Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(9): 113020, 2023 Sep 26.
Article in English | MEDLINE | ID: mdl-37610874

ABSTRACT

Structural and functional deficits in brain connectivity are reported in patients with fetal alcohol spectrum disorders (FASDs), but whether and how prenatal alcohol exposure (PAE) affects axonal development of neurons and disrupts wiring between brain regions is unknown. Here, we develop a mouse model of moderate alcohol exposure during prenatal brain wiring to study the effects of PAE on corpus callosum (CC) development. PAE induces aberrant navigation of interhemispheric CC axons that persists even after exposure ends, leading to ectopic termination in the contralateral cortex. The neuronal miR-17-5p and its target ephrin type A receptor 4 (EphA4) mediate the effect of alcohol on the contralateral targeting of CC axons. Thus, altered microRNA-mediated regulation of axonal guidance may have implications for interhemispheric cortical connectivity and associated behaviors in FASD.

2.
Liver Int ; 43(9): 1850-1864, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37402699

ABSTRACT

BACKGROUND AND AIMS: The liver is an innervated organ that develops a variety of chronic liver disease (CLD). Axon guidance cues (AGCs), of which ephrins, netrins, semaphorins and slits are the main representative, are secreted or membrane-bound proteins that can attract or repel axons through interactions with their growth cones that contain receptors recognizing these messengers. While fundamentally implicated in the physiological development of the nervous system, the expression of AGCs can also be reinduced under acute or chronic conditions, such as CLD, that necessitate redeployment of neural networks. METHODS: This review considers the ad hoc literature through the neglected canonical neural function of these proteins that is also applicable to the diseased liver (and not solely their observed parenchymal impact). RESULTS: AGCs impact fibrosis regulation, immune functions, viral/host interactions, angiogenesis, and cell growth, both at the CLD and HCC levels. Special attention has been paid to distinguishing correlative and causal data in such datasets in order to streamline data interpretation. While hepatic mechanistic insights are to date limited, bioinformatic evidence for the identification of AGCs mRNAs positive cells, protein expression, quantitative regulation, and prognostic data have been provided. Liver-pertinent clinical studies based on the US Clinical Trials database are listed. Future research directions derived from AGC targeting are proposed. CONCLUSION: This review highlights frequent implication of AGCs in CLD, linking traits of liver disorders and the local autonomic nervous system. Such data should contribute to diversifying current parameters of patient stratification and our understanding of CLD.

3.
J Theor Biol ; 556: 111301, 2023 01 07.
Article in English | MEDLINE | ID: mdl-36270328

ABSTRACT

The nervous system is today recognized to play an important role in the development of cancer. Indeed, neurons extend long processes (axons) that grow and infiltrate tumors in order to regulate the progression of the disease in a positive or negative way, depending on the type of neuron considered. Mathematical modeling of this biological process allows to formalize the nerve-tumor interactions and to test hypotheses in silico to better understand this phenomenon. In this work, we introduce a system of differential equations modeling the progression of pancreatic ductal adenocarcinoma (PDAC) coupled with associated changes in axonal innervation. The study of the asymptotic behavior of the model confirms the experimental observations that PDAC development is correlated with the type and densities of axons in the tissue. We study then the identifiability and the sensitivity of the model parameters. The identifiability analysis informs on the adequacy between the parameters of the model and the experimental data and the sensitivity analysis on the most contributing factors on the development of cancer. It leads to significant insights on the main neural checkpoints and mechanisms controlling the progression of pancreatic cancer. Finally, we give an example of a simulation of the effects of partial or complete denervation that sheds lights on complex correlation between the healthy, pre-cancerous and cancerous cell densities and axons with opposite functions.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Axons , Cell Transformation, Neoplastic , Carcinogenesis , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Pancreatic Neoplasms
4.
Mol Cell Neurosci ; 120: 103726, 2022 05.
Article in English | MEDLINE | ID: mdl-35367368

ABSTRACT

VPS35 is a core component of the retromer complex involved in familial forms of neurodegenerative diseases such as Parkinson's and Alzheimer's disease. In mice, VPS35 is expressed during early brain development. However, previous studies have reported that VPS35 activity is largely dispensable for normal neuronal development and initial elaboration of axonal projections. Here, we evaluated the role of VPS35 in the mouse embryonic brain using two Cre-driver lines that remove Vps35 from the cortex at different prenatal stages. We found that Vps35 mutant mice displayed microcephaly and decreased cortical thickness from the embryonic stages to adulthood. VPS35 also regulates cortical development by affecting a subpopulation of neural progenitor cells and the survival of postmitotic neurons. In addition, we showed that a lack of VPS35 leads to hypoplasia and misrouting of several axonal projections, including the anterior commissure and fornix. Furthermore, VPS35 deficiency impairs the non-autonomous development of thalamocortical axons (TCAs), which show severe disruption of innervation and terminal arborization in the cortex. Together, these data demonstrate that VPS35 plays a greater role in embryonic development of the mammalian brain than it was previously thought.


Subject(s)
Neurodegenerative Diseases , Vesicular Transport Proteins , Animals , Axons/metabolism , Mammals , Mice , Neurodegenerative Diseases/metabolism , Neurogenesis , Neurons/metabolism , Vesicular Transport Proteins/metabolism
5.
Nat Commun ; 13(1): 1985, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35418199

ABSTRACT

Neuronal nerve processes in the tumor microenvironment were highlighted recently. However, the origin of intra-tumoral nerves remains poorly known, in part because of technical difficulties in tracing nerve fibers via conventional histological preparations. Here, we employ three-dimensional (3D) imaging of cleared tissues for a comprehensive analysis of sympathetic innervation in a murine model of pancreatic ductal adenocarcinoma (PDAC). Our results support two independent, but coexisting, mechanisms: passive engulfment of pre-existing sympathetic nerves within tumors plus an active, localized sprouting of axon terminals into non-neoplastic lesions and tumor periphery. Ablation of the innervating sympathetic nerves increases tumor growth and spread. This effect is explained by the observation that sympathectomy increases intratumoral CD163+ macrophage numbers, which contribute to the worse outcome. Altogether, our findings provide insights into the mechanisms by which the sympathetic nervous system exerts cancer-protective properties in a mouse model of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Macrophages , Mice , Sympathetic Nervous System/physiology , Tumor Microenvironment , Pancreatic Neoplasms
6.
Cancer Discov ; 12(4): 1128-1151, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34930787

ABSTRACT

Gut dysbiosis has been associated with intestinal and extraintestinal malignancies, but whether and how carcinogenesis drives compositional shifts of the microbiome to its own benefit remains an open conundrum. Here, we show that malignant processes can cause ileal mucosa atrophy, with villous microvascular constriction associated with dominance of sympathetic over cholinergic signaling. The rapid onset of tumorigenesis induced a burst of REG3γ release by ileal cells, and transient epithelial barrier permeability that culminated in overt and long-lasting dysbiosis dominated by Gram-positive Clostridium species. Pharmacologic blockade of ß-adrenergic receptors or genetic deficiency in Adrb2 gene, vancomycin, or cohousing of tumor bearers with tumor-free littermates prevented cancer-induced ileopathy, eventually slowing tumor growth kinetics. Patients with cancer harbor distinct hallmarks of this stress ileopathy dominated by Clostridium species. Hence, stress ileopathy is a corollary disease of extraintestinal malignancies requiring specific therapies. SIGNIFICANCE: Whether gut dysbiosis promotes tumorigenesis and how it controls tumor progression remain open questions. We show that 50% of transplantable extraintestinal malignancies triggered a ß-adrenergic receptor-dependent ileal mucosa atrophy, associated with increased gut permeability, sustained Clostridium spp.-related dysbiosis, and cancer growth. Vancomycin or propranolol prevented cancer-associated stress ileopathy. This article is highlighted in the In This Issue feature, p. 873.


Subject(s)
Dysbiosis , Receptors, Adrenergic, beta , Carcinogenesis/pathology , Dysbiosis/chemically induced , Dysbiosis/complications , Dysbiosis/pathology , Humans , Intestinal Mucosa/pathology , Signal Transduction
7.
Mol Cell Neurosci ; 100: 103397, 2019 10.
Article in English | MEDLINE | ID: mdl-31454665

ABSTRACT

The corpus callosum is the largest bundle of commissural fibres that transfer information between the two cerebral hemispheres. Callosal projection neurons (CPNs) are a diverse population of pyramidal neurons within the neocortex that mainly interconnect homotopic regions of the opposite cortices. Nevertheless, some CPNs are involved in heterotopic projections between distinct cortical areas or to subcortical regions such as the striatum. In this study, we showed that the axon guidance receptor PlexinD1 is expressed by a large proportion of heterotopically projecting CPNs in layer 5A of the primary somatosensory (S1) and motor (M1) areas. Retrograde tracing of M1 CPNs projecting to the contralateral striatum revealed the presence of ectopic neurons aberrantly located in layers 2/3 of Plxnd1 and Sema3e mutant cortices. These results showed that Sema3E/PlexinD1 signalling controls the laminar distribution of heterotopically projecting CPNs.


Subject(s)
Corpus Callosum/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Glycoproteins/metabolism , Neurons/metabolism , Semaphorins/metabolism , Animals , Corpus Callosum/metabolism , Female , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Motor Cortex/cytology , Motor Cortex/metabolism , Neuroanatomical Tract-Tracing Techniques , Neurons/cytology , Neurons/physiology , Semaphorins/genetics , Somatosensory Cortex/cytology , Somatosensory Cortex/metabolism
8.
Curr Opin Neurobiol ; 53: 183-191, 2018 12.
Article in English | MEDLINE | ID: mdl-30273799

ABSTRACT

Twenty-five years after the discovery of the first chemotropic molecules for growing axons, what are the new findings? This review describes the latest progress made in our understanding of the molecular control of axonal guidance in the vertebrate nervous system. Special focus will be given to new molecular players, their source and location in vivo, and the role of membrane/receptor trafficking and RNA-based mechanisms in axon guidance cue signalling.


Subject(s)
Axon Guidance/physiology , Axons/metabolism , Biological Transport/physiology , Central Nervous System/growth & development , Nerve Net/growth & development , Animals , Humans
9.
Curr Biol ; 28(11): 1768-1782.e4, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29779877

ABSTRACT

The corpus callosum is the largest commissure in the brain, whose main function is to ensure communication between homotopic regions of the cerebral cortex. During fetal development, corpus callosum axons (CCAs) grow toward and across the brain midline and then away on the contralateral hemisphere to their targets. A particular feature of this circuit, which raises a key developmental question, is that the outgoing trajectory of post-crossing CCAs is mirror-symmetric with the incoming trajectory of pre-crossing axons. Here, we show that post-crossing CCAs switch off their response to axon guidance cues, among which the secreted Semaphorin-3C (Sema3C), that act as attractants for pre-crossing axons on their way to the midline. This change is concomitant with an upregulation of the surface protein Ephrin-B1, which acts in CCAs to inhibit Sema3C signaling via interaction with the Neuropilin-1 (Nrp1) receptor. This silencing activity is independent of Eph receptors and involves a N-glycosylation site (N-139) in the extracellular domain of Ephrin-B1. Together, our results reveal a molecular mechanism, involving interaction between the two unrelated guidance receptors Ephrin-B1 and Nrp1, that is used to control the navigation of post-crossing axons in the corpus callosum.


Subject(s)
Axons/physiology , Corpus Callosum/physiology , Ephrin-B1/genetics , Gene Expression Regulation, Developmental , Neuropilin-1/genetics , Semaphorins/genetics , Animals , Ephrin-B1/metabolism , Gene Silencing , Mice , Neuropilin-1/metabolism , Semaphorins/metabolism
10.
J Med Virol ; 89(11): 2051-2054, 2017 11.
Article in English | MEDLINE | ID: mdl-28617960

ABSTRACT

The aim of the study was to assess whether high-risk pregnant women have a higher prevalence of HEV during the perinatal period. This was a cross-sectional study of 428 patients: Group 1, 127 women with a high-risk pregnancy; Group 2, 97 asymptomatic people with reactivity to HCV or HBV; Group 3, 94 patients with clinical symptoms suggestive of HEV infection; and Group 4, 110 healthy blood donors from an urban area of Mexico City. ELISA was used to measure antibody to HEV genotypes 1 and 3. The prevalence rates of anti-HEV IgG antibodies were 0.79% in Group 1, 2.1% in Group 2, 7.4% in Group 3, and 0% in Group 4. Women with a high-risk pregnancy did not have a higher prevalence of HEV infection in this clinical setting.


Subject(s)
Hepatitis Antibodies/blood , Hepatitis E virus/genetics , Hepatitis E virus/immunology , Hepatitis E/epidemiology , Pregnancy Complications, Infectious/epidemiology , Pregnancy, High-Risk , Adolescent , Adult , Antigens, Viral/immunology , Asymptomatic Infections/epidemiology , Blood Donors , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Female , Genotype , Hepatitis Antibodies/immunology , Hepatitis E/immunology , Hepatitis E/virology , Humans , Immunoglobulin G/blood , Mexico/epidemiology , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Prevalence , Young Adult
11.
Nat Commun ; 8: 14508, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28224988

ABSTRACT

Local endocytic events involving receptors for axon guidance cues play a central role in controlling growth cone behaviour. Yet, little is known about the fate of internalized receptors, and whether the sorting events directing them to distinct endosomal pathways control guidance decisions. Here, we show that the receptor Plexin-D1 contains a sorting motif that interacts with the adaptor protein GIPC1 to facilitate transport to recycling endosomes. This sorting process promotes colocalization of Plexin-D1 with vesicular pools of active R-ras, leading to its inactivation. In the absence of interaction with GIPC1, missorting of Plexin-D1 results in loss of signalling activity. Consequently, Gipc1 mutant mice show specific defects in axonal projections, as well as vascular structures, that rely on Plexin-D1 signalling for their development. Thus, intracellular sorting steps that occur after receptor internalization by endocytosis provide a critical level of control of cellular responses to guidance signals.


Subject(s)
Axons/metabolism , Blood Vessels/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Endocytosis , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Animals , Body Patterning , Cell Adhesion Molecules, Neuronal/chemistry , Cell Membrane/metabolism , Cytoskeletal Proteins , Endosomes/metabolism , Epistasis, Genetic , Female , Glycoproteins/metabolism , Green Fluorescent Proteins/metabolism , Growth Cones/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Male , Membrane Glycoproteins/chemistry , Membrane Proteins/metabolism , Mice , Nerve Tissue Proteins/chemistry , PDZ Domains , Protein Transport , Semaphorins , ras Proteins/metabolism
12.
Methods Mol Biol ; 1493: 223-235, 2017.
Article in English | MEDLINE | ID: mdl-27787854

ABSTRACT

Semaphorin guidance molecules act through different receptor complexes to activate multiple signaling cascades leading to changes in axonal growth cone behavior and morphology. We describe here approaches for studying the effect of individual Semaphorins on isolated forebrain neurons from mouse embryos and dissecting downstream signaling pathways. These approaches include the production of recombinant Semaphorin ligands, the culture of dissociated primary neurons, the manipulation of gene expression by electroporation in primary neurons, and functional assays to assess axon outgrowth and growth cone collapse.


Subject(s)
Neurons/metabolism , Semaphorins/metabolism , Signal Transduction , Animals , Axons/metabolism , Cells, Cultured , Electroporation , Gene Expression , Growth Cones/metabolism , HEK293 Cells , Humans , Mice , Prosencephalon/cytology , Prosencephalon/metabolism , Transfection
13.
Nat Commun ; 6: 7246, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26037503

ABSTRACT

Structural microtubule associated proteins (MAPs) stabilize microtubules, a property that was thought to be essential for development, maintenance and function of neuronal circuits. However, deletion of the structural MAPs in mice does not lead to major neurodevelopment defects. Here we demonstrate a role for MAP6 in brain wiring that is independent of microtubule binding. We find that MAP6 deletion disrupts brain connectivity and is associated with a lack of post-commissural fornix fibres. MAP6 contributes to fornix development by regulating axonal elongation induced by Semaphorin 3E. We show that MAP6 acts downstream of receptor activation through a mechanism that requires a proline-rich domain distinct from its microtubule-stabilizing domains. We also show that MAP6 directly binds to SH3 domain proteins known to be involved in neurite extension and semaphorin function. We conclude that MAP6 is critical to interface guidance molecules with intracellular signalling effectors during the development of cerebral axon tracts.


Subject(s)
Axons/metabolism , Fornix, Brain/embryology , Glycoproteins/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Neurons/metabolism , Animals , Brain/metabolism , Brain/pathology , Cytoskeletal Proteins , Diffusion Tensor Imaging , Fornix, Brain/metabolism , Fornix, Brain/pathology , HEK293 Cells , Humans , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Neural Pathways/embryology , Neural Pathways/metabolism , Neurites/metabolism , Neuroanatomical Tract-Tracing Techniques , Organ Size , Semaphorins , src Homology Domains
14.
J Clin Invest ; 125(6): 2413-28, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25985275

ABSTRACT

Individuals with an inherited deficiency in gonadotropin-releasing hormone (GnRH) have impaired sexual reproduction. Previous genetic linkage studies and sequencing of plausible gene candidates have identified mutations associated with inherited GnRH deficiency, but the small number of affected families and limited success in validating candidates have impeded genetic diagnoses for most patients. Using a combination of exome sequencing and computational modeling, we have identified a shared point mutation in semaphorin 3E (SEMA3E) in 2 brothers with Kallmann syndrome (KS), which causes inherited GnRH deficiency. Recombinant wild-type SEMA3E protected maturing GnRH neurons from cell death by triggering a plexin D1-dependent (PLXND1-dependent) activation of PI3K-mediated survival signaling. In contrast, recombinant SEMA3E carrying the KS-associated mutation did not protect GnRH neurons from death. In murine models, lack of either SEMA3E or PLXND1 increased apoptosis of GnRH neurons in the developing brain, reducing innervation of the adult median eminence by GnRH-positive neurites. GnRH neuron deficiency in male mice was accompanied by impaired testes growth, a characteristic feature of KS. Together, these results identify SEMA3E as an essential gene for GnRH neuron development, uncover a neurotrophic function for SEMA3E in the developing brain, and elucidate SEMA3E/PLXND1/PI3K signaling as a mechanism that prevents GnRH neuron deficiency.


Subject(s)
Glycoproteins/metabolism , Gonadotropin-Releasing Hormone/deficiency , Kallmann Syndrome/metabolism , Membrane Proteins/metabolism , Mutation , Neurons/metabolism , Semaphorins/metabolism , Adult , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cytoskeletal Proteins , Exome , Glycoproteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Kallmann Syndrome/genetics , Kallmann Syndrome/pathology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Semaphorins/genetics , Signal Transduction/genetics
15.
Nat Commun ; 5: 4265, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24969029

ABSTRACT

During the development of the cerebral cortex, Cajal-Retzius (CR) cells settle in the preplate and coordinate the precise growth of the neocortex. Indeed, CR cells migrate tangentially from specific proliferative regions of the telencephalon (for example, the cortical hem (CH)) to populate the entire cortical surface. This is a very finely tuned process regulated by an emerging number of factors that has been sequentially revealed in recent years. However, the putative participation of one of the major families of axon guidance molecules in this process, the Semaphorins, was not explored. Here we show that Semaphorin-3E (Sema3E) is a natural negative regulator of the migration of PlexinD1-positive CR cells originating in the CH. Our results also indicate that Sema3E/PlexinD1 signalling controls the motogenic potential of CR cells in vitro and in vivo. Indeed, absence of Sema3E/PlexinD1 signalling increased the migratory properties of CR cells. This modulation implies negative effects on CXCL12/CXCR4 signalling and increased ADF/Cofilin activity.


Subject(s)
Cell Movement , Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Neocortex/embryology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , RNA, Messenger/genetics , Actin Depolymerizing Factors/metabolism , Animals , Cerebral Cortex/embryology , Chemokine CXCL12/metabolism , Cytoskeletal Proteins , Destrin/metabolism , Gene Expression Regulation, Developmental , Glycoproteins/genetics , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Neurons/cytology , Receptors, CXCR4/metabolism , Semaphorins , Signal Transduction
16.
Neuron ; 80(2): 262-5, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24139030

ABSTRACT

In this issue of Neuron, Oh and Gu (2013) present a model in which intimately related embryonic nerves and blood vessels are patterned independently in response to different guidance cues from a central organizer: the whisker.


Subject(s)
Blood Vessels/physiology , Body Patterning/physiology , Sensory Receptor Cells/physiology , Vibrissae/innervation , Vibrissae/physiology , Animals
17.
Cancer Cell ; 24(5): 673-85, 2013 Nov 11.
Article in English | MEDLINE | ID: mdl-24139859

ABSTRACT

The semaphorin guidance molecules and their receptors, the plexins, are often inappropriately expressed in cancers. However, the signaling processes mediated by plexins in tumor cells are still poorly understood. Here, we demonstrate that the Semaphorin 3E (Sema3E) regulates tumor cell survival by suppressing an apoptotic pathway triggered by the Plexin D1 dependence receptor. In mouse models of breast cancer, a ligand trap that sequesters Sema3E inhibited tumor growth and reduced metastasis through a selective tumor cytocidal effect. We further showed that Plexin D1 triggers apoptosis via interaction with the orphan nuclear receptor NR4A1. These results define a critical role of Sema3E/Plexin D1 interaction in tumor resistance to apoptosis and suggest a therapeutic approach based on activation of a dependence receptor pathway.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Cell Adhesion Molecules, Neuronal/physiology , Lung Neoplasms/secondary , Semaphorins/physiology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Female , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Membrane Glycoproteins , Mice , Mice, Inbred BALB C , Mice, Nude , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptide Fragments/physiology , Protein Interaction Domains and Motifs , Semaphorins/chemistry , Semaphorins/pharmacology , Signal Transduction , Xenograft Model Antitumor Assays
18.
Cell Mol Life Sci ; 70(10): 1685-703, 2013 May.
Article in English | MEDLINE | ID: mdl-23475066

ABSTRACT

Many organs, such as lungs, nerves, blood and lymphatic vessels, consist of complex networks that carry flows of information, gases, and nutrients within the body. The morphogenetic patterning that generates these organs involves the coordinated action of developmental signaling cues that guide migration of specialized cells. Precision guidance of endothelial tip cells by vascular endothelial growth factors (VEGFs) is well established, and several families of neural guidance molecules have been identified to exert guidance function in both the nervous and the vascular systems. This review discusses recent advances in VEGF research, focusing on the emerging role of neural guidance molecules as key regulators of VEGF function during vascular development and on the novel role of VEGFs in neural cell migration and nerve wiring.


Subject(s)
Blood Vessels/metabolism , Lymphatic Vessels/metabolism , Neurons/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Axons/metabolism , Humans , Neuropilins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Eph Family/metabolism , Semaphorins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
Neuron ; 77(3): 472-84, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23395374

ABSTRACT

Major outputs of the neocortex are conveyed by corticothalamic axons (CTAs), which form reciprocal connections with thalamocortical axons, and corticosubcerebral axons (CSAs) headed to more caudal parts of the nervous system. Previous findings establish that transcriptional programs define cortical neuron identity and suggest that CTAs and thalamic axons may guide each other, but the mechanisms governing CTA versus CSA pathfinding remain elusive. Here, we show that thalamocortical axons are required to guide pioneer CTAs away from a default CSA-like trajectory. This process relies on a hold in the progression of cortical axons, or waiting period, during which thalamic projections navigate toward cortical axons. At the molecular level, Sema3E/PlexinD1 signaling in pioneer cortical neurons mediates a "waiting signal" required to orchestrate the mandatory meeting with reciprocal thalamic axons. Our study reveals that temporal control of axonal progression contributes to spatial pathfinding of cortical projections and opens perspectives on brain wiring.


Subject(s)
Cerebral Cortex/physiology , Neural Pathways/physiology , Thalamus/physiology , Age Factors , Animals , Axons/physiology , Body Patterning/genetics , Calbindin 2 , Cerebral Cortex/cytology , Contactin 2/metabolism , Cytoskeletal Proteins , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Glycoproteins/genetics , Homeodomain Proteins/genetics , Intracellular Signaling Peptides and Proteins , Leukocyte L1 Antigen Complex/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , POU Domain Factors/genetics , Repressor Proteins/metabolism , S100 Calcium Binding Protein G/metabolism , Semaphorins , T-Box Domain Proteins , Thalamus/cytology , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Wnt3A Protein/genetics , tau Proteins/genetics
20.
Circ Res ; 110(1): 34-46, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22076636

ABSTRACT

RATIONALE: Positive signals, such as vascular endothelial growth factor, direct endothelial cells (ECs) to specific locations during blood vessel formation. Less is known about repulsive signal contribution to shaping vessels. Recently, "neuronal guidance cues" have been shown to influence EC behavior, particularly in directing sprouting angiogenesis by repelling ECs. However, their role during de novo blood vessel formation remains unexplored. OBJECTIVE: To identify signals that guide and pattern the first mammalian blood vessels. METHODS AND RESULTS: Using genetic mouse models, we show that blood vessels are sculpted through the generation of stereotyped avascular zones by EC-repulsive cues. We demonstrate that Semaphorin3E (Sema3E) is a key factor that shapes the paired dorsal aortae in mouse, as sema3E(-/-) embryos develop an abnormally branched aortic plexus with a markedly narrowed avascular midline. In vitro cultures and avian grafting experiments show strong repulsion of ECs by Sema3E-expressing cells. We further identify the mouse notochord as a rich source of multiple redundant neuronal guidance cues. Mouse embryos that lack notochords fail to form cohesive aortic vessels because of loss of the avascular midline, yet maintain lateral avascular zones. We demonstrate that lateral avascular zones are directly generated by the lateral plate mesoderm, a critical source of Sema3E. CONCLUSIONS: These findings demonstrate that Sema3E-generated avascular zones are critical regulators of mammalian cardiovascular patterning and are the first to identify a repulsive role for the lateral plate mesoderm. Integration of multiple, and in some cases redundant, repulsive cues from various tissues is critical to patterning the first embryonic blood vessels.


Subject(s)
Blood Vessels/embryology , Embryo, Mammalian/blood supply , Endothelium, Vascular/embryology , Glycoproteins/physiology , Membrane Proteins/physiology , Neovascularization, Physiologic/physiology , Signal Transduction/physiology , Animals , Aorta/cytology , Aorta/embryology , Blood Vessels/cytology , Cells, Cultured , Cytoskeletal Proteins , Endothelium, Vascular/cytology , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/physiology , Glycoproteins/deficiency , Glycoproteins/genetics , Hepatocyte Nuclear Factor 3-beta/deficiency , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/physiology , In Vitro Techniques , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mesoderm/cytology , Mesoderm/embryology , Mice , Mice, Knockout , Models, Animal , Notochord/cytology , Notochord/embryology , Semaphorins
SELECTION OF CITATIONS
SEARCH DETAIL
...