Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Cell Mol Life Sci ; 80(2): 54, 2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36715759

ABSTRACT

Neural stem cells reside in the subgranular zone, a specialized neurogenic niche of the hippocampus. Throughout adulthood, these cells give rise to neurons in the dentate gyrus, playing an important role in learning and memory. Given that these core cognitive processes are disrupted in numerous disease states, understanding the underlying mechanisms of neural stem cell proliferation in the subgranular zone is of direct practical interest. Here, we report that mature neurons, neural stem cells and neural precursor cells each secrete the neurovascular protein epidermal growth factor-like protein 7 (EGFL7) to shape this hippocampal niche. We further demonstrate that EGFL7 knock-out in a Nestin-CreERT2-based mouse model produces a pronounced upregulation of neurogenesis within the subgranular zone. RNA sequencing identified that the increased expression of the cytokine VEGF-D correlates significantly with the ablation of EGFL7. We substantiate this finding with intraventricular infusion of VEGF-D upregulating neurogenesis in vivo and further show that VEGF-D knock-out produces a downregulation of neurogenesis. Finally, behavioral studies in EGFL7 knock-out mice demonstrate greater maintenance of spatial memory and improved memory consolidation in the hippocampus by modulation of pattern separation. Taken together, our findings demonstrate that both EGFL7 and VEGF-D affect neurogenesis in the adult hippocampus, with the ablation of EGFL7 upregulating neurogenesis, increasing spatial learning and memory, and correlating with increased VEGF-D expression.


Subject(s)
Neural Stem Cells , Mice , Animals , Neural Stem Cells/metabolism , Spatial Learning , Vascular Endothelial Growth Factor D/metabolism , Cell Proliferation/physiology , Hippocampus/metabolism , Neurogenesis/genetics , Mice, Knockout , Intercellular Signaling Peptides and Proteins/metabolism
3.
J Neurochem ; 159(3): 452-478, 2021 11.
Article in English | MEDLINE | ID: mdl-34478569

ABSTRACT

Autism spectrum disorder (ASD) comprises a group of multifactorial neurodevelopmental disorders primarily characterized by deficits in social interaction and repetitive behavior. Although the onset is typically in early childhood, ASD poses a lifelong challenge for both patients and caretakers. Adult neurogenesis (AN) is the process by which new functional neurons are created from neural stem cells existing in the post-natal brain. The entire event is based on a sequence of cellular processes, such as proliferation, specification of cell fate, maturation, and ultimately, synaptic integration into the existing neural circuits. Hence, AN is implicated in structural and functional brain plasticity throughout life. Accumulating evidence shows that impaired AN may underlie some of the abnormal behavioral phenotypes seen in ASD. In this review, we approach the interconnections between the molecular pathways related to AN and ASD. We also discuss existing therapeutic approaches targeting such pathways both in preclinical and clinical studies. A deeper understanding of how ASD and AN reciprocally affect one another could reveal important converging pathways leading to the emergence of psychiatric disorders.


Subject(s)
Autism Spectrum Disorder/pathology , Neurogenesis/physiology , Adult , Animals , Autism Spectrum Disorder/genetics , Child, Preschool , Humans , Neural Stem Cells , Neurodevelopmental Disorders , Neurogenesis/genetics
4.
EMBO Mol Med ; 10(9)2018 09.
Article in English | MEDLINE | ID: mdl-30065025

ABSTRACT

Glioblastoma (GBM) is a typically lethal type of brain tumor with a median survival of 15 months postdiagnosis. This negative prognosis prompted the exploration of alternative treatment options. In particular, the reliance of GBM on angiogenesis triggered the development of anti-VEGF (vascular endothelial growth factor) blocking antibodies such as bevacizumab. Although its application in human GBM only increased progression-free periods but did not improve overall survival, physicians and researchers still utilize this treatment option due to the lack of adequate alternatives. In an attempt to improve the efficacy of anti-VEGF treatment, we explored the role of the egfl7 gene in malignant glioma. We found that the encoded extracellular matrix protein epidermal growth factor-like protein 7 (EGFL7) was secreted by glioma blood vessels but not glioma cells themselves, while no major role could be assigned to the parasitic miRNAs miR-126/126*. EGFL7 expression promoted glioma growth in experimental glioma models in vivo and stimulated tumor vascularization. Mechanistically, this was mediated by an upregulation of integrin α5ß1 on the cellular surface of endothelial cells, which enhanced fibronectin-induced angiogenic sprouting. Glioma blood vessels that formed in vivo were more mature as determined by pericyte and smooth muscle cell coverage. Furthermore, these vessels were less leaky as measured by magnetic resonance imaging of extravasating contrast agent. EGFL7-inhibition using a specific blocking antibody reduced the vascularization of experimental gliomas and increased the life span of treated animals, in particular in combination with anti-VEGF and the chemotherapeutic agent temozolomide. Data allow for the conclusion that this combinatorial regimen may serve as a novel treatment option for GBM.


Subject(s)
Brain Neoplasms/pathology , Endothelial Growth Factors/metabolism , Glioblastoma/pathology , Integrin alpha5beta1/metabolism , Neovascularization, Pathologic/physiopathology , Animals , Antineoplastic Agents, Immunological/administration & dosage , Calcium-Binding Proteins , Cell Proliferation , Disease Models, Animal , EGF Family of Proteins , Endothelial Cells/metabolism , Endothelial Growth Factors/antagonists & inhibitors , Heterografts , Human Umbilical Vein Endothelial Cells , Humans , Mice , Neoplasm Transplantation , Survival Analysis , Treatment Outcome
5.
Nat Commun ; 9(1): 819, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29483510

ABSTRACT

Extracellular matrix (ECM) proteins secreted by blood-brain barrier (BBB) endothelial cells (ECs) are implicated in cell trafficking. We discovered that the expression of ECM epidermal growth factor-like protein 7 (EGFL7) is increased in the CNS vasculature of patients with multiple sclerosis (MS), and in mice with experimental autoimmune encephalomyelitis (EAE). Perivascular CD4 T lymphocytes colocalize with ECM-bound EGFL7 in MS lesions. Human and mouse activated T cells upregulate EGFL7 ligand αvß3 integrin and can adhere to EGFL7 through integrin αvß3. EGFL7-knockout (KO) mice show earlier onset of EAE and increased brain and spinal cord parenchymal infiltration of T lymphocytes. Importantly, EC-restricted EGFL7-KO is associated with a similar EAE worsening. Finally, treatment with recombinant EGFL7 improves EAE, reduces MCAM expression, and tightens the BBB in mouse. Our data demonstrate that EGFL7 can limit CNS immune infiltration and may represent a novel therapeutic avenue in MS.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Endothelial Growth Factors/genetics , Spinal Cord/drug effects , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/immunology , Brain/metabolism , Brain/pathology , CD146 Antigen/genetics , CD146 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Calcium-Binding Proteins , EGF Family of Proteins , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Growth Factors/deficiency , Endothelial Growth Factors/immunology , Endothelial Growth Factors/pharmacology , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/immunology , Female , Gene Expression Regulation , Humans , Integrin alpha5/genetics , Integrin alpha5/immunology , Integrin beta3/genetics , Integrin beta3/immunology , Lymphocyte Activation , Male , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Spinal Cord/immunology
6.
Nat Commun ; 8: 15922, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28656980

ABSTRACT

Adult neural stem cells reside in a specialized niche in the subventricular zone (SVZ). Throughout life they give rise to adult-born neurons in the olfactory bulb (OB), thus contributing to neural plasticity and pattern discrimination. Here, we show that the neurovascular protein EGFL7 is secreted by endothelial cells and neural stem cells (NSCs) of the SVZ to shape the vascular stem-cell niche. Loss of EGFL7 causes an accumulation of activated NSCs, which display enhanced activity and re-entry into the cell cycle. EGFL7 pushes activated NSCs towards quiescence and neuronal progeny towards differentiation. This is achieved by promoting Dll4-induced Notch signalling at the blood vessel-stem cell interface. Fewer inhibitory neurons form in the OB of EGFL7-knockout mice, which increases the absolute signal conducted from the mitral cell layer of the OB but decreases neuronal network synchronicity. Consequently, EGFL7-knockout mice display severe physiological defects in olfactory behaviour and perception.


Subject(s)
Adult Stem Cells/metabolism , Lateral Ventricles/metabolism , Neurogenesis , Olfactory Perception , Proteins/metabolism , Adult Stem Cells/cytology , Animals , Calcium-Binding Proteins , Cell Cycle , EGF Family of Proteins , Lateral Ventricles/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neuronal Plasticity , Proteins/genetics , Signal Transduction
7.
Acta Neuropathol ; 129(2): 279-95, 2015 02.
Article in English | MEDLINE | ID: mdl-25500713

ABSTRACT

The contribution of microglia to ischemic cortical stroke is of particular therapeutic interest because of the impact on the survival of brain tissue in the ischemic penumbra, a region that is potentially salvable upon a brain infarct. Whether or not tissue in the penumbra survives critically depends on blood flow and vessel perfusion. To study the role of microglia in cortical stroke and blood vessel stability, CX3CR1(+/GFP) mice were subjected to transient middle cerebral artery occlusion and then microglia were investigated using time-lapse two-photon microscopy in vivo. Soon after reperfusion, microglia became activated in the stroke penumbra and started to expand cellular protrusions towards adjacent blood vessels. All microglia in the penumbra were found associated with blood vessels within 24 h post reperfusion and partially fully engulfed them. In the same time frame blood vessels became permissive for blood serum components. Migration assays in vitro showed that blood serum proteins leaking into the tissue provided molecular cues leading to the recruitment of microglia to blood vessels and to their activation. Subsequently, these perivascular microglia started to eat up endothelial cells by phagocytosis, which caused an activation of the local endothelium and contributed to the disintegration of blood vessels with an eventual break down of the blood brain barrier. Loss-of-microglia-function studies using CX3CR1(GFP/GFP) mice displayed a decrease in stroke size and a reduction in the extravasation of contrast agent into the brain penumbra as measured by MRI. Potentially, medication directed at inhibiting microglia activation within the first day after stroke could stabilize blood vessels in the penumbra, increase blood flow, and serve as a valuable treatment for patients suffering from ischemic stroke.


Subject(s)
Brain Ischemia/physiopathology , Brain/blood supply , Brain/physiopathology , Microglia/physiology , Stroke/physiopathology , Animals , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Brain/pathology , Brain Ischemia/pathology , CX3C Chemokine Receptor 1 , Cell Line , Disease Models, Animal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Phagocytosis/physiology , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Stroke/pathology
8.
Blood ; 121(15): 3041-50, 2013 04 11.
Article in English | MEDLINE | ID: mdl-23386126

ABSTRACT

Angiogenesis, defined as blood vessel formation from a preexisting vasculature, is governed by multiple signal cascades including integrin receptors, in particular integrin αVß3. Here we identify the endothelial cell (EC)-secreted factor epidermal growth factor-like protein 7 (EGFL7) as a novel specific ligand of integrin αVß3, thus providing mechanistic insight into its proangiogenic actions in vitro and in vivo. Specifically, EGFL7 attaches to the extracellular matrix and by its interaction with integrin αVß3 increases the motility of EC, which allows EC to move on a sticky underground during vessel remodeling. We provide evidence that the deregulation of EGFL7 in zebrafish embryos leads to a severe integrin-dependent malformation of the caudal venous plexus, pointing toward the significance of EGFL7 in vessel development. In biopsy specimens of patients with neurologic diseases, vascular EGFL7 expression rose with increasing EC proliferation. Further, EGFL7 became upregulated in vessels of the stroke penumbra using a mouse model of reversible middle cerebral artery occlusion. Our data suggest that EGFL7 expression depends on the remodeling state of the existing vasculature rather than on the phenotype of neurologic disease analyzed. In sum, our work sheds a novel light on the molecular mechanism EGFL7 engages to govern physiological and pathological angiogenesis.


Subject(s)
Blood Vessels/metabolism , Endothelial Growth Factors/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Integrin alphaVbeta3/metabolism , Amino Acid Motifs/genetics , Animals , Calcium-Binding Proteins , Cell Adhesion/genetics , Cell Movement/genetics , EGF Family of Proteins , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/metabolism , Endothelial Growth Factors/genetics , Endothelial Growth Factors/pharmacology , Extracellular Matrix/metabolism , Gene Expression , HEK293 Cells , Humans , Immunohistochemistry , Immunoprecipitation , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Integrin alphaVbeta3/genetics , Mice , Mice, Nude , Phosphorylation/drug effects , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Zebrafish
9.
Cell Cycle ; 9(7): 1263-9, 2010 04 01.
Article in English | MEDLINE | ID: mdl-20372059

ABSTRACT

EGFL7 drives the formation of neurons from neural stem cells. In the embryonic and adult brain this process is essential for neurogenesis and homeostasis of the nervous system. The function of adult neurogenesis is not fully understood but maybe it supports life-long learning and brain repair after injuries such as stroke. The transition of neural stem cells into mature neurons is tightly regulated. One of the essential signaling pathways governing this process is the Notch pathway, which controls metazoan development. In a recent publication, we identified a novel non-canonical Notch ligand, EGFL7, and described its impact on neural stem cells. We explored the molecular mechanisms, which this molecule affects to regulate the self-renewal capacity of neural stem cells and to promote their differentiation into neurons. In this review, we discuss the implications of our findings for adult neurogenesis and illustrate the potential of EGFL7 to serve as an agent to increase neurogenesis and the self-renewal potential of the brain


Subject(s)
Endothelial Growth Factors/metabolism , Neural Stem Cells/metabolism , Animals , Calcium-Binding Proteins , Cell Differentiation/genetics , Cell Differentiation/physiology , EGF Family of Proteins , Endothelial Growth Factors/genetics , Humans , Mice , Nervous System/metabolism , Neural Stem Cells/cytology , Neurogenesis/genetics , Neurogenesis/physiology , Proteins/genetics , Proteins/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
10.
Nat Cell Biol ; 11(7): 873-80, 2009 07.
Article in English | MEDLINE | ID: mdl-19503073

ABSTRACT

Epidermal growth factor-like domain 7 (EGFL7) is a secreted factor implicated in cellular responses such as cell migration and blood vessel formation; however the molecular mechanisms underlying the effects of EGFL7 are largely unknown. Here we have identified transmembrane receptors of the Notch family as EGFL7-binding molecules. Secreted EGFL7 binds to a region in Notch involved in ligand-mediated receptor activation, thus acting as an antagonist of Notch signalling. Expression of EGFL7 in neural stem cells (NSCs) in vitro decreased Notch-specific signalling and consequently, reduced proliferation and self-renewal of NSCs. Such altered Notch signalling caused a shift in the differentiation pattern of cultured NSCs towards an excess of neurons and oligodendrocytes. We identified neurons as a source of EGFL7 in the brain, suggesting that brain-derived EGFL7 acts as an endogenous antagonist of Notch signalling that regulates proliferation and differentiation of subventricular zone-derived adult NSCs.


Subject(s)
Endothelial Growth Factors/physiology , Neurons/cytology , Neurons/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Calcium-Binding Proteins , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cell Proliferation , Cells, Cultured , EGF Family of Proteins , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Humans , Mice , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Two-Hybrid System Techniques
11.
Plant J ; 39(1): 98-112, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15200645

ABSTRACT

Heat stress transcription factors (Hsfs) are the major regulators of the plant heat stress (hs) response. Sequencing of the Arabidopsis genome revealed the existence of 21 open-reading frames (ORFs) encoding putative Hsfs assigned to classes A-C. Here we present results of a functional genomics approach to the Arabidopsis Hsf family focused on the analysis of their C-terminal domains (CTDs) harboring conserved modules for their function as transcription factors and their intracellular localization. Using reporter assays in tobacco protoplasts and yeast as well as glutathione-S-transferase (GST) pull-down assays, we demonstrate that short peptide motifs enriched with aromatic and large hydrophobic amino acid (aa) residues embedded in an acidic surrounding (AHA motifs) are essential for transcriptional activity of class A Hsfs. In contrast to this, class B and C Hsfs lack AHA motifs and have no activator function on their own. We also provide evidence for the function of a leucine (Leu)-rich region centered around a conserved QMGPhiL motif at the very C-terminus as a nuclear export signal (NES) of class A Hsfs. Sequence comparison indicates that the combination of a C-terminal AHA motif with the consensus sequence FWxxF/L,F/I/L as well as the adjacent NES represents a signature domain for plant class A Hsfs, which allowed to identify more than 60 new Hsfs from the expressed sequence tag (EST) database.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/genetics , DNA-Binding Proteins/chemistry , Heat-Shock Proteins/chemistry , Transcription Factors/chemistry , Amino Acid Motifs , Amino Acid Sequence , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Conserved Sequence , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Genes, Reporter , Heat Shock Transcription Factors , Heat-Shock Proteins/genetics , Molecular Sequence Data , Nuclear Localization Signals/metabolism , Physical Chromosome Mapping , Plant Proteins , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Nicotiana/genetics , Nicotiana/metabolism , Transcription Factors/metabolism , Yeasts/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...