Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4879, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849354

ABSTRACT

The mammalian neocortex comprises an enormous diversity regarding cell types, morphology, and connectivity. In this work, we discover a post-transcriptional mechanism of gene expression regulation, protein translation, as a determinant of cortical neuron identity. We find specific upregulation of protein synthesis in the progenitors of later-born neurons and show that translation rates and concomitantly protein half-lives are inherent features of cortical neuron subtypes. In a small molecule screening, we identify Ire1α as a regulator of Satb2 expression and neuronal polarity. In the developing brain, Ire1α regulates global translation rates, coordinates ribosome traffic, and the expression of eIF4A1. Furthermore, we demonstrate that the Satb2 mRNA translation requires eIF4A1 helicase activity towards its 5'-untranslated region. Altogether, we show that cortical neuron diversity is generated by mechanisms operating beyond gene transcription, with Ire1α-safeguarded proteostasis serving as an essential regulator of brain development.


Subject(s)
Matrix Attachment Region Binding Proteins , Neocortex , Neurons , Protein Biosynthesis , Protein Serine-Threonine Kinases , Animals , Neocortex/metabolism , Neocortex/cytology , Neocortex/embryology , Neurons/metabolism , Neurons/cytology , Mice , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Matrix Attachment Region Binding Proteins/metabolism , Matrix Attachment Region Binding Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Gene Expression Regulation, Developmental , Proteostasis , Neurogenesis/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , 5' Untranslated Regions/genetics , Ribosomes/metabolism , Ribosomes/genetics , Humans , Endoribonucleases/metabolism , Endoribonucleases/genetics , Cell Differentiation/genetics
2.
medRxiv ; 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38260255

ABSTRACT

SPOUT1/CENP-32 encodes a putative SPOUT RNA methyltransferase previously identified as a mitotic chromosome associated protein. SPOUT1/CENP-32 depletion leads to centrosome detachment from the spindle poles and chromosome misalignment. Aided by gene matching platforms, we identified 24 individuals with neurodevelopmental delays from 18 families with bi-allelic variants in SPOUT1/CENP-32 detected by exome/genome sequencing. Zebrafish spout1/cenp-32 mutants showed reduction in larval head size with concomitant apoptosis likely associated with altered cell cycle progression. In vivo complementation assays in zebrafish indicated that SPOUT1/CENP-32 missense variants identified in humans are pathogenic. Crystal structure analysis of SPOUT1/CENP-32 revealed that most disease-associated missense variants mapped to the catalytic domain. Additionally, SPOUT1/CENP-32 recurrent missense variants had reduced methyltransferase activity in vitro and compromised centrosome tethering to the spindle poles in human cells. Thus, SPOUT1/CENP-32 pathogenic variants cause an autosomal recessive neurodevelopmental disorder: SpADMiSS ( SPOUT1 Associated Development delay Microcephaly Seizures Short stature) underpinned by mitotic spindle organization defects and consequent chromosome segregation errors.

3.
Cereb Cortex ; 33(5): 1752-1767, 2023 02 20.
Article in English | MEDLINE | ID: mdl-35462405

ABSTRACT

Abnormal development of corpus callosum is relatively common and causes a broad spectrum of cognitive impairments in humans. We use acallosal Neurod2/6-deficient mice to study callosal axon guidance within the ipsilateral cerebral cortex. Initial callosal tracts form but fail to traverse the ipsilateral cingulum and are not attracted towards the midline in the absence of Neurod2/6. We show that the restoration of Ephrin-A4 (EfnA4) expression in the embryonic neocortex of Neurod2/6-deficient embryos is sufficient to partially rescue targeted callosal axon growth towards the midline. EfnA4 cannot directly mediate reverse signaling within outgrowing axons, but it forms co-receptor complexes with TrkB (Ntrk2). The ability of EfnA4 to rescue the guided growth of a subset of callosal axons in Neurod2/6-deficient mice is abolished by the co-expression of dominant negative TrkBK571N (kinase-dead) or TrkBY515F (SHC-binding deficient) variants, but not by TrkBY816F (PLCγ1-binding deficient). Additionally, EphA4 is repulsive to EfnA4-positive medially projecting axons in organotypic brain slice culture. Collectively, we suggest that EfnA4-mediated reverse signaling acts via TrkB-SHC and is required for ipsilateral callosal axon growth accuracy towards the midline downstream of Neurod family factors.


Subject(s)
Neocortex , Neuropeptides , Mice , Animals , Humans , Corpus Callosum/metabolism , Axons/physiology , Neocortex/metabolism , Nerve Fibers , Phosphotransferases/metabolism , Neuropeptides/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism
4.
Sci Adv ; 7(27)2021 Jul.
Article in English | MEDLINE | ID: mdl-34215578

ABSTRACT

The neocortex is stereotypically organized into layers of excitatory neurons arranged in a precise parallel orientation. Here we show that dynamic adhesion both preceding and following radial migration is essential for this organization. Neuronal adhesion is regulated by the Mowat-Wilson syndrome-associated transcription factor Zeb2 (Sip1/Zfhx1b) through direct repression of independent adhesion pathways controlled by Neuropilin-1 (Nrp1) and Cadherin-6 (Cdh6). We reveal that to initiate radial migration, neurons must first suppress adhesion to the extracellular matrix. Zeb2 regulates the multipolar stage by transcriptional repression of Nrp1 and thereby downstream inhibition of integrin signaling. Upon completion of migration, neurons undergo an orientation process that is independent of migration. The parallel organization of neurons within the neocortex is controlled by Cdh6 through atypical regulation of integrin signaling via its RGD motif. Our data shed light on the mechanisms that regulate initiation of radial migration and the postmigratory orientation of neurons during neocortical development.

5.
Sci Rep ; 11(1): 1144, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441772

ABSTRACT

Mortality of patients with end-stage renal disease tremendously exceeds that of the general population due to excess cardiovascular morbidity. Large middle-sized molecules (LMM) including pro-inflammatory cytokines are major drivers of uremic cardiovascular toxicity and cannot be removed sufficiently by conventional high-flux (HFL) hemodialysis. We tested the ability of plasma from 19 hemodialysis patients participating in a trial comparing HFL with high cut-off (HCO) membranes facilitating removal of LMM to induce calcification in mesenchymal stromal cells (MSC) functioning as vascular progenitors. HCO dialysis favorably changed plasma composition resulting in reduced pro-calcific activity. LMM were removed more effectively by HCO dialysis including FGF23, a typical LMM we found to promote osteoblastic differentiation of MSC. Protein-bound uremic retention solutes with known cardiovascular toxicity but not LMM inhibited proliferation of MSC without direct toxicity in screening experiments. We could not attribute the effect of HCO dialysis on MSC calcification to distinct mediators. However, we found evidence of sustained reduced inflammation that might parallel other anti-calcifying mechanisms such as altered generation of extracellular vesicles. Our findings imply protection of MSC from dysfunctional differentiation by novel dialysis techniques targeted at removal of LMM. HCO dialysis might preserve their physiologic role in vascular regeneration and improve outcomes in dialysis patients.


Subject(s)
Mesenchymal Stem Cells/pathology , Osteoblasts/pathology , Renal Dialysis/adverse effects , Vascular Calcification/etiology , Aged , Aged, 80 and over , Cell Differentiation , Female , Fibroblast Growth Factor-23 , Humans , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/pathology , Kidney Failure, Chronic/therapy , Male , Mesenchymal Stem Cells/cytology , Middle Aged , Osteoblasts/cytology , Renal Dialysis/instrumentation , Renal Dialysis/methods , Vascular Calcification/blood , Vascular Calcification/pathology
6.
Mol Cell ; 81(2): 304-322.e16, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33357414

ABSTRACT

Protein synthesis must be finely tuned in the developing nervous system as the final essential step of gene expression. This study investigates the architecture of ribosomes from the neocortex during neurogenesis, revealing Ebp1 as a high-occupancy 60S peptide tunnel exit (TE) factor during protein synthesis at near-atomic resolution by cryoelectron microscopy (cryo-EM). Ribosome profiling demonstrated Ebp1-60S binding is highest during start codon initiation and N-terminal peptide elongation, regulating ribosome occupancy of these codons. Membrane-targeting domains emerging from the 60S tunnel, which recruit SRP/Sec61 to the shared binding site, displace Ebp1. Ebp1 is particularly abundant in the early-born neural stem cell (NSC) lineage and regulates neuronal morphology. Ebp1 especially impacts the synthesis of membrane-targeted cell adhesion molecules (CAMs), measured by pulsed stable isotope labeling by amino acids in cell culture (pSILAC)/bioorthogonal noncanonical amino acid tagging (BONCAT) mass spectrometry (MS). Therefore, Ebp1 is a central component of protein synthesis, and the ribosome TE is a focal point of gene expression control in the molecular specification of neuronal morphology during development.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Neocortex/metabolism , Neurons/metabolism , Protein Biosynthesis , Proteostasis/genetics , RNA-Binding Proteins/genetics , Ribosome Subunits, Large, Eukaryotic/genetics , Animals , Animals, Newborn , Binding Sites , Cell Adhesion Molecules, Neuronal/chemistry , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Line, Tumor , Cryoelectron Microscopy , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Female , Male , Mice , Neocortex/cytology , Neocortex/growth & development , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurons/cytology , Primary Cell Culture , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosome Subunits, Large, Eukaryotic/ultrastructure , Signal Recognition Particle/chemistry , Signal Recognition Particle/genetics , Signal Recognition Particle/metabolism
7.
Mol Psychiatry ; 26(6): 1980-1995, 2021 06.
Article in English | MEDLINE | ID: mdl-32249816

ABSTRACT

Kaufman oculocerebrofacial syndrome (KOS) is a severe autosomal recessive disorder characterized by intellectual disability, developmental delays, microcephaly, and characteristic dysmorphisms. Biallelic mutations of UBE3B, encoding for a ubiquitin ligase E3B are causative for KOS. In this report, we characterize neuronal functions of its murine ortholog Ube3b and show that Ube3b regulates dendritic branching in a cell-autonomous manner. Moreover, Ube3b knockout (KO) neurons exhibit increased density and aberrant morphology of dendritic spines, altered synaptic physiology, and changes in hippocampal circuit activity. Dorsal forebrain-specific Ube3b KO animals show impaired spatial learning, altered social interactions, and repetitive behaviors. We further demonstrate that Ube3b ubiquitinates the catalytic γ-subunit of calcineurin, Ppp3cc, the overexpression of which phenocopies Ube3b loss with regard to dendritic spine density. This work provides insights into the molecular pathologies underlying intellectual disability-like phenotypes in a genetically engineered mouse model.


Subject(s)
Intellectual Disability , Microcephaly , Animals , Calcineurin , Dendritic Spines , Eye Abnormalities , Facies , Intellectual Disability/genetics , Limb Deformities, Congenital , Mice , Mice, Knockout , Microcephaly/genetics , Mutation/genetics , Synapses , Ubiquitin-Protein Ligases/genetics
8.
Sci Rep ; 10(1): 3740, 2020 Feb 25.
Article in English | MEDLINE | ID: mdl-32099063

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

9.
Sci Rep ; 9(1): 20071, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882658

ABSTRACT

Vascular regeneration depends on intact function of progenitors of vascular smooth muscle cells such as pericytes and their circulating counterparts, mesenchymal stromal cells (MSC). Deregulated MSC differentiation and maladaptive cell fate programs associated with age and metabolic diseases may exacerbate arteriosclerosis due to excessive transformation to osteoblast-like calcifying cells. Targeting mTOR, a central controller of differentiation and cell fates, could offer novel therapeutic perspectives. In a cell culture model for osteoblastic differentiation of pluripotent human MSC we found distinct roles for mTORC1 and mTORC2 in the regulation of differentiation towards calcifying osteoblasts via cell fate programs in a temporally-controlled sequence. Activation of mTORC1 with induction of cellular senescence and apoptosis were hallmarks of transition to a calcifying phenotype. Inhibition of mTORC1 with Rapamycin elicited reciprocal activation of mTORC2, enhanced autophagy and recruited anti-apoptotic signals, conferring protection from calcification. Pharmacologic and genetic negative interference with mTORC2 function or autophagy both abolished regenerative programs but induced cellular senescence, apoptosis, and calcification. Overexpression of the mTORC2 constituent rictor revealed that enhanced mTORC2 signaling without altered mTORC1 function was sufficient to inhibit calcification. Studies in mice reproduced the in vitro effects of mTOR modulation with Rapamycin on cell fates in vascular cells in vivo. Amplification of mTORC2 signaling promotes protective cell fates including autophagy to counteract osteoblast differentiation and calcification of MSC, representing a novel mTORC2 function. Regenerative approaches aimed at modulating mTOR network activation patterns hold promise for delaying age-related vascular diseases and treatment of accelerated arteriosclerosis in chronic metabolic conditions.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Mechanistic Target of Rapamycin Complex 1/physiology , Mechanistic Target of Rapamycin Complex 2/physiology , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Adolescent , Adult , Animals , Child , Child, Preschool , Female , Humans , Infant , Male , Mice , Mice, Inbred C57BL , Young Adult
10.
Neuron ; 100(5): 1097-1115.e15, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30392800

ABSTRACT

The establishment of axon-dendrite polarity is fundamental for radial migration of neurons during cortex development of mammals. We demonstrate that the E3 ubiquitin ligases WW-Containing Proteins 1 and 2 (Wwp1 and Wwp2) are indispensable for proper polarization of developing neurons. We show that knockout of Wwp1 and Wwp2 results in defects in axon-dendrite polarity in pyramidal neurons, and their aberrant laminar cortical distribution. Knockout of miR-140, encoded in Wwp2 intron, engenders phenotypic changes analogous to those upon Wwp1 and Wwp2 deletion. Intriguingly, transcription of the Wwp1 and Wwp2/miR-140 loci in neurons is induced by the transcription factor Sox9. Finally, we provide evidence that miR-140 supervises the establishment of axon-dendrite polarity through repression of Fyn kinase mRNA. Our data delineate a novel regulatory pathway that involves Sox9-[Wwp1/Wwp2/miR-140]-Fyn required for axon specification, acquisition of pyramidal morphology, and proper laminar distribution of cortical neurons.


Subject(s)
Cell Polarity , Cerebral Cortex/growth & development , MicroRNAs/physiology , Neurons/physiology , SOX9 Transcription Factor/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Axons/physiology , Cerebral Cortex/cytology , Dendrites/physiology , Female , Gene Expression Regulation, Developmental , Male , Mice, Knockout , MicroRNAs/genetics , Neurons/cytology , SOX9 Transcription Factor/genetics , Ubiquitin-Protein Ligases/genetics
11.
Sci Rep ; 8(1): 12087, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30108259

ABSTRACT

Severe vascular calcification develops almost invariably in chronic kidney patients posing a substantial risk to quality of life and survival. This unmet medical need demands identification of novel therapeutic modalities. We aimed to pinpoint components of the uremic microenvironment triggering differentiation of vascular progenitors to calcifying osteoblast-like cells. In an unbiased approach, assessing the individual potency of 63 uremic retention solutes to enhance calcific phenotype conversion of vascular progenitor cells, the pro-inflammatory cytokines IL-1ß and TNF-α were identified as the strongest inducers followed by FGF-2, and PTH. Pharmacologic targeting of these molecules alone or in combination additively antagonized pro-calcifying properties of sera from uremic patients. Our findings stress the importance of pro-inflammatory cytokines above other characteristic components of the uremic microenvironment as key mediators of calcifying osteoblastic differentiation in vascular progenitors. Belonging to the group of "middle-sized molecules", they are neither effectively removed by conventional dialysis nor influenced by established supportive therapies. Specific pharmacologic interventions or novel extracorporeal approaches may help preserve regenerative capacity and control vascular calcification due to uremic environment.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cytokines/antagonists & inhibitors , Kidney Failure, Chronic/therapy , Uremia/therapy , Vascular Calcification/prevention & control , Adolescent , Anti-Inflammatory Agents/therapeutic use , Cell Differentiation/drug effects , Child , Child, Preschool , Cytokines/immunology , Female , Healthy Volunteers , Humans , Infant , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/complications , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Monocytes , Osteoblasts/physiology , Primary Cell Culture , Renal Dialysis , Uremia/blood , Uremia/immunology , Vascular Calcification/blood , Vascular Calcification/immunology , Young Adult
12.
PLoS One ; 11(4): e0153101, 2016.
Article in English | MEDLINE | ID: mdl-27054717

ABSTRACT

INTRODUCTION: Obliterative vasculopathy and fibrosis are hallmarks of systemic sclerosis (SSc), a severe systemic autoimmune disease. Bone marrow-derived mesenchymal stromal cells (MSCs) from SSc patients may harbor disease-specific abnormalities. We hypothesized disturbed vascular smooth muscle cell (VSMC) differentiation with increased propensity towards myofibroblast differentiation in response to SSc-microenvironment defining growth factors and determined responsible mechanisms. METHODS: We studied responses of multipotent MSCs from SSc-patients (SSc-MSCs) and healthy controls (H-MSCs) to long-term exposure to CTGF, b-FGF, PDGF-BB or TGF-ß1. Differentiation towards VSMC and myofibroblast lineages was analyzed on phenotypic, biochemical, and functional levels. Intracellular signaling studies included analysis of TGF-ß receptor regulation, SMAD, AKT, ERK1/2 and autocrine loops. RESULTS: VSMC differentiation towards both, contractile and synthetic VSMC phenotypes in response to CTGF and b-FGF was disturbed in SSc-MSCs. H-MSCs and SSc-MSCs responded equally to PDGF-BB with prototypic fibroblastic differentiation. TGF-ß1 initiated myofibroblast differentiation in both cell types, yet with striking phenotypic and functional differences: In relation to H-MSC-derived myofibroblasts induced by TGF-ß1, those obtained from SSc-MSCs expressed more contractile proteins, migrated towards TGF-ß1, had low proliferative capacity, and secreted higher amounts of collagen paralleled by reduced MMP expression. Higher levels of TGF-ß receptor 1 and enhanced canonical and noncanonical TGF-ß signaling in SSc-MSCs accompanied aberrant differentiation response of SSc-MSCs in comparison to H-MSCs. CONCLUSIONS: Deregulated VSMC differentiation with a shift towards myofibroblast differentiation expands the concept of disturbed endogenous regenerative capacity of MSCs from SSc patients. Disease related intrinsic hyperresponsiveness to TGF-ß1 with increased collagen production may represent one responsible mechanism. Better understanding of repair barriers and harnessing beneficial differentiation processes in MSCs could widen options of autologous MSC application in SSc patients.


Subject(s)
Mesenchymal Stem Cells/cytology , Muscle, Smooth, Vascular/cytology , Myofibroblasts/cytology , Scleroderma, Systemic/pathology , Adult , Aged , Becaplermin , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Proliferation , Cells, Cultured , Connective Tissue Growth Factor/pharmacology , Female , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation , Humans , Male , Mesenchymal Stem Cells/drug effects , Middle Aged , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Proto-Oncogene Proteins c-sis/pharmacology , Scleroderma, Systemic/genetics , Scleroderma, Systemic/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology
14.
J Cell Sci ; 122(Pt 16): 2946-56, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19638409

ABSTRACT

In this study, we characterize the molecular and functional features of a novel protein called SPOC1. SPOC1 RNA expression was previously reported to be highest in highly proliferating tissues and increased in a subset of ovarian carcinoma patients, which statistically correlated with poor prognosis and residual disease. These observations implied that SPOC1 might play a role in cellular proliferation and oncogenesis. Here we show that the endogenous SPOC1 protein is labile, primarily chromatin associated and its expression as well as localization are regulated throughout the cell cycle. SPOC1 is dynamically regulated during mitosis with increased expression levels and biphasic localization to mitotic chromosomes indicating a functional role of SPOC1 in mitotic processes. Consistent with this postulate, SPOC1 siRNA knockdown experiments resulted in defects in mitotic chromosome condensation, alignment and aberrant sister chromatid segregation. Finally, we have been able to show, using micrococcal nuclease (MNase) chromatin-digestion assays that SPOC1 expression levels proportionally influence the degree of chromatin compaction. Collectively, our findings show that SPOC1 modulates chromatin structure and that tight regulation of its expression levels and subcellular localization during mitosis are crucial for proper chromosome condensation and cell division.


Subject(s)
Chromatin/metabolism , Chromosomes, Human/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Mitosis , Transcription Factors/chemistry , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Eukaryotic Cells , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Metaphase , Prophase , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protein Transport , RNA, Small Interfering/metabolism , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...