinction, Psychological physiology, Female, Forkhead Transcription Factors genetics, Forkhead Transcription Factors physiology, Homeodomain Proteins, Male, Mice, Mice, Knockout, Motor Activity genetics, Phenotype, Pregnancy, Repressor Proteins physiology, Amygdala pathology, Depression genetics, Depression psychology, Fear psychology, Interneurons pathology, Interpersonal Relations, Mutation genetics, Repressor Proteins genetics
- Abstract
The intercalated cells (ITCs) of the amygdala have been shown to be critical regulatory components of amygdalar circuits, which control appropriate fear responses. Despite this, the molecular processes guiding ITC development remain poorly understood. Here we establish the zinc finger transcription factor Tshz1 as a marker of ITCs during their migration from the dorsal lateral ganglionic eminence through maturity. Using germline and conditional knock-out (cKO) mouse models, we show that Tshz1 is required for the proper migration and differentiation of ITCs. In the absence of Tshz1 , migrating ITC precursors fail to settle in their stereotypical locations encapsulating the lateral amygdala and BLA. Furthermore, they display reductions in the ITC marker Foxp2 and ectopic persistence of the dorsal lateral ganglionic eminence marker Sp8. Tshz1 mutant ITCs show increased cell death at postnatal time points, leading to a dramatic reduction by 3 weeks of age. In line with this, Foxp2 -null mutants also show a loss of ITCs at postnatal time points, suggesting that Foxp2 may function downstream of Tshz1 in the maintenance of ITCs. Behavioral analysis of male Tshz1 cKOs revealed defects in fear extinction as well as an increase in floating during the forced swim test, indicative of a depression-like phenotype. Moreover, Tshz1 cKOs display significantly impaired social interaction (i.e., increased passivity) regardless of partner genetics. Together, these results suggest that Tshz1 plays a critical role in the development of ITCs and that fear, depression-like and social behavioral deficits arise in their absence. SIGNIFICANCE STATEMENT We show here that the zinc finger transcription factor Tshz1 is expressed during development of the intercalated cells (ITCs) within the mouse amygdala. These neurons have previously been shown to play a crucial role in fear extinction. Tshz1 mouse mutants exhibit severely reduced numbers of ITCs as a result of abnormal migration, differentiation, and survival of these neurons. Furthermore, the loss of ITCs in mouse Tshz1 mutants correlates well with defects in fear extinction as well as the appearance of depression-like and abnormal social interaction behaviors reminiscent of depressive disorders observed in human patients with distal 18q deletions, including the Tshz1 locus., (Copyright © 2018 the authors 0270-6474/18/381160-18$15.00/0.)
- Published
- 2018
- Full Text
- View/download PDF
17. Characterization of Glcci1 expression in a subpopulation of lateral ganglionic eminence progenitors in the mouse telencephalon.
- Author
-
Kohli V, Nardini D, Ehrman LA, and Waclaw RR
- Subjects
- Animals, Mice, Mice, Transgenic, Neurons metabolism, Receptors, Glucocorticoid genetics, Telencephalon embryology, Gene Expression Regulation, Developmental, Neural Stem Cells metabolism, Receptors, Glucocorticoid metabolism, Telencephalon metabolism
- Abstract
Background: The lateral ganglionic eminence (LGE) in the ventral telencephalon is a diverse progenitor domain subdivided by distinct gene expression into a dorsal region (dLGE) that gives rise to olfactory bulb and amygdalar interneurons and a ventral region (vLGE) that gives rise to striatal projection neurons. The homeobox gene, Gsx2, is an enriched marker of the LGE and is expressed in a high dorsal to low ventral gradient in the ventricular zone (VZ) as development proceeds. Aside from Gsx2, markers restricted to the VZ in the dLGE and/or vLGE remain largely unknown., Results: Here, we show that the gene and protein expression of Glucocorticoid-induced transcript 1 (Glcci1) has a similar dorsal to ventral gradient of expression in the VZ as Gsx2. We found that Glcci1 gene and protein expression are reduced in Gsx2 mutants, and are increased in the cortex after early and late Gsx2 misexpression. Moreover, Glcci1 expressing cells are restricted to a subpopulation of Gsx2 positive cells on the basal side of the VZ and co-express Ascl1, but not the subventricular zone dLGE marker, Sp8., Conclusions: These findings suggest that Glcci1 is a new marker of a subpopulation of LGE VZ progenitor cells in the Gsx2 lineage. Developmental Dynamics 247:222-228, 2018. © 2017 Wiley Periodicals, Inc., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
18. Selective neuronal expression of the SoxE factor, Sox8, in direct pathway striatal projection neurons of the developing mouse brain.
- Author
-
Merchan-Sala P, Nardini D, Waclaw RR, and Campbell K
- Subjects
- Animals, Animals, Newborn, Embryo, Mammalian, Forkhead Transcription Factors metabolism, Gene Expression Regulation, Developmental genetics, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, LIM-Homeodomain Proteins genetics, LIM-Homeodomain Proteins metabolism, Mice, Mice, Transgenic, Mutation genetics, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Receptors, Dopamine D1 genetics, Receptors, Dopamine D1 metabolism, Receptors, Dopamine D2 genetics, Receptors, Dopamine D2 metabolism, Repressor Proteins metabolism, SOXE Transcription Factors genetics, Transcription Factors genetics, Transcription Factors metabolism, Corpus Striatum cytology, Corpus Striatum embryology, Corpus Striatum growth & development, Corpus Striatum metabolism, Gene Expression Regulation, Developmental physiology, Neural Pathways physiology, Neurons metabolism, SOXE Transcription Factors metabolism, Signal Transduction physiology
- Abstract
The striatum is the major component of the basal ganglia and is well known to play a key role in the control of motor function via balanced output from the indirect (iSPNs) and direct pathway striatal projection neurons (dSPNs). Little is known, however, about the molecular genetic mechanisms that control the formation of the iSPNs versus dSPNs. We show here that the SoxE family member, Sox8, is co-expressed with the dSPN markers, Isl1 and Ebf1, in the developing striatum. Moreover, dSPNs, as marked by Isl1-cre fate map, express Sox8 in the embryonic striatum and Sox8-EGFP BAC transgenic mice specifically reveal the direct pathway axons during development. These EGFP
+ axons are first observed to reach their midbrain target, the substantia nigra pars reticulata (SNr), at E14 in the mouse with a robust connection observed already at birth. The selective expression of EGFP in dSPNs of Sox8-EGFP BAC mice is maintained at postnatal timepoints. Sox8 is known to be expressed in oligodendrocyte precursor cells (OPCs) together with other SoxE factors and we show here that the EGFP signal co-localizes with the OPC markers throughout the brain. Finally, we show that Sox8-EGFP BAC mice can be used to interrogate the altered dSPN development in Isl1 conditional mutants including aberrant axonal projections detected already at embryonic timepoints. Thus, Sox8 represents an early and specific marker of embryonic dSPNs and the Sox8-EGFP BAC transgenic mice are an excellent tool to study the development of basal ganglia circuitry., (© 2017 Wiley Periodicals, Inc.)- Published
- 2017
- Full Text
- View/download PDF
19. Septal contributions to olfactory bulb interneuron diversity in the embryonic mouse telencephalon: role of the homeobox gene Gsx2.
- Author
-
Qin S, Ware SM, Waclaw RR, and Campbell K
- Subjects
- Animals, Cell Differentiation physiology, Embryo, Mammalian, Mice, Mice, Transgenic, Neural Stem Cells cytology, Homeodomain Proteins genetics, Interneurons cytology, Neurogenesis physiology, Olfactory Bulb embryology, Septum of Brain embryology
- Abstract
Background: Olfactory bulb (OB) interneurons are known to represent diverse neuronal subtypes, which are thought to originate from a number of telencephalic regions including the embryonic dorsal lateral ganglionic eminence (dLGE) and septum. These cells migrate rostrally toward the OB, where they then radially migrate to populate different OB layers including the granule cell layer (GCL) and the outer glomerular layer (GL). Although previous studies have attempted to investigate regional contributions to OB interneuron diversity, few genetic tools have been used to address this question at embryonic time points when the earliest populations are specified., Methods: In this study, we utilized Zic3-lacZ and Gsx2e-CIE transgenic mice as genetic fate-mapping tools to study OB interneuron contributions derived from septum and LGE, respectively. Moreover, to address the regional (i.e. septal) requirements of the homeobox gene Gsx2 for OB interneuron diversity, we conditionally inactivated Gsx2 in the septum, leaving it largely intact in the dLGE, by recombining the Gsx2 floxed allele using Olig2
Cre/+ mice., Results: Our fate mapping studies demonstrated that the dLGE and septum gave rise to OB interneuron subtypes differently. Notably, the embryonic septum was found to give rise largely to the calretinin+ (CR+ ) GL subtype, while the dLGE was more diverse, generating all major GL subpopulations as well as many GCL interneurons. Moreover, Gsx2 conditional mutants (cKOs), with septum but not dLGE recombination, showed impaired generation of CR+ interneurons within the OB GL. These Gsx2 cKOs exhibited reduced proliferation within the septal subventricular zone (SVZ), which correlated well with the reduced number of CR+ interneurons observed., Conclusions: Our findings indicate that the septum and LGE contribute differently to OB interneuron diversity. While the dLGE provides a wide range of OB interneuron subtypes, the septum is more restricted in its contribution to the CR+ subtype. Gsx2 is required in septal progenitors for the correct expansion of SVZ progenitors specified toward the CR+ subtype. Finally, the septum has been suggested to be the exclusive source of CR+ interneurons in postnatal studies. Our results here demonstrate that dLGE progenitors in the embryo also contribute to this OB neuronal subtype.- Published
- 2017
- Full Text
- View/download PDF
20. The transcription factor Olig2 is important for the biology of diffuse intrinsic pontine gliomas.
- Author
-
Anderson JL, Muraleedharan R, Oatman N, Klotter A, Sengupta S, Waclaw RR, Wu J, Drissi R, Miles L, Raabe EH, Weirauch ML, Fouladi M, Chow LM, Hoffman L, DeWire M, and Dasgupta B
- Subjects
- Animals, Brain Stem Neoplasms genetics, Child, Preschool, Gene Expression Regulation genetics, Humans, Male, Mice, Nerve Tissue Proteins metabolism, Astrocytoma metabolism, Brain Stem Neoplasms metabolism, Glioma metabolism, Oligodendrocyte Transcription Factor 2 metabolism
- Abstract
Background: Diffuse intrinsic pontine glioma (DIPG) is a high-grade brainstem glioma of children with dismal prognosis. There is no single unifying model about the cell of origin of DIPGs. Proliferating cells in the developing human and mouse pons, the site of DIPGs, express neural stem/progenitor cell (NPC) markers, including Sox2, nestin, vimentin, Olig2, and glial fibrillary acidic protein, in an overlapping and non-overlapping manner, suggesting progenitor cell heterogeneity in the pons. It is thought that during a restricted window of postnatal pons development, a differentiation block caused by genetic/epigenetic changes leads to unrestrained progenitor proliferation and DIPG development. Nearly 80% of DIPGs harbor a mutation in the H3F3A or the related HIST1H3B gene. Supporting the impaired differentiation model, NPCs derived from human induced pluripotent stem cells expressing the H3F3A mutation showed complete differentiation block. However, the mechanisms regulating an altered differentiation program in DIPG are unknown., Methods: We established syngeneic serum-dependent and independent primary DIPG lines, performed molecular characterization of DIPG lines in vitro and in an orthotopic xenograft model, and used small hairpin RNA to examine Olig2 function in DIPG., Results: The transcription factor Olig2 is highly expressed in 70%-80% of DIPGs. Here we report that Olig2 expression and DIPG differentiation are mutually exclusive events in vitro, and only DIPG cells that retained Olig2 in vitro formed robust Olig2-positive brainstem glioma with 100% penetrance in a xenograft model., Conclusion: Our results indicate Olig2 as an onco-requisite factor in DIPG and propose investigation of Olig2 target genes as novel candidates in DIPG therapy., (© The Author(s) 2017. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com)
- Published
- 2017
- Full Text
- View/download PDF
21. Foxo1 is a downstream effector of Isl1 in direct pathway striatal projection neuron development within the embryonic mouse telencephalon.
- Author
-
Waclaw RR, Ehrman LA, Merchan-Sala P, Kohli V, Nardini D, and Campbell K
- Subjects
- Age Factors, Animals, Basic-Leucine Zipper Transcription Factors metabolism, Cell Differentiation physiology, Corpus Striatum embryology, Corpus Striatum metabolism, Embryo, Mammalian, Forkhead Box Protein O1 genetics, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, LIM-Homeodomain Proteins genetics, Luminescent Proteins genetics, Luminescent Proteins metabolism, Mice, Mice, Transgenic, Neural Pathways physiology, Receptors, Dopamine D1 genetics, Receptors, Dopamine D1 metabolism, Trans-Activators metabolism, Transcription Factors genetics, Corpus Striatum cytology, Forkhead Box Protein O1 metabolism, Gene Expression Regulation, Developmental genetics, LIM-Homeodomain Proteins metabolism, Neurons metabolism, Signal Transduction genetics, Transcription Factors metabolism
- Abstract
Recent studies have shown that the LIM-homeodomain transcription factor Isl1 is required for the survival and differentiation of direct pathway striatonigral neurons during embryonic development. The downstream effectors of Isl1 in these processes are presently unknown. We show here that Foxo1, a transcription factor that has been implicated in cell survival, is expressed in striatal projection neurons (SPNs) that derive from the Isl1 lineage (i.e. direct pathway SPNs). Moreover, Isl1 conditional knockouts (cKOs) show a severe loss of Foxo1 expression at E15.5 with a modest recovery by E18.5. Although Foxo1 is enriched in the direct pathway SPNs at embryonic stages, it is expressed in both direct and indirect pathway SPNs at postnatal time points as evidenced by co-localization with EGFP in both Drd1-EGFP and Drd2-EGFP BAC transgenic mice. Foxo1 was not detected in striatal interneurons as marked by the transcription factor Nkx2.1. Conditional knockout of Foxo1 using Dlx5/6-CIE mice results in reduced expression of the SPN marker Darpp-32, as well as in the direct pathway SPN markers Ebf1 and Zfp521 within the embryonic striatum at E15.5. However, this phenotype improves in the conditional mutants by E18.5. Interestingly, the Foxo family members, Foxo3 and Foxo6, remain expressed at late embryonic stages in the Foxo1 cKOs unlike the Isl1 cKOs where Foxo1/3/6 as well as the Foxo1/3 target Bach2 are all reduced. Taken together, these findings suggest that Foxo-regulated pathways are downstream of Isl1 in the survival and/or differentiation of direct pathway SPNs., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
22. Characterization of a new Gsx2-cre line in the developing mouse telencephalon.
- Author
-
Qin S, Madhavan M, Waclaw RR, Nakafuku M, and Campbell K
- Subjects
- Amygdala growth & development, Animals, Ganglion Cysts pathology, Gene Expression Regulation, Developmental, Green Fluorescent Proteins biosynthesis, Integrases genetics, Mice, Mice, Transgenic, Neurons metabolism, Neurons pathology, Olfactory Bulb growth & development, Telencephalon metabolism, Ganglion Cysts genetics, Green Fluorescent Proteins genetics, Homeodomain Proteins genetics, Telencephalon growth & development
- Abstract
In this study, we generated a transgenic mouse line driving Cre and EGFP expression with two putative cis-regulatory modules (CRMs) (i.e., hs687 and hs678) upstream of the homeobox gene Gsx2 (formerly Gsh2), a critical gene for establishing lateral ganglionic eminence (LGE) identity. The combination of these two CRMs drives transgene expression within the endogenous Gsx2 expression domains along the anterior-posterior neuraxis. By crossing this transgenic line with the Rosa
tdTomato (Ai14) reporter mouse line, we observed a unique recombination pattern in the lateral ventral telencephalon, namely the LGE and the dorsal half of the medial GE (MGE), but not in the septum. We found robust recombination in many cell types derived from these embryonic regions, including olfactory bulb and amygdala interneurons and striatal projection neurons from the LGE, as well as cortical interneurons from the MGE and caudal GE (CGE). In summary, this transgenic mouse line represents a new tool for genetic manipulation in the LGE/CGE and the dorsal half of MGE., (© 2016 Wiley Periodicals, Inc.)- Published
- 2016
- Full Text
- View/download PDF
23. Concomitant inactivation of foxo3a and fancc or fancd2 reveals a two-tier protection from oxidative stress-induced hydrocephalus.
- Author
-
Li X, Li L, Li J, Sipple J, Schick J, Mehta PA, Davies SM, Dasgupta B, Waclaw RR, and Pang Q
- Subjects
- Animals, Fanconi Anemia genetics, Fanconi Anemia metabolism, Fanconi Anemia Complementation Group C Protein deficiency, Fanconi Anemia Complementation Group D2 Protein deficiency, Forkhead Box Protein O3, Forkhead Transcription Factors deficiency, Forkhead Transcription Factors genetics, Hydrocephalus genetics, Mice, Mice, Knockout, Reactive Oxygen Species metabolism, Fanconi Anemia Complementation Group C Protein metabolism, Fanconi Anemia Complementation Group D2 Protein metabolism, Forkhead Transcription Factors metabolism, Hydrocephalus metabolism, Oxidative Stress
- Abstract
Aims: This study seeks at investigating the cause of hydrocephalus, and at identifying therapeutic targets for the prevention of hydrocephalus., Results: In this study, we show that inactivation of the Foxo3a gene in two mouse models of Fanconi anemia (FA) leads to the development of hydrocephalus in late embryonic stage and after birth. More than 50% of Foxo3a(-/-) Fancc(-/-) or Foxo3a(-/-) Fancd2(-/-) mice die during embryonic development or within 6 months of life as a result of hydrocephalus characterized by cranial distortion, dilation of the ventricular system, reduced thickness of the cerebral cortex, and disorganization of the ependymal cilia and subcommissural organ. Combined deficiency of Foxo3a and Fancc or Fancd2 not only impairs the self-renewal capacity but also markedly increases the apoptosis of neural stem and progenitor cells (NSPCs), leading to defective neurogenesis. Increased accumulation of reactive oxygen species (ROS) and subsequently de-regulated mitosis and ultimately apoptosis in the neural stem or progenitor cells is identified as one of the potential mechanisms of congenital obstructive hydrocephalus., Innovation: The work unravels a two-tier protective mechanism for preventing oxidative stress-induced hydrocephalus., Conclusion: The deletion of Foxo3a in FA mice increased the accumulation of ROS and subsequently de-regulated mitosis and ultimately apoptosis in the NSPCs, leading to hydrocephalus development.
- Published
- 2014
- Full Text
- View/download PDF
24. The protein tyrosine phosphatase Shp2 is required for the generation of oligodendrocyte progenitor cells and myelination in the mouse telencephalon.
- Author
-
Ehrman LA, Nardini D, Ehrman S, Rizvi TA, Gulick J, Krenz M, Dasgupta B, Robbins J, Ratner N, Nakafuku M, and Waclaw RR
- Subjects
- Animals, Cell Differentiation physiology, Mice, Mice, Transgenic, Telencephalon cytology, Nerve Fibers, Myelinated enzymology, Oligodendroglia enzymology, Protein Tyrosine Phosphatase, Non-Receptor Type 11 biosynthesis, Stem Cells enzymology, Telencephalon embryology, Telencephalon enzymology
- Abstract
The protein tyrosine phosphatase Shp2 (PTPN11) is crucial for normal brain development and has been implicated in dorsal telencephalic neuronal and astroglia cell fate decisions. However, its roles in the ventral telencephalon and during oligodendrogenesis in the telencephalon remain largely unknown. Shp2 gain-of-function (GOF) mutations are observed in Noonan syndrome, a type of RASopathy associated with multiple phenotypes, including cardiovascular, craniofacial, and neurocognitive abnormalities. To gain insight into requirements for Shp2 (LOF) and the impact of abnormal Shp2 GOF mutations, we used a Shp2 conditional mutant allele (LOF) and a cre inducible Shp2-Q79R GOF transgenic mouse in combination with Olig2(cre/+) mice to target embryonic ventral telencephalic progenitors and the oligodendrocyte lineage. In the absence of Shp2 (LOF), neuronal cell types originating from progenitors in the ventral telencephalon were generated, but oligodendrocyte progenitor cell (OPC) generation was severely impaired. Late embryonic and postnatal Shp2 cKOs showed defects in the generation of OPCs throughout the telencephalon and subsequent reductions in white matter myelination. Conversely, transgenic expression of the Shp2 GOF Noonan syndrome mutation resulted in elevated OPC numbers in the embryo and postnatal brain. Interestingly, expression of this mutation negatively influenced myelination as mice displayed abnormal myelination and fewer myelinated axons in the white matter despite elevated OPC numbers. Increased proliferating OPCs and elevated MAPK activity were also observed during oligodendrogenesis after expression of Shp2 GOF mutation. These results support the notion that appropriate Shp2 activity levels control the number as well as the differentiation of oligodendrocytes during development.
- Published
- 2014
- Full Text
- View/download PDF
25. The LIM homeobox gene Isl1 is required for the correct development of the striatonigral pathway in the mouse.
- Author
-
Ehrman LA, Mu X, Waclaw RR, Yoshida Y, Vorhees CV, Klein WH, and Campbell K
- Subjects
- Animals, Behavior, Animal physiology, Cell Adhesion Molecules, Neuronal genetics, Cell Adhesion Molecules, Neuronal metabolism, Corpus Striatum cytology, Cytoskeletal Proteins, Glycoproteins genetics, Glycoproteins metabolism, Intracellular Signaling Peptides and Proteins, LIM-Homeodomain Proteins genetics, Membrane Glycoproteins, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, Mice, Knockout, Nerve Tissue Proteins genetics, Semaphorins, Substantia Nigra cytology, Transcription Factors genetics, Corpus Striatum embryology, LIM-Homeodomain Proteins metabolism, Nerve Tissue Proteins metabolism, Signal Transduction physiology, Substantia Nigra embryology, Transcription Factors metabolism
- Abstract
The mammalian striatum controls the output of the basal ganglia via two distinct efferent pathways, the direct (i.e., striatonigral) and the indirect (i.e., striatopallidal) pathways. The LIM homeodomain transcription factor Islet1 (Isl1) is expressed in a subpopulation of striatal progenitors; however, its specific role in striatal development remains unknown. Our genetic fate-mapping results show that Isl1-expressing progenitors give rise to striatal neurons belonging to the striatonigral pathway. Conditional inactivation of Isl1 in the telencephalon resulted in a smaller striatum with fewer striatonigral neurons and reduced projections to the substantia nigra. Additionally, conditional inactivation in the ventral forebrain (including both the telencephalon and diencephalon) revealed a unique role for Isl1 in diencephalic cells bordering the internal capsule for the normal development of the striatonigral pathway involving PlexinD1-Semaphorin 3e (Sema3e) signaling. Finally, Isl1 conditional mutants displayed a hyperlocomotion phenotype, and their locomotor response to psychostimulants was significantly blunted, indicating that the alterations in basal ganglia circuitry contribute to these mutant behaviors.
- Published
- 2013
- Full Text
- View/download PDF
26. The homeobox gene Gsx2 controls the timing of oligodendroglial fate specification in mouse lateral ganglionic eminence progenitors.
- Author
-
Chapman H, Waclaw RR, Pei Z, Nakafuku M, and Campbell K
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors metabolism, Cell Lineage, Green Fluorescent Proteins metabolism, Homeodomain Proteins genetics, Mice, Microscopy, Fluorescence, Mutation, Nerve Tissue Proteins metabolism, Neurons physiology, Oligodendrocyte Transcription Factor 2, Receptor, Platelet-Derived Growth Factor alpha metabolism, SOXE Transcription Factors metabolism, Stem Cells physiology, Telencephalon physiology, Time Factors, Ganglia growth & development, Gene Expression Regulation, Developmental, Homeodomain Proteins physiology, Oligodendroglia physiology
- Abstract
The homeobox gene Gsx2 has previously been shown to be required for the specification of distinct neuronal subtypes derived from lateral ganglionic eminence (LGE) progenitors at specific embryonic time points. However, its role in the subsequent generation of oligodendrocytes from these progenitors remains unclear. We have utilized conditional gain-of-function and loss-of-function approaches in order to elucidate the role of Gsx2 in the switch between neurogenesis and oligodendrogenesis within the embryonic ventral telencephalon. In the absence of Gsx2 expression, an increase in oligodendrocyte precursor cells (OPCs) with a concomitant decrease in neurogenesis is observed in the subventricular zone of the LGE at mid-stages of embryogenesis (i.e. E12.5-15.5), which subsequently leads to an increased number of Gsx2-derived OPCs within the adjacent mantle regions of the cortex before birth at E18.5. Moreover, using Olig2(cre) to conditionally inactivate Gsx2 throughout the ventral telencephalon with the exception of the dorsal (d)LGE, we found that the increase in cortical OPCs in Gsx2 germline mutants are derived from dLGE progenitors. We also show that Ascl1 is required for the expansion of these dLGE-derived OPCs in the cortex of Gsx2 mutants. Complementing these results, gain-of-function experiments in which Gsx2 was expressed throughout most of the late-stage embryonic telencephalon (i.e. E15.5-18.5) result in a significant decrease in the number of cortical OPCs. These results support the notion that high levels of Gsx2 suppress OPC specification in dLGE progenitors and that its downregulation is required for the transition from neurogenesis to oligodendrogenesis.
- Published
- 2013
- Full Text
- View/download PDF
27. Loss of Gsx1 and Gsx2 function rescues distinct phenotypes in Dlx1/2 mutants.
- Author
-
Wang B, Long JE, Flandin P, Pla R, Waclaw RR, Campbell K, and Rubenstein JL
- Subjects
- Animals, Brain embryology, Embryo, Mammalian, Fluorescent Antibody Technique, In Situ Hybridization, Mice, Mice, Mutant Strains, Phenotype, Brain physiology, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Neurogenesis physiology, Transcription Factors genetics
- Abstract
Mice lacking the Dlx1 and Dlx2 homeobox genes (Dlx1/2 mutants) have severe deficits in subpallial differentiation, including overexpression of the Gsx1 and Gsx2 homeobox genes. To investigate whether Gsx overexpression contributes to the Dlx1/2 mutant phenotypes, we made compound loss-of-function mutants. Eliminating Gsx2 function from the Dlx1/2 mutants rescued the increased expression of Ascl1 and Hes5 (Notch signaling mediators) and Olig2 (oligodendrogenesis mediator). In addition, Dlx1/2;Gsx2 mutants, like Dlx1/2;Ascl1 mutants, exacerbated the Gsx2 and Dlx1/2 patterning and differentiation phenotypes, particularly in the lateral ganglionic eminence (LGE) caudal ganglionic eminence (CGE), and septum, including loss of GAD1 expression. On the other hand, eliminating Gsx1 function from the Dlx1/2 mutants (Dlx1/2;Gsx1 mutants) did not severely exacerbate their phenotype; on the contrary, it resulted in a partial rescue of medial ganglionic eminence (MGE) properties, including interneuron migration to the cortex. Thus, despite their redundant properties, Gsx1 and -2 have distinct interactions with Dlx1 and -2. Gsx2 interaction is strongest in the LGE, CGE, and septum, whereas the Gsx1 interaction is strongest in the MGE. From these studies, and earlier studies, we present a model of the transcriptional network that regulates early steps of subcortical development., (Copyright © 2012 Wiley Periodicals, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
28. Tangentially migrating transient glutamatergic neurons control neurogenesis and maintenance of cerebral cortical progenitor pools.
- Author
-
Teissier A, Waclaw RR, Griveau A, Campbell K, and Pierani A
- Subjects
- Age Factors, Animals, Animals, Newborn, Basic Helix-Loop-Helix Transcription Factors genetics, Body Patterning genetics, Bromodeoxyuridine metabolism, Cadherins metabolism, Cell Cycle genetics, Cell Differentiation, Cell Movement genetics, Cell Proliferation, Cerebral Ventricles cytology, Cerebral Ventricles embryology, Cerebral Ventricles growth & development, Embryo, Mammalian, Gene Expression Regulation, Developmental, Green Fluorescent Proteins genetics, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, MEF2 Transcription Factors, Mice, Mice, Inbred C57BL, Mice, Transgenic, Mutation genetics, Myogenic Regulatory Factors metabolism, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Nuclear Proteins metabolism, Nuclear Receptor Subfamily 2, Group C, Member 1 metabolism, Repressor Proteins metabolism, Cell Movement physiology, Cerebral Cortex cytology, Cerebral Cortex embryology, Cerebral Cortex growth & development, Glutamates metabolism, Neural Stem Cells physiology, Neurogenesis genetics, Neurons physiology
- Abstract
The relative contribution of intrinsic and extrinsic cues in the regulation of cortical neurogenesis remains a crucial challenge in developmental neurobiology. We previously reported that a transient population of glutamatergic neurons, the cortical plate (CP) transient neurons, migrates from the ventral pallium (VP) over long distances and participate in neocortical development. Here, we show that the genetic ablation of this population leads to a reduction in the number of cortical neurons especially fated to superficial layers. These defects result from precocious neurogenesis followed by a depletion of the progenitor pools. Notably, these changes progress from caudolateral to rostrodorsal pallial territories between E12.5 and E14.5 along the expected trajectory of the ablated cells. Conversely, we describe enhanced proliferation resulting in an increase in the number of cortical neurons in the Gsx2 mutants which present an expansion of the VP and a higher number of CP transient neurons migrating into the pallium. Our findings indicate that these neurons act to maintain the proliferative state of neocortical progenitors and delay differentiation during their migration from extraneocortical regions and, thus, participate in the extrinsic control of cortical neuronal numbers.
- Published
- 2012
- Full Text
- View/download PDF
29. Ascl1 participates in Cajal-Retzius cell development in the neocortex.
- Author
-
Dixit R, Zimmer C, Waclaw RR, Mattar P, Shaker T, Kovach C, Logan C, Campbell K, Guillemot F, and Schuurmans C
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors metabolism, Cell Lineage, Immunohistochemistry, In Situ Hybridization, Mice, Mice, Transgenic, Neocortex cytology, Neocortex metabolism, Neural Stem Cells metabolism, Neurons metabolism, Reelin Protein, Basic Helix-Loop-Helix Transcription Factors genetics, Cell Differentiation genetics, Neocortex embryology, Neural Stem Cells cytology, Neurogenesis genetics, Neurons cytology
- Abstract
Cajal-Retzius cells are essential pioneer neurons that guide neuronal migration in the developing neocortex. During development, Cajal-Retzius cells arise from distinct progenitor domains that line the margins of the dorsal telencephalon, or pallium. Here, we show that the proneural gene Ascl1 is expressed in Cajal-Retzius cell progenitors in the pallial septum, ventral pallium, and cortical hem. Using a short-term lineage trace, we demonstrate that it is primarily the Ascl1-expressing progenitors in the pallial septum and ventral pallium that differentiate into Cajal-Retzius cells. Accordingly, we found a small, albeit significant reduction in the number of Reelin(+) and Trp73(+) Cajal-Retzius cells in the Ascl1(-/-) neocortex. Conversely, using a gain-of-function approach, we found that Ascl1 induces the expression of both Reelin, a Cajal-Retzius marker, and Tbr1, a marker of pallial-derived neurons, in a subset of early-stage pallial progenitors, an activity that declines over developmental time. Taken together, our data indicate that the proneural gene Ascl1 is required and sufficient to promote the differentiation of a subset of Cajal-Retzius neurons during early neocortical development. Notably, this is the first study that reports a function for Ascl1 in the pallium, as this gene is best known for its role in specifying subpallial neuronal identities.
- Published
- 2011
- Full Text
- View/download PDF
30. Spatial and temporal requirements for sonic hedgehog in the regulation of thalamic interneuron identity.
- Author
-
Jeong Y, Dolson DK, Waclaw RR, Matise MP, Sussel L, Campbell K, Kaestner KH, and Epstein DJ
- Subjects
- Animals, Diencephalon cytology, Diencephalon embryology, Diencephalon metabolism, Gene Expression Regulation, Developmental, Hedgehog Proteins genetics, Immunohistochemistry, In Situ Hybridization, Mice, Mice, Mutant Strains, Thalamus embryology, Hedgehog Proteins metabolism, Interneurons cytology, Interneurons metabolism, Thalamus cytology, Thalamus metabolism
- Abstract
In caudal regions of the diencephalon, sonic hedgehog (Shh) is expressed in the ventral midline of prosomeres 1-3 (p1-p3), which underlie the pretectum, thalamus and prethalamus, respectively. Shh is also expressed in the zona limitans intrathalamica (zli), a dorsally projecting spike that forms at the p2-p3 boundary. The presence of two Shh signaling centers in the thalamus has made it difficult to determine the specific roles of either one in regional patterning and neuronal fate specification. To investigate the requirement of Shh from a focal source of expression in the ventral midline of the diencephalon, we used a newly generated mouse line carrying a targeted deletion of the 525 bp intronic sequence mediating Shh brain enhancer-1 (SBE1) activity. In SBE1 mutant mice, Shh transcription was initiated but not maintained in the ventral midline of the rostral midbrain and caudal diencephalon, yet expression in the zli was unaffected. In the absence of ventral midline Shh, rostral thalamic progenitors (pTH-R) adopted the molecular profile of a more caudal thalamic subtype (pTH-C). Surprisingly, despite their early mis-specification, neurons derived from the pTH-R domain continued to migrate to their proper thalamic nucleus, extended axons along their normal trajectory and expressed some, but not all, of their terminal differentiation markers. Our results, and those of others, suggest a model whereby Shh signaling from distinct spatial and temporal domains in the diencephalon exhibits unique and overlapping functions in the development of discrete classes of thalamic interneurons.
- Published
- 2011
- Full Text
- View/download PDF
31. Developmental origin of the neuronal subtypes that comprise the amygdalar fear circuit in the mouse.
- Author
-
Waclaw RR, Ehrman LA, Pierani A, and Campbell K
- Subjects
- Age Factors, Analysis of Variance, Animals, Animals, Newborn, Behavior, Animal, Brain Mapping, DNA-Binding Proteins genetics, Embryo, Mammalian, Gene Expression Regulation, Developmental genetics, Green Fluorescent Proteins genetics, Homeodomain Proteins genetics, LIM-Homeodomain Proteins, Mice, Mice, Transgenic, Mutation genetics, Nerve Tissue Proteins metabolism, Neural Pathways physiology, Neurons classification, Transcription Factors genetics, Amygdala cytology, Amygdala growth & development, Fear, Gene Expression Regulation, Developmental physiology, Neurons physiology
- Abstract
We have taken a genetic-based fate-mapping approach to determine the specific contributions of telencephalic progenitors to the structures that comprise the amygdalar fear circuit including the central (CA), lateral (LA), and basolateral (BLA) amygdala. Our data indicate that progenitors in the ventral pallium (VP) contribute projection neurons to the LA and BLA but not the CA. Rather, the CA appears to derive, at least in part, from progenitors located in the ventral lateral ganglionic eminence (vLGE). Diverse groups of interneurons populate these amygdalar nuclei, and as predicted our data support the notion that they originate from subpallial progenitors. A rather specific population of amygdalar interneurons, the intercalated cells (ITCs), is known to play a fundamental role in fear-related behaviors. However, no information on their specific origin has, as yet, been provided. Our findings suggest that the ITCs arise from the dorsal lateral ganglionic eminence (dLGE) and migrate in the lateral migratory stream to populate the paracapsular regions as well as the main intercalated mass of the amygdala (IA). Germ-line Gsx2 mutants are known to exhibit an expansion of the VP into the LGE and a concomitant reduction in the dLGE and vLGE. Accordingly, Gsx2 conditional mutants display a significantly enlarged LA and a significant reduction in ITCs both within the paracapsular regions and the IA. Additional support for a dLGE origin of the ITCs was obtained in conditional mutants of the dLGE gene Sp8. Thus, our findings indicate diverse origins for the neuronal components that comprise the amygdalar fear circuit.
- Published
- 2010
- Full Text
- View/download PDF
32. Sonic hedgehog signaling confers ventral telencephalic progenitors with distinct cortical interneuron fates.
- Author
-
Xu Q, Guo L, Moore H, Waclaw RR, Campbell K, and Anderson SA
- Subjects
- Animals, Animals, Genetically Modified, Calbindin 2, Embryo, Mammalian, Eye Proteins genetics, Eye Proteins metabolism, Gene Expression Regulation, Developmental physiology, Green Fluorescent Proteins genetics, Hedgehog Proteins genetics, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Mice, Mice, Inbred C57BL, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism, Parvalbumins metabolism, S100 Calcium Binding Protein G metabolism, Somatostatin metabolism, Thyroid Nuclear Factor 1, Transcription Factors genetics, Transcription Factors metabolism, Homeobox Protein SIX3, Cerebral Cortex cytology, Embryonic Stem Cells physiology, Hedgehog Proteins metabolism, Interneurons physiology, Signal Transduction physiology, Telencephalon cytology
- Abstract
Interneurons in the cerebral cortex regulate cortical functions through the actions of distinct subgroups that express parvalbumin, somatostatin, or calretinin. The genesis of the first two subgroups requires the expression of NKX2.1, which is maintained by SHH signaling during neurogenesis. In this paper, we report that mosaic elimination in the medial ganglionic eminence (MGE) of Smo, a key effector of SHH signaling, reveals that MGE progenitors retain a remarkable degree of plasticity during the neurogenic period. SHH signaling prevents the upregulation of GSX2 and conversion of some MGE progenitors to a caudal ganglionic eminence-like, bipolar calretinin-expressing cell fate that is promoted by GSX2. In addition, a higher level of SHH signaling promotes the generation of the somatostatin-expressing interneuron at the expense of parvalbumin-expressing subgroup. These results indicate that cortical interneuron diversity, a major determinant of cortical function, is critically influenced by differential levels of SHH signaling within the ventral telencephalon., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
33. Regional control of cortical lamination.
- Author
-
Waclaw RR and Campbell K
- Subjects
- Animals, Humans, Visual Pathways physiology, Cerebral Cortex cytology, Cerebral Cortex physiology, Neurons physiology, Transcription Factor AP-2 physiology
- Published
- 2009
- Full Text
- View/download PDF
34. Distinct temporal requirements for the homeobox gene Gsx2 in specifying striatal and olfactory bulb neuronal fates.
- Author
-
Waclaw RR, Wang B, Pei Z, Ehrman LA, and Campbell K
- Subjects
- Animals, Corpus Striatum embryology, Female, Homeodomain Proteins biosynthesis, Homeodomain Proteins physiology, Interneurons physiology, Mice, Mice, Knockout, Mice, Transgenic, Mutagenesis, Site-Directed, Olfactory Bulb embryology, Pregnancy, Time Factors, Corpus Striatum pathology, Genes, Homeobox physiology, Homeodomain Proteins genetics, Interneurons pathology, Neurogenesis genetics, Olfactory Bulb pathology
- Abstract
The homeobox gene Gsx2 (formerly Gsh2) is known to be required for striatal and olfactory bulb neurogenesis; however, its specific role in the specification of these two neuronal subtypes remains unclear. To address this, we have employed a temporally regulated gain-of-function approach in transgenic mice and found that misexpression of Gsx2 at early stages of telencephalic neurogenesis favors the specification of striatal projection neuron identity over that of olfactory bulb interneurons. In contrast, delayed activation of the Gsx2 transgene until later stages exclusively promotes olfactory bulb interneuron identity. In a complementary approach, we have conditionally inactivated Gsx2 in a temporally progressive manner. Unlike germline Gsx2 mutants, which exhibit severe alterations in both striatal and olfactory bulb neurogenesis at birth, the conditional mutants exhibited defects restricted to olfactory bulb interneurons. These results demonstrate that Gsx2 specifies striatal projection neuron and olfactory bulb interneuron identity at distinct time points during telencephalic neurogenesis.
- Published
- 2009
- Full Text
- View/download PDF
35. Ascl1 is a required downstream effector of Gsx gene function in the embryonic mouse telencephalon.
- Author
-
Wang B, Waclaw RR, Allen ZJ 2nd, Guillemot F, and Campbell K
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors deficiency, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors physiology, Corpus Striatum embryology, Embryo, Mammalian, Homeodomain Proteins metabolism, Immunohistochemistry, In Situ Hybridization, Mice, Mice, Transgenic, Mutation, Polymerase Chain Reaction, Telencephalon embryology, Basic Helix-Loop-Helix Transcription Factors metabolism, Corpus Striatum metabolism, Homeodomain Proteins genetics, Telencephalon metabolism
- Abstract
Background: The homeobox gene Gsx2 (formerly Gsh2) is known to regulate patterning in the lateral ganglionic eminence (LGE) of the embryonic telencephalon. In its absence, the closely related gene Gsx1 (previously known as Gsh1) can partially compensate in the patterning and differentiation of ventral telencephalic structures, such as the striatum. However, the cellular and molecular mechanisms underlying this compensation remain unclear., Results: We show here that in the Gsx2 mutants Gsx1 is expressed in only a subset of the ventral telencephalic progenitors that normally express Gsx2. Based on the similarities in the expression of Gsx1 and Ascl1 (Mash1) within the Gsx2 mutant LGE, we examined whether Ascl1 plays an integral part in the Gsx1-based recovery. Ascl1 mutants show only modest alterations in striatal development; however, in Gsx2;Ascl1 double mutants, striatal development is severely affected, similar to that seen in the Gsx1;Gsx2 double mutants. This is despite the fact that Gsx1 is expressed, and even expands, in the Gsx2;Ascl1 mutant LGE, comparable to that seen in the Gsx2 mutant. Finally, Notch signaling has recently been suggested to be required for normal striatal development. In spite of the fact that Notch signaling is severely disrupted in Ascl1 mutants, it actually appears to be improved in the Gsx2;Ascl1 double mutants., Conclusion: These results, therefore, reveal a non-proneural requirement of Ascl1 that together with Gsx1 compensates for the loss of Gsx2 in a subset of LGE progenitors.
- Published
- 2009
- Full Text
- View/download PDF
36. Molecular identity of olfactory bulb interneurons: transcriptional codes of periglomerular neuron subtypes.
- Author
-
Allen ZJ 2nd, Waclaw RR, Colbert MC, and Campbell K
- Subjects
- Animals, Calbindin 2, Calbindins, DNA-Binding Proteins metabolism, Dopamine metabolism, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Interneurons cytology, Mice, Mice, Transgenic, S100 Calcium Binding Protein G metabolism, Stem Cells metabolism, Transcription, Genetic, Interneurons metabolism, Olfactory Bulb cytology, Olfactory Bulb metabolism, Transcription Factors metabolism
- Abstract
Interneurons in the glomerular layer (GL) of the olfactory bulb represent a diverse set of cells, which can be identified by distinct expression of different neurotransmitters as well as calcium binding proteins. Using genetic based fate mapping, we show here that at least three of these different interneurons subtypes (i.e. dopaminergic, calbindin- and calretinin-expressing) derive from cells that express the homeobox genes Dlx5/6. The transcription factors ER81, Meis2, Pax6 and Sp8 have all been implicated in olfactory bulb interneuron development and each of these can be observed in Dlx5/6-derived periglomerular cells. Conversely, the T-box factors Tbr1 and Tbx21, which mark olfactory bulb projection neurons, are not expressed in the Dlx5/6-derived periglomerular cells. While the interneuron subtypes that are marked by Pax6 and Sp8 have been described, little information exists as to the specific subtypes that express ER81 or Meis2. We show here that ER81 is expressed in dopaminergic cells and in a subset of calretinin-expressing cells in the GL. Meis2 is found in dopaminergic and calbindin-expressing cells as well as in a subpopulation of the calretinin-expressing interneurons of the glomerular layer. These findings suggest that distinct transcriptional codes may underlie the differentiation of specific olfactory bulb interneuron subtypes.
- Published
- 2007
- Full Text
- View/download PDF
37. Rac1 controls the formation of midline commissures and the competency of tangential migration in ventral telencephalic neurons.
- Author
-
Chen L, Liao G, Waclaw RR, Burns KA, Linquist D, Campbell K, Zheng Y, and Kuan CY
- Subjects
- Animals, Axons physiology, Cell Differentiation genetics, Cell Differentiation physiology, Cell Movement genetics, Cells, Cultured, Female, Mice, Mice, Knockout, Mice, Transgenic, Neurons cytology, Neuropeptides genetics, Pregnancy, Telencephalon cytology, rac GTP-Binding Proteins genetics, rac1 GTP-Binding Protein, Cell Movement physiology, Neurons physiology, Neuropeptides physiology, Telencephalon embryology, Telencephalon physiology, rac GTP-Binding Proteins physiology
- Abstract
Previous studies using dominant-mutant constructs have implicated Rac1 GTPase in neuritogenesis and neuronal migration. However, overexpression of dominant mutants generally blocks Rho-GTPase activity; thus, it may not reveal the specific or physiological functions of Rac1. To address this issue, we have applied a conditional gene-targeting strategy, using Foxg1-Cre mice to delete Rac1 in the ventricular zone (VZ) of telencephalon and Dlx5/6-Cre-IRES (internal ribosomal entry site)-EGFP (enhanced green fluorescent protein) (Dlx5/6-CIE) in the subventricular zone (SVZ) of ventral telencephalon, respectively. Surprisingly, the deletion of Rac1 in VZ progenitors did not prevent axonal outgrowth of telencephalic neurons. However, the anterior commissure was absent, and the corpus callosal as well as hippocampal commissural axons failed to cross the midline in Rac1/Foxg1-Cre knock-out embryos. The thalamocortical and corticothalamic axons also showed defasciculation or projection defects. These results suggest that Rac1 controls axon guidance rather than neuritogenesis. In addition, although Rac1/Foxg1-Cre knock-out embryos showed delayed radial migration of cortical projection neurons and severe impairment of tangential migration by the ventral telencephalon-derived interneurons, deletion of Rac1 in the SVZ by Dlx5/6-CIE mice produced no discernible defects in tangential migration. These contrasting effects of Rac1 deletion on tangential migration suggest that Rac1 is dispensable for cellular motility per se during neuronal migration. Together, these results underscore the challenge of deciphering the biological functions of Rac1, and Rho-GTPases in general, during mammalian brain development. Moreover, they indicate that Rac1 has a critical role in axon guidance and in acquisition of migratory competency during differentiation of the progenitors for the ventral telencephalon-derived interneurons.
- Published
- 2007
- Full Text
- View/download PDF
38. The zinc finger transcription factor Sp8 regulates the generation and diversity of olfactory bulb interneurons.
- Author
-
Waclaw RR, Allen ZJ 2nd, Bell SM, Erdélyi F, Szabó G, Potter SS, and Campbell K
- Subjects
- Animals, Animals, Newborn, Bromodeoxyuridine metabolism, Calbindin 2, Calbindins, Cell Count methods, Cell Death genetics, Cell Differentiation physiology, Cell Movement physiology, DNA-Binding Proteins deficiency, Embryo, Mammalian, Fluorescent Antibody Technique methods, Glutamate Decarboxylase metabolism, Green Fluorescent Proteins metabolism, Homeodomain Proteins genetics, Homeodomain Proteins physiology, In Situ Hybridization methods, In Situ Nick-End Labeling, Interneurons classification, Isoenzymes metabolism, Mice, Mice, Knockout, Models, Biological, PAX2 Transcription Factor genetics, S100 Calcium Binding Protein G metabolism, Transcription Factors deficiency, Tyrosine 3-Monooxygenase metabolism, gamma-Aminobutyric Acid metabolism, DNA-Binding Proteins physiology, Gene Expression Regulation, Developmental physiology, Interneurons physiology, Olfactory Bulb cytology, Olfactory Bulb embryology, Olfactory Bulb growth & development, Transcription Factors physiology
- Abstract
The molecular mechanisms that regulate the production and diversity of olfactory bulb interneurons remain poorly understood. With the exception of the GABAergic/dopaminergic subtype in the glomerular layer, no information exists concerning the generation of the other subtypes. Here we show that the recently identified zinc finger transcription factor Sp8 is expressed in neurogenic regions, which give rise to olfactory bulb interneurons at embryonic and postnatal time points and remains expressed in the calretinin-expressing and GABAergic/nondopaminergic interneurons of the glomerular layer. Conditional inactivation of Sp8 in the embryonic ventral telencephalon reveals a requirement for the normal generation of these interneuron subtypes. Sp8 conditional mutants exhibit an increase in cell death within the lateral ganglionic eminence and rostral migratory stream. Moreover, mutant neuroblasts/interneurons are misspecified and display abnormal migration patterns in the olfactory bulb, indicating that Sp8 contributes to olfactory bulb interneuron diversity by regulating the survival, migration, and molecular specification of neuroblasts/interneurons.
- Published
- 2006
- Full Text
- View/download PDF
39. The homeobox gene Gsh2 is required for retinoid production in the embryonic mouse telencephalon.
- Author
-
Waclaw RR, Wang B, and Campbell K
- Subjects
- Animals, Cell Differentiation drug effects, Cell Differentiation physiology, Eye Proteins, Homeodomain Proteins genetics, Mice, Mutation, Neurons drug effects, Neurons physiology, PAX6 Transcription Factor, Paired Box Transcription Factors, Repressor Proteins, Retinoids pharmacology, Telencephalon embryology, Homeodomain Proteins metabolism, Retinoids biosynthesis, Telencephalon metabolism
- Abstract
We have examined the role of the homeobox gene Gsh2 in retinoid production and signaling within the ventral telencephalon of mouse embryos. Gsh2 mutants exhibit altered ventral telencephalic development, including a smaller striatum with fewer DARPP-32 neurons than wild types. We show that the expression of the retinoic acid (RA) synthesis enzyme, retinaldehyde dehydrogenase 3 (Raldh3, also known as Aldh1a3), is reduced in the lateral ganglionic eminence (LGE) of Gsh2 mutants. Moreover, using a retinoid reporter cell assay, we found that retinoid production in the Gsh2 mutants is markedly reduced. The striatal defects in Gsh2 mutants are thought to result from ectopic expression of Pax6 in the LGE. Previously, we had shown that removal of Pax6 from the Gsh2 mutant background improves the molecular identity of the LGE in these double mutants; however, Raldh3 expression is not improved. The Pax6;Gsh2 double mutants possess a larger striatum than the Gsh2 mutants, but the disproportionate reduction in DARPP-32 neurons is not improved. These findings suggest that reduced retinoid production in the Gsh2 mutant contributes to the striatal differentiation defects. As RA promotes the expression of DARPP-32 in differentiating LGE cells in vitro, we examined whether exogenous RA can improve striatal neuron differentiation in the Gsh2 mutants. Indeed, RA supplementation of Gsh2 mutants, during the period of striatal neurogenesis, results in a significant increase in DARPP-32 expression. Thus, in addition to the previously described role for Gsh2 to maintain correct molecular identity in the LGE, our results demonstrate a novel requirement of this gene for retinoid production within the ventral telencephalon.
- Published
- 2004
- Full Text
- View/download PDF
40. Patterns of expression of cyclins A, B1, D, E and cdk 2 in preimplantation mouse embryos.
- Author
-
Waclaw RR and Chatot CL
- Subjects
- Animals, Blastocyst cytology, Cell Cycle physiology, Cyclin B1, Cyclin D, Cyclin-Dependent Kinase 2, Female, Fluorescent Antibody Technique, Immunohistochemistry, Mice, Zona Pellucida physiology, Blastocyst physiology, CDC2-CDC28 Kinases metabolism, Cyclin A metabolism, Cyclin B metabolism, Cyclin E metabolism, Cyclins metabolism
- Abstract
Cell cycle regulatory proteins have been characterized in somatic cells and exhibit phase-specific expression patterns. Changes in expression of these regulatory proteins have not been clearly characterized in early preimplantation mouse embryos. This study utilized indirect immunofluorescence to determine the expression pattern of G1/S phase cyclins D and E; S, G2/M phase cyclins A and B1, and cdk 2 during the first three cell cycles of mouse embryo development. Cyclin D demonstrated low expression throughout the first cell cycle but had a somatic-like pattern of expression in cycles 2 and 3 with peak expression at G1 declining through S phase to a low during G2. Cyclin E was present at peak levels in G1 declining through S to a low in G2 during both the first and third cell cycles, but remained at moderate levels during the entire second cell cycle. Cyclin A was maintained at moderate levels throughout the first two cell cycles but showed a somatic-like pattern with a low level in G1 increasing during S phase with peak levels during G2 of the third cell cycle. Cyclin B consistently demonstrated a pattern opposite to a somatic G2/M cyclin, with peak levels in G1 declining through S phase to a low in G2 during each of the three cell cycles examined. Cdk 2 was present at consistent levels during G1 and S phases of all three cell cycles declining slightly in G2.
- Published
- 2004
- Full Text
- View/download PDF
41. Sp8 is crucial for limb outgrowth and neuropore closure.
- Author
-
Bell SM, Schreiner CM, Waclaw RR, Campbell K, Potter SS, and Scott WJ
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Body Patterning, DNA genetics, Gene Targeting, In Situ Hybridization, Mice, Mice, Knockout, Phenotype, Signal Transduction, Transcription Factors genetics, Zinc Fingers genetics, Zinc Fingers physiology, Extremities embryology, Transcription Factors physiology
- Abstract
In this report we describe the developmental expression and function of Sp8, a member of the Sp family of zinc finger transcription factors, and provide evidence that the legless transgene insertional mutant is a hypomorphic allele of the Sp8 gene. Sp8 is expressed during embryogenesis in the forming apical ectodermal ridge (AER), restricted regions of the central nervous system, and tail bud. Targeted deletion of the Sp8 gene gives a striking phenotype, with severe truncation of both forelimbs and hindlimbs, absent tail, as well as defects in anterior and posterior neuropore closure leading to exencephaly and spina bifida. Outgrowth of the limb depends on formation of the AER, a signaling center that forms at the limb bud apex. In Sp8 mutants, the AER precursor cells are induced and initially express multiple appropriate marker genes, but expression of these genes is not maintained and progression to a mature AER is blocked. These observations indicate that Sp8 functions downstream of Wnt3, Fgf10, and Bmpr1a in the signaling cascade that mediates AER formation.
- Published
- 2003
- Full Text
- View/download PDF
42. NFATc3 and NFATc4 are required for cardiac development and mitochondrial function.
- Author
-
Bushdid PB, Osinska H, Waclaw RR, Molkentin JD, and Yutzey KE
- Subjects
- Animals, Bromodeoxyuridine metabolism, Cell Division genetics, DNA-Binding Proteins metabolism, Embryo, Mammalian abnormalities, Embryo, Mammalian metabolism, Embryonic and Fetal Development genetics, Embryonic and Fetal Development physiology, Female, Fetal Heart abnormalities, Fetal Heart metabolism, Gene Expression Regulation, Developmental, In Situ Hybridization, Male, Mice, Mice, Mutant Strains, Mice, Transgenic, Microscopy, Electron, Mitochondria ultrastructure, Mutation, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, NFATC Transcription Factors, Time Factors, Transcription Factors metabolism, DNA-Binding Proteins genetics, Fetal Heart embryology, Mitochondria physiology, Transcription Factors genetics
- Abstract
Activation of the nuclear factor of activated T-cell (NFAT) family of transcription factors is associated with changes in gene expression and myocyte function in adult cardiac and skeletal muscle. However, the role of NFATs in normal embryonic heart development is not well characterized. In this report, the function of NFATc3 and NFATc4 in embryonic heart development was examined in mice with targeted disruption of both nfatc3 and nfatc4 genes. The nfatc3-/-nfatc4-/- mice demonstrate embryonic lethality after embryonic day 10.5 and have thin ventricles, pericardial effusion, and a reduction in ventricular myocyte proliferation. Cardiac mitochondria are swollen with abnormal cristae, indicative of metabolic failure, but hallmarks of apoptosis are not evident. Furthermore, enzymatic activity of complex II and IV of the respiratory chain and mitochondrial oxidative activity are reduced in nfatc3-/-nfatc4-/- cardiomyocytes. Cardiac-specific expression of constitutively active NFATc4 in nfatc3-/-nfatc4-/- embryos prolongs embryonic viability to embryonic day 12 and preserves ventricular myocyte proliferation, compact zone density, and trabecular formation. The rescued embryos also maintain cardiac mitochondrial ultrastructure and complex II enzyme activity. Together, these data support the hypothesis that loss of NFAT activity in the heart results in a deficiency in mitochondrial energy metabolism required for cardiac morphogenesis and function.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.