Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Nature ; 628(8006): 204-211, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38418880

ABSTRACT

The eye, an anatomical extension of the central nervous system (CNS), exhibits many molecular and cellular parallels to the brain. Emerging research demonstrates that changes in the brain are often reflected in the eye, particularly in the retina1. Still, the possibility of an immunological nexus between the posterior eye and the rest of the CNS tissues remains unexplored. Here, studying immune responses to herpes simplex virus in the brain, we observed that intravitreal immunization protects mice against intracranial viral challenge. This protection extended to bacteria and even tumours, allowing therapeutic immune responses against glioblastoma through intravitreal immunization. We further show that the anterior and posterior compartments of the eye have distinct lymphatic drainage systems, with the latter draining to the deep cervical lymph nodes through lymphatic vasculature in the optic nerve sheath. This posterior lymphatic drainage, like that of meningeal lymphatics, could be modulated by the lymphatic stimulator VEGFC. Conversely, we show that inhibition of lymphatic signalling on the optic nerve could overcome a major limitation in gene therapy by diminishing the immune response to adeno-associated virus and ensuring continued efficacy after multiple doses. These results reveal a shared lymphatic circuit able to mount a unified immune response between the posterior eye and the brain, highlighting an understudied immunological feature of the eye and opening up the potential for new therapeutic strategies in ocular and CNS diseases.


Subject(s)
Brain , Eye , Lymphatic System , Animals , Female , Humans , Male , Mice , Rabbits , Bacteria/immunology , Brain/anatomy & histology , Brain/immunology , Dependovirus/immunology , Eye/anatomy & histology , Eye/immunology , Glioblastoma/immunology , Herpesvirus 2, Human/immunology , Intravitreal Injections , Lymphatic System/anatomy & histology , Lymphatic System/immunology , Lymphatic Vessels/anatomy & histology , Lymphatic Vessels/immunology , Macaca mulatta , Meninges/immunology , Optic Nerve/immunology , Swine , Zebrafish , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/pharmacology
2.
Nature ; 577(7792): 689-694, 2020 01.
Article in English | MEDLINE | ID: mdl-31942068

ABSTRACT

Immune surveillance against pathogens and tumours in the central nervous system is thought to be limited owing to the lack of lymphatic drainage. However, the characterization of the meningeal lymphatic network has shed light on previously unappreciated ways that an immune response can be elicited to antigens that are expressed in the brain1-3. Despite progress in our understanding of the development and structure of the meningeal lymphatic system, the contribution of this network in evoking a protective antigen-specific immune response in the brain remains unclear. Here, using a mouse model of glioblastoma, we show that the meningeal lymphatic vasculature can be manipulated to mount better immune responses against brain tumours. The immunity that is mediated by CD8 T cells to the glioblastoma antigen is very limited when the tumour is confined to the central nervous system, resulting in uncontrolled tumour growth. However, ectopic expression of vascular endothelial growth factor C (VEGF-C) promotes enhanced priming of CD8 T cells in the draining deep cervical lymph nodes, migration of CD8 T cells into the tumour, rapid clearance of the glioblastoma and a long-lasting antitumour memory response. Furthermore, transfection of an mRNA construct that expresses VEGF-C works synergistically with checkpoint blockade therapy to eradicate existing glioblastoma. These results reveal the capacity of VEGF-C to promote immune surveillance of tumours, and suggest a new therapeutic approach to treat brain tumours.


Subject(s)
Brain Neoplasms/immunology , Glioblastoma/immunology , Immunologic Surveillance/immunology , Lymph Nodes/immunology , Lymphatic Vessels/immunology , Vascular Endothelial Growth Factor C/metabolism , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/immunology , Cell Line, Tumor , Cell Movement , Central Nervous System/immunology , Central Nervous System/pathology , Cross-Priming , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , HEK293 Cells , Humans , Immunologic Memory/immunology , Lymphangiogenesis , Male , Melanoma/drug therapy , Melanoma/immunology , Meninges/immunology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Vascular Endothelial Growth Factor C/administration & dosage , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/therapeutic use
4.
Cell Mol Life Sci ; 80(7): 179, 2023 Jun 14.
Article in English | MEDLINE | ID: mdl-37314567

ABSTRACT

Glioblastoma (GBM) is the most common and fatal primary tumor of the central nervous system (CNS) and current treatments have limited success. Chemokine signaling regulates both malignant cells and stromal cells of the tumor microenvironment (TME), constituting a potential therapeutic target against brain cancers. Here, we investigated the C-C chemokine receptor type 7 (CCR7) and the chemokine (C-C-motif) ligand 21 (CCL21) for their expression and function in human GBM and then assessed their therapeutic potential in preclinical mouse GBM models. In GBM patients, CCR7 expression positively associated with a poor survival. CCL21-CCR7 signaling was shown to regulate tumor cell migration and proliferation while also controlling tumor associated microglia/macrophage recruitment and VEGF-A production, thereby controlling vascular dysmorphia. Inhibition of CCL21-CCR7 signaling led to an increased sensitivity to temozolomide-induced tumor cell death. Collectively, our data indicate that drug targeting of CCL21-CCR7 signaling in tumor and TME cells is a therapeutic option against GBM.


Subject(s)
Glioblastoma , Microglia , Animals , Mice , Humans , Glioblastoma/drug therapy , Receptors, CCR7/genetics , Macrophages , Central Nervous System , Tumor Microenvironment , Chemokine CCL21
6.
Proc Natl Acad Sci U S A ; 115(35): E8246-E8255, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30108144

ABSTRACT

Oligodendrocyte precursor cells (OPCs) constitute the main proliferative cells in the adult brain, and deregulation of OPC proliferation-differentiation balance results in either glioma formation or defective adaptive (re)myelination. OPC differentiation requires significant genetic reprogramming, implicating chromatin remodeling. Mounting evidence indicates that chromatin remodelers play important roles during normal development and their mutations are associated with neurodevelopmental defects, with CHD7 haploinsuficiency being the cause of CHARGE syndrome and CHD8 being one of the strongest autism spectrum disorder (ASD) high-risk-associated genes. Herein, we report on uncharacterized functions of the chromatin remodelers Chd7 and Chd8 in OPCs. Their OPC-chromatin binding profile, combined with transcriptome and chromatin accessibility analyses of Chd7-deleted OPCs, demonstrates that Chd7 protects nonproliferative OPCs from apoptosis by chromatin closing and transcriptional repression of p53 Furthermore, Chd7 controls OPC differentiation through chromatin opening and transcriptional activation of key regulators, including Sox10, Nkx2.2, and Gpr17 However, Chd7 is dispensable for oligodendrocyte stage progression, consistent with Chd8 compensatory function, as suggested by their common chromatin-binding profiles and genetic interaction. Finally, CHD7 and CHD8 bind in OPCs to a majority of ASD risk-associated genes, suggesting an implication of oligodendrocyte lineage cells in ASD neurological defects. Our results thus offer new avenues to understand and modulate the CHD7 and CHD8 functions in normal development and disease.


Subject(s)
Chromatin Assembly and Disassembly , DNA-Binding Proteins/metabolism , Oligodendroglia/metabolism , Stem Cells/metabolism , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , CHARGE Syndrome/genetics , CHARGE Syndrome/metabolism , CHARGE Syndrome/pathology , Cell Survival , DNA-Binding Proteins/genetics , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Mice , Mice, Knockout , Nuclear Proteins , Oligodendroglia/pathology , Stem Cells/pathology , Transcription Factors
7.
Dev Biol ; 442(1): 138-154, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30016639

ABSTRACT

Macrophages are well characterized as immune cells. However, in recent years, a multitude of non-immune functions have emerged many of which play essential roles in a variety of developmental processes (Wynn et al., 2013; DeFalco et al., 2014). In adult animals, macrophages are derived from circulating monocytes originating in the bone marrow, but much of the tissue-resident population arise from erythro-myeloid progenitors (EMPs) in the extra-embryonic yolk sac, appearing around the same time as primitive erythroblasts (Schulz et al., 2012; Kierdorf et al., 2013; McGrath et al., 2015; Gomez Perdiguero et al., 2015; Mass et al., 2016). Of particular interest to our group, macrophages have been shown to act as pro-angiogenic regulators during development (Wynn et al., 2013; DeFalco et al., 2014; Hsu et al., 2015), but there is still much to learn about these early cells. The goal of the present study was to isolate and expand progenitors of yolk-sac-derived Embryonic Macrophages (EMs) in vitro to generate a new platform for mechanistic studies of EM differentiation. To accomplish this goal, we isolated pure (>98%) EGFP+ populations by flow cytometry from embryonic day 9.5 (E9.5) Csf1r-EGFP+/tg mice, then evaluated the angiogenic potential of EMs relative to Bone Marrow-Derived Macrophages (BMDMs). We found that EMs expressed more pro-angiogenic and less pro-inflammatory macrophage markers than BMDMs. EMs also promoted more endothelial cell (EC) cord formation in vitro, as compared to BMDMs in a manner that required direct cell-to-cell contact. Importantly, EMs preferentially matured into microglia when co-cultured with mouse Neural Stem/Progenitor Cells (NSPCs). In conclusion, we have established a protocol to isolate and propagate EMs in vitro, have further defined specialized properties of yolk-sac-derived macrophages, and have identified EM-EC and EM-NSPC interactions as key inducers of EC tube formation and microglial cell maturation, respectively.


Subject(s)
Erythroid Precursor Cells/physiology , Macrophages/physiology , Myeloid Progenitor Cells/physiology , Animals , Cell Culture Techniques/methods , Cell Differentiation/physiology , Coculture Techniques/methods , Flow Cytometry/methods , Hematopoietic Stem Cells/physiology , Macrophages/cytology , Mice/embryology , Phenotype , Yolk Sac/cytology
8.
Cereb Cortex ; 28(7): 2479-2481, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29746611

ABSTRACT

In this issue of Cerebral Cortex, Cipriani et al. are following up on the recent report of Sorrels et al. to add novel immunohistological observations indicating that, unlike rodents, adult and aging humans do not acquire new neurons in the hippocampus. The common finding emerging from these 2 different, but almost simultaneous studies is highly significant because the dentate gyrus of the hippocampus was, until recently, considered as the only structure in the human brain that may continue neurogenesis throughout the full life span.


Subject(s)
Alzheimer Disease , Adult , Dentate Gyrus , Fetus , Hippocampus , Humans , Neurogenesis , Neurons
9.
Genes Dev ; 25(8): 831-44, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21498572

ABSTRACT

Neural stem cells (NSCs) are slowly dividing astrocytes that are intimately associated with capillary endothelial cells in the subventricular zone (SVZ) of the brain. Functionally, members of the vascular endothelial growth factor (VEGF) family can stimulate neurogenesis as well as angiogenesis, but it has been unclear whether they act directly via VEGF receptors (VEGFRs) expressed by neural cells, or indirectly via the release of growth factors from angiogenic capillaries. Here, we show that VEGFR-3, a receptor required for lymphangiogenesis, is expressed by NSCs and is directly required for neurogenesis. Vegfr3:YFP reporter mice show VEGFR-3 expression in multipotent NSCs, which are capable of self-renewal and are activated by the VEGFR-3 ligand VEGF-C in vitro. Overexpression of VEGF-C stimulates VEGFR-3-expressing NSCs and neurogenesis in the SVZ without affecting angiogenesis. Conversely, conditional deletion of Vegfr3 in neural cells, inducible deletion in subventricular astrocytes, and blocking of VEGFR-3 signaling with antibodies reduce SVZ neurogenesis. Therefore, VEGF-C/VEGFR-3 signaling acts directly on NSCs and regulates adult neurogenesis, opening potential approaches for treatment of neurodegenerative diseases.


Subject(s)
Neurogenesis/physiology , Vascular Endothelial Growth Factor Receptor-3/metabolism , Animals , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Lymphangiogenesis/genetics , Lymphangiogenesis/physiology , Mice , Mice, Mutant Strains , Microscopy, Electron, Transmission , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-3/genetics
10.
Circulation ; 135(23): 2288-2298, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28356442

ABSTRACT

BACKGROUND: Bone morphogenetic protein (BMP) signaling has multiple roles in the development and function of the blood vessels. In humans, mutations in BMP receptor type 2 (BMPR2), a key component of BMP signaling, have been identified in the majority of patients with familial pulmonary arterial hypertension (PAH). However, only a small subset of individuals with BMPR2 mutation develops PAH, suggesting that additional modifiers of BMPR2 function play an important role in the onset and progression of PAH. METHODS: We used a combination of studies in zebrafish embryos and genetically engineered mice lacking endothelial expression of Vegfr3 to determine the interaction between vascular endothelial growth factor receptor 3 (VEGFR3) and BMPR2. Additional in vitro studies were performed by using human endothelial cells, including primary lung endothelial cells from subjects with PAH. RESULTS: Attenuation of Vegfr3 in zebrafish embryos abrogated Bmp2b-induced ectopic angiogenesis. Endothelial cells with disrupted VEGFR3 expression failed to respond to exogenous BMP stimulation. Mechanistically, VEGFR3 is physically associated with BMPR2 and facilitates ligand-induced endocytosis of BMPR2 to promote phosphorylation of SMADs and transcription of ID genes. Conditional, endothelial-specific deletion of Vegfr3 in mice resulted in impaired BMP signaling responses, and significantly worsened hypoxia-induced pulmonary hypertension. Consistent with these data, we found significant decrease in VEGFR3 expression in pulmonary arterial endothelial cells from human PAH subjects, and reconstitution of VEGFR3 expression in PAH pulmonary arterial endothelial cells restored BMP signaling responses. CONCLUSIONS: Our findings identify VEGFR3 as a key regulator of endothelial BMPR2 signaling and a potential determinant of PAH penetrance in humans.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/biosynthesis , Endothelium, Vascular/metabolism , Hypertension, Pulmonary/metabolism , Vascular Endothelial Growth Factor Receptor-3/biosynthesis , Animals , Bone Morphogenetic Protein Receptors, Type II/genetics , Cells, Cultured , Endothelium, Vascular/pathology , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Vascular Endothelial Growth Factor Receptor-3/genetics , Zebrafish
11.
Mult Scler ; 24(11): 1421-1432, 2018 10.
Article in English | MEDLINE | ID: mdl-28752787

ABSTRACT

BACKGROUND: In multiple sclerosis, development of screening tools for remyelination-promoting molecules is timely. OBJECTIVE: A Xenopus transgenic line allowing conditional ablation of myelinating oligodendrocytes has been adapted for in vivo screening of remyelination-favoring molecules. METHODS: In this transgenic, the green fluorescent protein reporter is fused to E. coli nitroreductase and expressed specifically in myelinating oligodendrocytes. Nitroreductase converts the innocuous pro-drug metronidazole to a cytotoxin. Spontaneous remyelination occurs after metronidazole-induced demyelinating responses. As tadpoles are transparent, these events can be monitored in vivo and quantified. At the end of metronidazole-induced demyelination, tadpoles were screened in water containing the compounds tested. After 72 h, remyelination was assayed by counting numbers of oligodendrocytes per optic nerve. RESULTS: Among a battery of molecules tested, siponimod, a dual agonist of sphingosine-1-phosphate receptor 1 and 5, was among the most efficient favoring remyelination. Crispr/cas9 gene editing showed that the promyelinating effect of siponimod involves the sphingosine-1-phosphate receptor 5. CONCLUSION: This Xenopus transgenic line constitutes a simple in vivo screening platform for myelin repair therapeutics. We validated several known promyelinating compounds and demonstrated that the strong remyelinating efficacy of siponimod implicates the sphingosine-1-phosphate receptor 5.


Subject(s)
Azetidines/pharmacology , Benzyl Compounds/pharmacology , Disease Models, Animal , Receptors, Lysosphingolipid/agonists , Remyelination/drug effects , Animals , Animals, Genetically Modified , Female , Larva , Male , Remyelination/physiology , Xenopus
13.
Circulation ; 133(4): 409-21, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26659946

ABSTRACT

BACKGROUND: Sprouting angiogenesis is a key process driving blood vessel growth in ischemic tissues and an important drug target in a number of diseases, including wet macular degeneration and wound healing. Endothelial cells forming the sprout must develop front-rear polarity to allow sprout extension. The adaptor proteins Nck1 and 2 are known regulators of cytoskeletal dynamics and polarity, but their function in angiogenesis is poorly understood. Here, we show that the Nck adaptors are required for endothelial cell front-rear polarity and migration downstream of the angiogenic growth factors VEGF-A and Slit2. METHODS AND RESULTS: Mice carrying inducible, endothelial-specific Nck1/2 deletions fail to develop front-rear polarized vessel sprouts and exhibit severe angiogenesis defects in the postnatal retina and during embryonic development. Inactivation of NCK1 and 2 inhibits polarity by preventing Cdc42 and Pak2 activation by VEGF-A and Slit2. Mechanistically, NCK binding to ROBO1 is required for both Slit2- and VEGF-induced front-rear polarity. Selective inhibition of polarized endothelial cell migration by targeting Nck1/2 prevents hypersprouting induced by Notch or Bmp signaling inhibition, and pathological ocular neovascularization and wound healing, as well. CONCLUSIONS: These data reveal a novel signal integration mechanism involving NCK1/2, ROBO1/2, and VEGFR2 that controls endothelial cell front-rear polarity during sprouting angiogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Polarity/physiology , Endothelial Cells/physiology , Gene Deletion , Neovascularization, Physiologic/physiology , Oncogene Proteins/genetics , Adaptor Proteins, Signal Transducing/deficiency , Amino Acid Sequence , Animals , Gene Targeting/methods , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Oncogene Proteins/deficiency
14.
Dev Neurosci ; 37(3): 232-42, 2015.
Article in English | MEDLINE | ID: mdl-25896276

ABSTRACT

We have generated a Xenopus laevis transgenic line, MBP-GFP-NTR, allowing conditional ablation of myelin-forming oligodendrocytes. In this transgenic line the transgene is driven by the proximal portion of the myelin basic protein regulatory sequence, specific to mature oligodendrocytes. The transgene protein is formed by the green fluorescent protein reporter fused to the Escherichia coli nitroreductase (NTR) selection enzyme. The NTR enzyme converts the innocuous prodrug metronidazole (MTZ) to a cytotoxin. Ablation of oligodendrocytes by MTZ treatment of the tadpole induced demyelination, and here we show that myelin debris are subsequently eliminated by microglial cells. After cessation of MTZ treatment, remyelination proceeded spontaneously. We questioned the origin of remyelinating cells. Our data suggest that Sox10+ oligodendrocyte precursor cells (OPCs), which are already present in the optic nerve prior to the experimentally induced demyelination, are responsible for remyelination, and this required only minimal (if any) cell division of OPCs. © 2015 S. Karger AG, Basel.


Subject(s)
Demyelinating Diseases/metabolism , Microglia/metabolism , Myelin Sheath/metabolism , Nerve Regeneration/physiology , Oligodendroglia/metabolism , Animals , Animals, Genetically Modified , Anti-Infective Agents/pharmacology , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Disease Models, Animal , Metronidazole/pharmacology , Neural Stem Cells , Oligodendroglia/drug effects , Oligodendroglia/pathology , Xenopus laevis
15.
J Neurosci ; 33(23): 9752-9768, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23739972

ABSTRACT

Oligodendrocytes are the myelin-forming cells of the CNS. They differentiate from oligodendrocyte precursor cells (OPCs) that are produced from progenitors throughout life but more actively during the neonatal period and in response to demyelinating insults. An accurate regulation of oligodendrogenesis is required to generate oligodendrocytes during these developmental or repair processes. We hypothesized that this regulation implicates transcription factors, which are expressed by OPCs and/or their progenitors. Ascl1/Mash1 is a proneural transcription factor previously implicated in embryonic oligodendrogenesis and operating in genetic interaction with Olig2, an essential transcriptional regulator in oligodendrocyte development. Herein, we have investigated the contribution of Ascl1 to oligodendrocyte development and remyelination in the postnatal cortex. During the neonatal period, Ascl1 expression was detected in progenitors of the cortical subventricular zone and in cortical OPCs. Different genetic approaches to delete Ascl1 in cortical progenitors or OPCs reduced neonatal oligodendrogenesis, showing that Ascl1 positively regulated both OPC specification from subventricular zone progenitors as well as the balance between OPC differentiation and proliferation. Examination of remyelination processes, both in the mouse model for focal demyelination of the corpus callosum and in multiple sclerosis lesions in humans, indicated that Ascl1 activity was upregulated along with increased oligodendrogenesis observed in remyelinating lesions. Additional genetic evidence indicated that remyelinating oligodendrocytes derived from Ascl1(+) progenitors/OPCs and that Ascl1 was required for proper remyelination. Together, our results show that Ascl1 function modulates multiple steps of OPC development in the postnatal brain and in response to demyelinating insults.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Brain/physiology , Myelin Sheath/physiology , Oligodendroglia/metabolism , Animals , Brain/cytology , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Nerve Fibers, Myelinated/metabolism , Neural Stem Cells/metabolism , Oligodendroglia/cytology
16.
Development ; 138(22): 4991-5001, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22028031

ABSTRACT

Homeoprotein transcription factors play fundamental roles in development, ranging from embryonic polarity to cell differentiation and migration. Research in recent years has underscored the physiological importance of homeoprotein intercellular transfer in eye field development, axon guidance and retino-tectal patterning, and visual cortex plasticity. Here, we have used the embryonic chick neural tube to investigate a possible role for homeoprotein Pax6 transfer in oligodendrocyte precursor cell (OPC) migration. We report the extracellular expression of Pax6 and the effects of gain and loss of extracellular Pax6 activity on OPCs. Open book cultures with recombinant Pax6 protein or Pax6 blocking antibodies, as well as in ovo gene transfer experiments involving expression of secreted Pax6 protein or secreted Pax6 antibodies, provide converging evidences that OPC migration is promoted by extracellular Pax6. The paracrine effect of Pax6 on OPC migration is thus a new example of direct non-cell autonomous homeoprotein activity.


Subject(s)
Cell Movement/genetics , Eye Proteins/physiology , Homeodomain Proteins/physiology , Neural Tube/embryology , Oligodendroglia/physiology , Paired Box Transcription Factors/physiology , Paracrine Communication , Repressor Proteins/physiology , Animals , Animals, Genetically Modified , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/drug effects , Chick Embryo , Extracellular Space/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Eye Proteins/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/pharmacology , Nerve Tissue Proteins/metabolism , Neural Tube/cytology , Neural Tube/metabolism , Neural Tube/physiology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Paired Box Transcription Factors/pharmacology , Paracrine Communication/physiology , Protein Transport/genetics , Protein Transport/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/physiology , Substrate Specificity , Tissue Distribution
17.
Circ Res ; 111(4): 437-45, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22723296

ABSTRACT

RATIONALE: The lymphatic vasculature plays a major role in fluid homeostasis, absorption of dietary lipids, and immune surveillance. Fluid transport depends on the presence of intraluminal valves within lymphatic collectors. Defective formation of lymphatic valves leads to lymphedema, a progressive and debilitating condition for which curative treatments are currently unavailable. How lymphatic valve formation is regulated remains largely unknown. OBJECTIVE: We investigated if the repulsive axon guidance molecule Semaphorin3A (Sema3A) plays a role in lymphatic valve formation. METHODS AND RESULTS: We show that Sema3A mRNA is expressed in lymphatic vessels and that Sema3A protein binds to lymphatic valves expressing the Neuropilin-1 (Nrp1) and PlexinA1 receptors. Using mouse knockout models, we show that Sema3A is selectively required for lymphatic valve formation, via interaction with Nrp1 and PlexinA1. Sema3a(-/-) mice exhibit defects in lymphatic valve formation, which are not due to abnormal lymphatic patterning or sprouting, and mice carrying a mutation in the Sema3A binding site of Nrp1, or deficient for Plxna1, develop lymphatic valve defects similar to those seen in Sema3a(-/-) mice. CONCLUSIONS: Our data demonstrate an essential direct function of Sema3A-Nrp1-PlexinA1 signaling in lymphatic valve formation.


Subject(s)
Lymphatic Vessels/metabolism , Nerve Tissue Proteins/metabolism , Neuropilin-1/metabolism , Receptors, Cell Surface/metabolism , Semaphorin-3A/metabolism , Animals , Animals, Newborn , Antibodies, Neutralizing/administration & dosage , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Genotype , Gestational Age , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lymphatic Vessels/embryology , Mice , Mice, Knockout , Mice, Transgenic , Morphogenesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neuropilin-1/deficiency , Neuropilin-1/genetics , Neuropilin-1/immunology , Phenotype , RNA, Messenger/metabolism , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Semaphorin-3A/deficiency , Semaphorin-3A/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
18.
Cell Mol Life Sci ; 70(10): 1673-4, 2013 May.
Article in English | MEDLINE | ID: mdl-23475064

ABSTRACT

Vascular endothelial growth factors (VEGFs) and their high-affinity tyrosine kinase VEGF receptors (VEGFRs) are key regulators of both angiogenesis and neurogenesis. The current issue of CMLS discusses recent literature and work implementing these signals in nervous system development, maintenance and disease pathology.


Subject(s)
Vascular Endothelial Growth Factors/metabolism , Animals , Humans , Neovascularization, Pathologic , Neurogenesis , Receptors, Vascular Endothelial Growth Factor/metabolism
19.
Cell Mol Life Sci ; 70(10): 1779-92, 2013 May.
Article in English | MEDLINE | ID: mdl-23479133

ABSTRACT

Notch cell interaction mechanism governs cell fate decisions in many different cell contexts throughout the lifetime of all Metazoan species. It links the fate of one cell to that of its neighbors through cell-to-cell contacts, and binding of Notch receptors expressed on one cell to their membrane bound ligands on an adjacent cell. Environmental cues, such as growth factors and extracellular matrix molecules, superimpose a dynamic regulation on this canonical Notch signaling pathway. In this review, we will focus on Notch signaling in the vertebrate vascular and nervous systems and examine its role in angiogenesis, neurogenesis, and neurovascular interactions. We will also highlight the molecular relationships of the Notch pathway with vascular endothelial growth factors (VEGFs) and their high-affinity tyrosine kinase VEGF receptors, key regulators of both angiogenesis and neurogenesis.


Subject(s)
Endothelial Cells/metabolism , Neurons/metabolism , Receptors, Notch/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Endothelial Cells/cytology , Humans , Neovascularization, Pathologic/metabolism , Neurogenesis , Neurons/cytology , Signal Transduction , Vascular Endothelial Growth Factors/metabolism
20.
Proc Natl Acad Sci U S A ; 108(42): 17498-503, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-21969550

ABSTRACT

The six members of the contactin (CNTN) family of neural cell adhesion molecules are involved in the formation and maintenance of the central nervous system (CNS) and have been linked to mental retardation and neuropsychiatric disorders such as autism. Five of the six CNTNs bind to the homologous receptor protein tyrosine phosphatases gamma (PTPRG) and zeta (PTPRZ), but the biological roles of these interactions remain unclear. We report here the cocrystal structure of the carbonic anhydrase-like domain of PTPRZ bound to tandem Ig repeats of CNTN1 and combine these structural data with binding assays to show that PTPRZ binds specifically to CNTN1 expressed at the surface of oligodendrocyte precursor cells. Furthermore, analyses of glial cell populations in wild-type and PTPRZ-deficient mice show that the binding of PTPRZ to CNTN1 expressed at the surface of oligodendrocyte precursor cells inhibits their proliferation and promotes their development into mature oligodendrocytes. Overall, these results implicate the PTPRZ/CNTN1 complex as a previously unknown modulator of oligodendrogenesis.


Subject(s)
Contactin 1/chemistry , Contactin 1/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Animals , Binding Sites , Cell Differentiation , Cell Proliferation , Contactin 1/genetics , Crystallography, X-Ray , Humans , Mice , Mice, Knockout , Models, Molecular , Models, Neurological , Multiprotein Complexes , Neurogenesis/genetics , Neurogenesis/physiology , Protein Structure, Tertiary , Receptor-Like Protein Tyrosine Phosphatases, Class 5/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL