Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
EMBO Rep ; 23(9): e54078, 2022 09 05.
Article in English | MEDLINE | ID: mdl-35861333

ABSTRACT

According to the current consensus, murine neural stem cells (NSCs) apically contacting the lateral ventricle generate differentiated progenitors by rare asymmetric divisions or by relocating to the basal side of the ventricular-subventricular zone (V-SVZ). Both processes will ultimately lead to the generation of adult-born olfactory bulb (OB) interneurons. In contrast to this view, we here find that adult-born OB interneurons largely derive from an additional NSC-type resident in the basal V-SVZ. Despite being both capable of self-renewal and long-term quiescence, apical and basal NSCs differ in Nestin expression, primary cilia extension and frequency of cell division. The expression of Notch-related genes also differs between the two NSC groups, and Notch activation is greatest in apical NSCs. Apical downregulation of Notch-effector Hes1 decreases Notch activation while increasing proliferation across the niche and neurogenesis from apical NSCs. Underscoring their different roles in neurogenesis, lactation-dependent increase in neurogenesis is paralleled by extra activation of basal but not apical NSCs. Thus, basal NSCs support OB neurogenesis, whereas apical NSCs impart Notch-mediated lateral inhibition across the V-SVZ.


Subject(s)
Lateral Ventricles , Neural Stem Cells , Animals , Cell Differentiation/genetics , Female , Lateral Ventricles/metabolism , Mice , Neural Stem Cells/metabolism , Neurogenesis/genetics , Olfactory Bulb/metabolism
2.
Int J Mol Sci ; 21(11)2020 May 29.
Article in English | MEDLINE | ID: mdl-32486089

ABSTRACT

BRCA1/2 variants are prognostic biomarkers for hereditary breast and/or ovarian cancer (HBOC) syndrome and predictive biomarkers for PARP inhibition. In this study, we benchmarked the classification of BRCA1/2 variants from patients with HBOC-related cancer using MH BRCA, a novel computational technology that combines the ACMG guidelines with expert-curated variant annotations. Evaluation of BRCA1/2 variants (n = 1040) taken from four HBOC studies showed strong concordance within the pathogenic (98.1%) subset. Comparison of MH BRCA's ACMG classification to ClinVar submitter content from ENIGMA, the international consortium of investigators on the clinical significance of BRCA1/2 variants, the ARUP laboratories, a clinical testing lab of the University of UTAH, and the German Cancer Consortium showed 99.98% concordance (4975 out of 4976 variants) in the pathogenic subset. In our patient cohort, refinement of patients with variants of unknown significance reduced the uncertainty of cancer-predisposing syndromes by 64.7% and identified three cases with potential family risk to HBOC due to a likely pathogenic variant BRCA1 p.V1653L (NM_007294.3:c.4957G > T; rs80357261). To assess whether classification results predict PARP inhibitor efficacy, contextualization with functional impact information on DNA repair activity were performed, using MH Guide. We found a strong correlation between treatment efficacy association and MH BRCA classifications. Importantly, low efficacy to PARP inhibition was predicted in 3.95% of pathogenic variants from four examined HBOC studies and our patient cohort, indicating the clinical relevance of the consolidated variant interpretation.


Subject(s)
Breast Neoplasms/genetics , Genes, BRCA1 , Genes, BRCA2 , Ovarian Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Biomarkers, Tumor/genetics , Breast Neoplasms/blood , Breast Neoplasms/diagnosis , Computational Biology , DNA Repair , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genetic Testing , Genetic Variation , Germ-Line Mutation , Germany , Humans , Japan , Male , Ovarian Neoplasms/blood , Ovarian Neoplasms/diagnosis , Prostatic Neoplasms/blood , Prostatic Neoplasms/genetics , Reproducibility of Results , Retrospective Studies
3.
Stem Cells ; 35(12): 2417-2429, 2017 12.
Article in English | MEDLINE | ID: mdl-28869691

ABSTRACT

The glycoprotein Prominin-1 and the carbohydrate Lewis X stage-specific embryonic antigen 1 (LeX-SSEA1) both have been extensively used as cell surface markers to purify neural stem cells (NSCs). While Prominin-1 labels a specialized membrane region in NSCs and ependymal cells, the specificity of LeX-SSEA1 expression and its biological significance are still unknown. To address these issues, we have here monitored the expression of the carbohydrate in neonatal and adult NSCs and in their progeny. Our results show that the percentage of immunopositive cells and the levels of LeX-SSEA1 immunoreactivity both increase with postnatal age across all stages of the neural lineage. This is associated with decreased proliferation in precursors including NSCs, which accumulate the carbohydrate at the cell surface while remaining quiescent. Exposure of precursors to bone morphogenetic protein (BMP) increases LEX-SSEA1 expression, which promotes cell cycle withdrawal by a mechanism involving LeX-SSEA1-mediated interaction at the cell surface. Conversely, interference with either BMP signaling or with LeX-SSEA1 promotes proliferation to a similar degree. Thus, in the postnatal germinal niche, the expression of LeX-SSEA1 increases with age and exposure to BMP signaling, thereby downregulating the proliferation of subependymal zone precursors including NSCs. Stem Cells 2017;35:2417-2429.


Subject(s)
Lewis X Antigen/metabolism , Neural Stem Cells/metabolism , AC133 Antigen/genetics , AC133 Antigen/metabolism , Animals , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Proliferation/genetics , Cell Proliferation/physiology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Lewis X Antigen/genetics , Mice , Neural Stem Cells/cytology , Signal Transduction/genetics , Signal Transduction/physiology
4.
J Biol Chem ; 290(7): 4343-55, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25540202

ABSTRACT

Activation of γ-aminobutyric A receptors (GABA(A)Rs) in the subependymal zone (SEZ) induces hyperpolarization and osmotic swelling in precursors, thereby promoting surface expression of the epidermal growth factor receptor (EGFR) and cell cycle entry. However, the mechanisms underlying the GABAergic modulation of cell swelling are unclear. Here, we show that GABA(A)Rs colocalize with the water channel aquaporin (AQP) 4 in prominin-1 immunopositive (P(+)) precursors in the postnatal SEZ, which include neural stem cells. GABA(A)R signaling promotes AQP4 expression by decreasing serine phosphorylation associated with the water channel. The modulation of AQP4 expression by GABA(A)R signaling is key to its effect on cell swelling and EGFR expression. In addition, GABA(A)R function also affects the ability of neural precursors to swell in response to an osmotic challenge in vitro and in vivo. Thus, the regulation of AQP4 by GABA(A)Rs is involved in controlling activation of neural stem cells and water exchange dynamics in the SEZ.


Subject(s)
Aquaporin 4/physiology , Ependyma/metabolism , Gene Expression Regulation , Lateral Ventricles/metabolism , Receptors, GABA-A/metabolism , Water/metabolism , Animals , Blotting, Western , Cell Size , Cells, Cultured , Ependyma/cytology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fluorescent Antibody Technique , Immunoprecipitation , Lateral Ventricles/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osmotic Pressure , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, GABA-A/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
5.
Stem Cells ; 31(8): 1621-32, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23630160

ABSTRACT

Cell-fate decisions and differentiation of embryonic and adult neural stem cells (NSC) are tightly controlled by lineage-restricted and temporal factors that interact with cell-intrinsic programs and extracellular signals through multiple regulatory loops. Imprinted genes are important players in neurodevelopment and mental health although their molecular and cellular functions remain poorly understood. Here, we show that the paternally expressed transcriptional regulator Zac1 (zinc finger protein regulating apoptosis and cell cycle arrest) is transiently induced during astroglial and neuronal differentiation of embryonic and adult NSC lines. Thereby, Zac1 transactivates Socs3 (suppressor of cytokine signaling 3), a potent inhibitor of prodifferentiative Jak/Stat3 signaling, in a lineage-specific manner to prevent precocious astroglial differentiation. In vivo, Zac1 and Socs3 colocalize in the neocortical ventricular zone during incipient astrogliogenesis. Zac1 overexpression in primary NSCs delays astroglial differentiation whereas knockdown of Zac1 or Socs3 facilitates formation of astroglial cells. This negative feedback loop is unrelated to Zac1's cell cycle arrest function and specific to the Jak/Stat3 pathway. Hence, reinstating Jak/Stat3 signaling in the presence of increased Zac1 expression allows for timely astroglial differentiation. Overall, we suggest that the imprinted gene Zac1 curtails astroglial differentiation of NSCs in the developing and adult brain.


Subject(s)
Astrocytes/cytology , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/metabolism , Neural Stem Cells/cytology , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription Factors/biosynthesis , Transcription Factors/metabolism , Tumor Suppressor Proteins/biosynthesis , Animals , Apoptosis/physiology , Astrocytes/metabolism , Cell Cycle Proteins/genetics , Cell Differentiation/physiology , DNA Methylation , Gene Expression Regulation , Genes, Tumor Suppressor , Humans , Janus Kinases/metabolism , Mice , Plasmids/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Transcription Factors/genetics , Transcriptional Activation , Transfection , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
World J Stem Cells ; 7(2): 300-14, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25815116

ABSTRACT

Neural stem cells (NSCs) and imprinted genes play an important role in brain development. On historical grounds, these two determinants have been largely studied independently of each other. Recent evidence suggests, however, that NSCs can reset select genomic imprints to prevent precocious depletion of the stem cell reservoir. Moreover, imprinted genes like the transcriptional regulator Zac1 can fine tune neuronal vs astroglial differentiation of NSCs. Zac1 binds in a sequence-specific manner to pro-neuronal and imprinted genes to confer transcriptional regulation and furthermore coregulates members of the p53-family in NSCs. At the genome scale, Zac1 is a central hub of an imprinted gene network comprising genes with an important role for NSC quiescence, proliferation and differentiation. Overall, transcriptional, epigenomic, and genomic mechanisms seem to coordinate the functional relationships of NSCs and imprinted genes from development to maturation, and possibly aging.

7.
Mol Cell Biol ; 34(6): 1020-30, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24396065

ABSTRACT

Imprinted genes play a critical role in brain development and mental health, although the underlying molecular and cellular mechanisms remain incompletely understood. The family of basic helix-loop-helix (bHLH) proteins directs the proliferation, differentiation, and specification of distinct neuronal progenitor populations. Here, we identified the bHLH factor gene Tcf4 as a direct target gene of Zac1/Plagl1, a maternally imprinted transcriptional regulator, during early neurogenesis. Zac1 and Tcf4 expression levels concomitantly increased during neuronal progenitor differentiation; moreover, Zac1 interacts with two cis-regulatory elements in the Tcf4 gene locus, and these elements together confer synergistic activation of the Tcf4 gene. Tcf4 upregulation enhances the expression of the cyclin-dependent kinase inhibitor gene p57(Kip2), a paternally imprinted Tcf4 target gene, and increases the number of cells in G1 phase. Overall, we show that Zac1 controls cell cycle arrest function in neuronal progenitors through induction of p57(Kip2) via Tcf4 and provide evidence for cooperation between imprinted genes and a bHLH factor in early neurodevelopment.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/genetics , Embryonic Stem Cells/metabolism , Transcription Factors/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Brain/growth & development , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cyclin-Dependent Kinase Inhibitor p57/metabolism , G1 Phase/genetics , Gene Expression Regulation, Developmental/genetics , Genes, Tumor Suppressor , Mice , Neurogenesis/genetics , Protein Binding/genetics , Transcription Factor 4 , Transcription Factors/metabolism , Transcriptional Activation/genetics , Up-Regulation/genetics
8.
Epigenomics ; 6(5): 515-32, 2014.
Article in English | MEDLINE | ID: mdl-25431944

ABSTRACT

Imprinted genes and neural stem cells (NSC) play an important role in the developing and mature brain. A central theme of imprinted gene function in NSCs is cell survival and G1 arrest to control cell division, cell-cycle exit, migration and differentiation. Moreover, genomic imprinting can be epigenetically switched off at some genes to ensure stem cell quiescence and differentiation. At the genome scale, imprinted genes are organized in dynamic networks formed by interchromosomal interactions and transcriptional coregulation of imprinted and nonimprinted genes. Such multilayered networks may synchronize NSC activity with the demand from the niche resembling their roles in adjusting fetal size.


Subject(s)
Genomic Imprinting , Neural Stem Cells/metabolism , Animals , Apoptosis/genetics , Brain/metabolism , Brain/physiology , Cell Cycle/genetics , Cell Differentiation , Epigenesis, Genetic , Gene Dosage , Gene Expression Regulation , Gene Regulatory Networks , Humans , Neural Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL