Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Immunol ; 204(1): 128-136, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31776203

ABSTRACT

Semaphorin 3E (Sema3E) is a secreted protein that was initially discovered as a neuronal guidance cue. Recent evidence showed that Sema3E plays an essential role in regulating the activities of various immune cells. However, the exact role of Sema3E in macrophage function, particularly during inflammation, is not fully understood. We studied the impact of Sema3E gene deletion on macrophage function during the LPS-induced acute inflammatory response. We found that Sema3E-deficient (Sema3e-/- ) mice were better protected from LPS-induced acute inflammation as exemplified by their superior clinical score and effective temperature control compared with their wild-type littermates. This superior control of inflammatory response in Sema3e-/- mice was associated with significantly lower phosphorylation of ERK1/2, AKT, STAT3, and NF-κB, and a concomitant reduction in inducible NO synthase expression and production of TNF and IL-6 compared with their Sema3e+/+ littermates. Sema3e-/- mice also contained significantly higher numbers of activated macrophages compared with their Sema3e+/+ littermates at both baselines and after LPS challenge. In vivo-specific deletion of the Sema3E high-affinity receptor, plexinD1, on macrophages led to the improvement in clinical disease following exposure to a lethal dose of LPS. Collectively, our data show that Sema3E plays an essential role in dampening the early inflammatory response to LPS by regulating macrophage function, suggesting an essential role of this pathway in macrophage inflammatory response.


Subject(s)
Inflammation/immunology , Macrophages/immunology , Semaphorins/immunology , Animals , Cells, Cultured , Inflammation/chemically induced , Lipopolysaccharides/administration & dosage , Mice , Mice, Knockout , Mice, Transgenic , Semaphorins/deficiency
2.
Cell Mol Life Sci ; 78(6): 2911-2927, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33128105

ABSTRACT

Descending serotonergic (5-HT) projections originating from the raphe nuclei form an important input to the spinal cord that control basic locomotion. The molecular signals that control this projection pattern are currently unknown. Here, we identify Semaphorin7A (Sema7A) as a critical cue that restricts serotonergic innervation in the spinal cord. Sema7A deficient mice show a marked increase in serotonergic fiber density in all layers of the spinal cord while the density of neurons expressing the corresponding 5-HTR2α receptor remains unchanged. These alterations appear to be successfully compensated as no obvious changes in rhythmic locomotion and skilled stepping are observed in adult mice. When the system is challenged with a spinal lesion, serotonergic innervation patterns in both Sema7A-deficient and -competent mice evolve over time with excessive innervation becoming most pronounced in the dorsal horn of Sema7A-deficient mice. These altered serotonergic innervation patterns correlate with diminished functional recovery that predominantly affects rhythmic locomotion. Our findings identify Sema7A as a critical regulator of serotonergic circuit formation in the injured spinal cord.


Subject(s)
Antigens, CD/metabolism , Recovery of Function , Semaphorins/metabolism , Spinal Cord Injuries/pathology , Animals , Antigens, CD/genetics , Behavior, Animal , Disease Models, Animal , Female , Locomotion , Male , Mice , Mice, Knockout , Semaphorins/deficiency , Semaphorins/genetics , Serotonin/metabolism , Signal Transduction , Spinal Cord/diagnostic imaging , Spinal Cord/metabolism , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Dorsal Horn/pathology , Spinal Cord Injuries/metabolism
3.
Hum Genet ; 140(8): 1183-1200, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34076780

ABSTRACT

Dyslexia is a common heritable developmental disorder involving impaired reading abilities. Its genetic underpinnings are thought to be complex and heterogeneous, involving common and rare genetic variation. Multigenerational families segregating apparent monogenic forms of language-related disorders can provide useful entrypoints into biological pathways. In the present study, we performed a genome-wide linkage scan in a three-generational family in which dyslexia affects 14 of its 30 members and seems to be transmitted with an autosomal dominant pattern of inheritance. We identified a locus on chromosome 7q21.11 which cosegregated with dyslexia status, with the exception of two cases of phenocopy (LOD = 2.83). Whole-genome sequencing of key individuals enabled the assessment of coding and noncoding variation in the family. Two rare single-nucleotide variants (rs144517871 and rs143835534) within the first intron of the SEMA3C gene cosegregated with the 7q21.11 risk haplotype. In silico characterization of these two variants predicted effects on gene regulation, which we functionally validated for rs144517871 in human cell lines using luciferase reporter assays. SEMA3C encodes a secreted protein that acts as a guidance cue in several processes, including cortical neuronal migration and cellular polarization. We hypothesize that these intronic variants could have a cis-regulatory effect on SEMA3C expression, making a contribution to dyslexia susceptibility in this family.


Subject(s)
Dyslexia/genetics , Genetic Predisposition to Disease , Inheritance Patterns , Polymorphism, Single Nucleotide , Semaphorins/genetics , Base Sequence , Cell Movement , Chromosomes, Human, Pair 7 , Dyslexia/diagnostic imaging , Dyslexia/metabolism , Dyslexia/physiopathology , Family , Female , Gene Expression , Genes, Dominant , Genetic Linkage , Genetic Loci , Genome-Wide Association Study , Haplotypes , Humans , Introns , Lod Score , Male , Neuroimaging , Neurons/metabolism , Neurons/pathology , Pedigree , Phenotype , Semaphorins/deficiency , Whole Genome Sequencing
4.
J Biol Chem ; 294(21): 8336-8347, 2019 05 24.
Article in English | MEDLINE | ID: mdl-30979723

ABSTRACT

Primary hyperparathyroidism (PHPT) is a common endocrinopathy characterized by hypercalcemia and elevated levels of parathyroid hormone. The primary cause of PHPT is a benign overgrowth of parathyroid tissue causing excessive secretion of parathyroid hormone. However, the molecular etiology of PHPT is incompletely defined. Here, we demonstrate that semaphorin3d (Sema3d), a secreted glycoprotein, is expressed in the developing parathyroid gland in mice. We also observed that genetic deletion of Sema3d leads to parathyroid hyperplasia, causing PHPT. In vivo and in vitro experiments using histology, immunohistochemistry, biochemical, RT-qPCR, and immunoblotting assays revealed that Sema3d inhibits parathyroid cell proliferation by decreasing the epidermal growth factor receptor (EGFR)/Erb-B2 receptor tyrosine kinase (ERBB) signaling pathway. We further demonstrate that EGFR signaling is elevated in Sema3d-/- parathyroid glands and that pharmacological inhibition of EGFR signaling can partially rescue the parathyroid hyperplasia phenotype. We propose that because Sema3d is a secreted protein, it may be possible to use recombinant Sema3d or derived peptides to inhibit parathyroid cell proliferation causing hyperplasia and hyperparathyroidism. Collectively, these findings identify Sema3d as a negative regulator of parathyroid growth.


Subject(s)
Cell Proliferation , Hyperparathyroidism, Primary/epidemiology , Parathyroid Glands/embryology , Semaphorins/deficiency , Signal Transduction , Animals , ErbB Receptors/genetics , ErbB Receptors/metabolism , Hyperparathyroidism, Primary/genetics , Hyperparathyroidism, Primary/pathology , Mice , Mice, Knockout , Parathyroid Glands/pathology , Semaphorins/metabolism
5.
Arterioscler Thromb Vasc Biol ; 38(2): 335-343, 2018 02.
Article in English | MEDLINE | ID: mdl-29269512

ABSTRACT

OBJECTIVE: Accumulating evidence suggests a role of semaphorins in vascular homeostasis. Here, we investigate the role of Sema7A (semaphorin 7A) in atherosclerosis and its underlying mechanism. APPROACH AND RESULTS: Using genetically engineered Sema7A-/-ApoE-/- mice, we showed that deletion of Sema7A attenuates atherosclerotic plaque formation primarily in the aorta of ApoE-/- mice on a high-fat diet. A higher level of Sema7A in the atheroprone lesser curvature suggests a correlation of Sema7A with disturbed flow. This notion is supported by elevated Sema7A expression in human umbilical venous endothelial cells either subjected to oscillatory shear stress or treated with the PKA (protein kinase A)/CREB (cAMP response element-binding protein) inhibitor H89 (N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide·2HCl hydrate). Further studies using the partial carotid artery ligation model showed that disturbed flow in the left carotid artery of Sema7A+/+ApoE-/- mice promoted the expression of endothelial Sema7A and cell adhesion molecules, leukocyte adhesion, and plaque formation, whereas such changes were attenuated in Sema7A-/-ApoE-/- mice. Further studies showed that blockage of ß1 integrin, a known Sema7A receptor, or inhibition of FAK (focal adhesion kinase), MEK1/2 (mitogen-activated protein kinase kinase 1/2), or NF-κB (nuclear factor-κB) significantly reduced the expression of cell adhesion molecules and THP-1 (human acute monocytic leukemia cell line) monocyte adhesion in Sema7A-overexpressing human umbilical venous endothelial cells. Studies using chimeric mice suggest that vascular, most likely endothelial, Sema7A plays a major role in atherogenesis. CONCLUSIONS: Our findings indicate a significant role of Sema7A in atherosclerosis by mediating endothelial dysfunction in a ß1 integrin-dependent manner.


Subject(s)
Antigens, CD/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Carotid Artery Diseases/metabolism , Endothelial Cells/metabolism , Integrin beta1/metabolism , Mechanotransduction, Cellular , Semaphorins/metabolism , Animals , Antigens, CD/genetics , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Cell Adhesion , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Endothelial Cells/pathology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Leukocyte Rolling , MAP Kinase Kinase Kinases/metabolism , Mice, Inbred C57BL , Mice, Knockout, ApoE , NF-kappa B/metabolism , Plaque, Atherosclerotic , Regional Blood Flow , Semaphorins/deficiency , Semaphorins/genetics , THP-1 Cells , Up-Regulation
6.
Am J Hum Genet ; 96(4): 581-96, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25839327

ABSTRACT

Innervation of the gut is segmentally lost in Hirschsprung disease (HSCR), a consequence of cell-autonomous and non-autonomous defects in enteric neuronal cell differentiation, proliferation, migration, or survival. Rare, high-penetrance coding variants and common, low-penetrance non-coding variants in 13 genes are known to underlie HSCR risk, with the most frequent variants in the ret proto-oncogene (RET). We used a genome-wide association (220 trios) and replication (429 trios) study to reveal a second non-coding variant distal to RET and a non-coding allele on chromosome 7 within the class 3 Semaphorin gene cluster. Analysis in Ret wild-type and Ret-null mice demonstrates specific expression of Sema3a, Sema3c, and Sema3d in the enteric nervous system (ENS). In zebrafish embryos, sema3 knockdowns show reduction of migratory ENS precursors with complete ablation under conjoint ret loss of function. Seven candidate receptors of Sema3 proteins are also expressed within the mouse ENS and their expression is also lost in the ENS of Ret-null embryos. Sequencing of SEMA3A, SEMA3C, and SEMA3D in 254 HSCR-affected subjects followed by in silico protein structure modeling and functional analyses identified five disease-associated alleles with loss-of-function defects in semaphorin dimerization and binding to their cognate neuropilin and plexin receptors. Thus, semaphorin 3C/3D signaling is an evolutionarily conserved regulator of ENS development whose dys-regulation is a cause of enteric aganglionosis.


Subject(s)
Epistasis, Genetic/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation , Hirschsprung Disease/genetics , Proto-Oncogene Proteins c-ret/genetics , Semaphorins/genetics , Animals , Base Sequence , Genome-Wide Association Study , Mice , Molecular Sequence Data , Semaphorins/deficiency , Semaphorins/metabolism , Sequence Analysis, DNA
7.
Cereb Cortex ; 27(11): 5054-5069, 2017 11 01.
Article in English | MEDLINE | ID: mdl-27655933

ABSTRACT

The thalamus is a central brain structure with topographically ordered long-range axonal projections that convey sensory information to the cortex via distinct nuclei. Although there is an increasing knowledge about genes important for thalamocortical (TC) development, the identification of genetic landmarks of the distinct thalamic nuclei during the embryonic development has not been addressed systematically. Indeed, a more comprehensive understanding of how the axons from the individual nuclei find their way and connect to their corresponding cortical area is called for. Here, we used a genetic dual labeling strategy in mice to purify distinct principal sensory thalamic neurons. Subsequent genome-wide transcriptome profiling revealed genes specifically expressed in each nucleus during embryonic development. Analysis of regulatory regions of the identified genes revealed key transcription factors and networks that likely underlie the specification of individual sensory-modality TC connections. Finally, the importance of correct axon targeting for the specific sensory-modality population transcriptome was evidenced in a Sema6A mutant, in which visual TC axons are derailed at embryonic life. In sum, our data determined the developmental transcriptional profile of the TC neurons that will eventually support sensory processing.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/embryology , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Thalamic Nuclei/cytology , Thalamic Nuclei/embryology , Animals , Axons/metabolism , Cerebral Cortex/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Mice, Transgenic , Mutation , Neural Pathways/cytology , Neural Pathways/embryology , Neural Pathways/metabolism , Semaphorins/deficiency , Semaphorins/genetics , Thalamic Nuclei/metabolism , Transcriptome
8.
Nature ; 470(7333): 259-63, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21270798

ABSTRACT

In the vertebrate retina, establishment of precise synaptic connections among distinct retinal neuron cell types is critical for processing visual information and for accurate visual perception. Retinal ganglion cells (RGCs), amacrine cells and bipolar cells establish stereotypic neurite arborization patterns to form functional neural circuits in the inner plexiform layer (IPL), a laminar region that is conventionally divided into five major parallel sublaminae. However, the molecular mechanisms governing distinct retinal subtype targeting to specific sublaminae within the IPL remain to be elucidated. Here we show that the transmembrane semaphorin Sema6A signals through its receptor PlexinA4 (PlexA4) to control lamina-specific neuronal stratification in the mouse retina. Expression analyses demonstrate that Sema6A and PlexA4 proteins are expressed in a complementary fashion in the developing retina: Sema6A in most ON sublaminae and PlexA4 in OFF sublaminae of the IPL. Mice with null mutations in PlexA4 or Sema6A exhibit severe defects in stereotypic lamina-specific neurite arborization of tyrosine hydroxylase (TH)-expressing dopaminergic amacrine cells, intrinsically photosensitive RGCs (ipRGCs) and calbindin-positive cells in the IPL. Sema6A and PlexA4 genetically interact in vivo for the regulation of dopaminergic amacrine cell laminar targeting. Therefore, neuronal targeting to subdivisions of the IPL in the mammalian retina is directed by repulsive transmembrane guidance cues present on neuronal processes.


Subject(s)
Cell Membrane/metabolism , Neurons/cytology , Neurons/metabolism , Retina/cytology , Retina/metabolism , Semaphorins/metabolism , Signal Transduction , Amacrine Cells/enzymology , Amacrine Cells/metabolism , Animals , Calbindins , Dopamine/metabolism , Gene Expression Profiling , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nerve Tissue Proteins , Neurites/metabolism , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Retina/embryology , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , S100 Calcium Binding Protein G/metabolism , Semaphorins/deficiency , Semaphorins/genetics , Tyrosine 3-Monooxygenase/metabolism
9.
J Virol ; 89(23): 11834-44, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26378166

ABSTRACT

UNLABELLED: Hantaan virus (HTNV) infection can cause a severe lethal hemorrhagic fever with renal syndrome (HFRS) in humans. CD8(+) T cells play a critical role in combating HTNV infections. However, the contributions of different CD8(+) T cell subsets to the immune response against viral infection are poorly understood. Here, we identified a novel subset of CD8(+) T cells characterized by the CD8(low) CD100(-) phenotype in HFRS patients. The CD8(low) CD100(-) subset accounted for a median of 14.3% of the total CD8(+) T cells in early phase of HFRS, and this percentage subsequently declined in the late phase of infection, whereas this subset was absent in healthy controls. Furthermore, the CD8(low) CD100(-) cells were associated with high activation and expressed high levels of cytolytic effector molecules and exhibited a distinct expression profile of effector CD8(+) T cells (CCR7(+/-) CD45RA(-) CD127(high) CD27(int) CD28(low) CD62L(-)). When stimulated with specific HTNV nucleocapsid protein-derived peptide pools, most responding CD8(+) cells (gamma interferon [IFN-γ] positive and/or tumor necrosis factor alpha [TNF-α] positive) were CD8(low) CD100(-) cells. The frequency of CD8(low) CD100(-) cells among HTNV-specific CD8(+) T cells was higher in milder cases than in more severe cases. Importantly, the proportion of the CD8(low) CD100(-) subset among CD8(+) T cells in early phase of HFRS was negatively correlated with the HTNV viral load, suggesting that CD8(low) CD100(-) cells may be associated with viral clearance. The contraction of the CD8(low) CD100(-) subset in late phase of infection may be related to the consistently high expression levels of PD-1. These results may provide new insights into our understanding of CD8(+) T cell-mediated protective immunity as well as immune homeostasis after HTNV infection in humans. IMPORTANCE: CD8(+) T cells play important roles in the antiviral immune response. We found that the proportion of CD8(low) CD100(-) cells among CD8(+) T cells from HFRS patients was negatively correlated with the HTNV viral load, and the frequency of CD8(low) CD100(-) cells among virus-specific CD8(+) T cells was higher in milder HFRS cases than in more severe cases. These results imply a beneficial role for the CD8(low) CD100(-) cell subset in viral control during human HTNV infection.


Subject(s)
Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Hantaan virus/immunology , Hemorrhagic Fever with Renal Syndrome/immunology , Semaphorins/immunology , T-Lymphocyte Subsets/immunology , Antibodies, Monoclonal , CD8-Positive T-Lymphocytes/virology , China , Flow Cytometry , Fluorescein-5-isothiocyanate , Humans , Real-Time Polymerase Chain Reaction , Semaphorins/deficiency , Statistics, Nonparametric , T-Lymphocyte Subsets/metabolism
10.
EMBO J ; 29(15): 2635-45, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20606624

ABSTRACT

The correct navigation of axons to their targets depends on guidance molecules in the extra-cellular environment. Differential responsiveness to a particular guidance cue is largely an outcome of disparity in the expression of its receptors on the reacting axons. Here, we show that the differential responsiveness of sympathetic and sensory neurons to the transmembrane Semaphorin Sema6A is mainly determined by its co-expression in the responding neurons. Both sympathetic and sensory neurons express the Sema6A receptor Plexin-A4, but only sympathetic neurons respond to it. The expression of Sema6A counteracts this responsiveness and is detected only in sensory neurons. Remarkably, sensory neurons that lack Sema6A gain sensitivity to it in a Plexin-A4-dependent manner. Using heterologus systems, we show that the co-expression of Sema6A and Plexin-A4 hinders the binding of exogenous ligand, suggesting that a Sema6A-Plexin-A4 cis interaction serves as an inhibitory mechanism. Finally, we provide evidence for differential modes of interaction in cis versus in trans. Thus, co-expression of a transmembrane cue together with its receptor can serve as a guidance response modulator.


Subject(s)
Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , Animals , Cells, Cultured , Chlorocebus aethiops , Humans , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Protein Binding , Receptors, Cell Surface/deficiency , Semaphorins/deficiency
11.
Blood ; 119(3): 745-55, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-22134167

ABSTRACT

Sema4D, also known as CD100, is a constitutively expressed immune semaphorin on T cells and NK cells. CD100 has important immune regulatory functions that improve antigen-specific priming by antigen-presenting cells, and can also act as a costimulatory molecule on T cells. We investigated the consequence of HIV-1 infection on CD100 expression by T cells, and whether CD100 expression signifies functionally competent effector cells. CD100 expression on T cells from healthy individuals was compared with HIV-1-infected subjects including elite controllers, noncontrollers, and patients receiving antiretroviral therapy. The frequency and fluorescence intensity of CD100 on CD8(+) and CD4(+) T cells were decreased during HIV-1 infection. Furthermore, the absolute number of CD100-expressing CD8(+) T cells was positively associated with the magnitude of HIV-1-specific T-cell responses. CD8(+) T cells lacking CD100 expression were functionally impaired and present in increased numbers in HIV-1-infected individuals. The number of CD100(-)CD8(+) T cells positively correlated with T-cell immunosenescence, immune activation, and viral load. Loss of CD100 expression appears to result from direct antigen stimulation, as in vitro cytokine exposure and viral replication did not significantly impact CD100 expression. These data suggest that loss of CD100 expression probably plays an important role in dysfunctional immunity in HIV-1 infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Semaphorins/deficiency , Antigen-Presenting Cells , Antigens, CD , Antiretroviral Therapy, Highly Active , CD8-Positive T-Lymphocytes/metabolism , Case-Control Studies , Cross-Sectional Studies , Cytokines/metabolism , Flow Cytometry , HIV Infections/drug therapy , Humans , Lymphocyte Activation , Viral Load , Virus Replication
12.
Blood ; 120(19): 4104-15, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-23007403

ABSTRACT

Formation of new vessels during development and in the mature mammal generally proceeds through angiogenesis. Although a variety of molecules and signaling pathways are known to underlie endothelial cell sprouting and remodeling during angiogenesis, many aspects of this complex process remain unexplained. Here we show that the transmembrane semaphorin6A (Sema6A) is expressed in endothelial cells, and regulates endothelial cell survival and growth by modulating the expression and signaling of VEGFR2, which is known to maintain endothelial cell viability by autocrine VEGFR signaling. The silencing of Sema6A in primary endothelial cells promotes cell death that is not rescued by exogenous VEGF-A or FGF2, attributable to the loss of prosurvival signaling from endogenous VEGF. Analyses of mouse tissues demonstrate that Sema6A is expressed in angiogenic and remodeling vessels. Mice with null mutations of Sema6A exhibit significant defects in hyaloid vessels complexity associated with increased endothelial cell death, and in retinal vessels development that is abnormally reduced. Adult Sema6A-null mice exhibit reduced tumor, matrigel, and choroidal angiogenesis compared with controls. Sema6A plays important roles in development of the nervous system. Here we show that it also regulates vascular development and adult angiogenesis.


Subject(s)
Neovascularization, Physiologic/genetics , Semaphorins/genetics , Semaphorins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Choroid/blood supply , Fibroblast Growth Factor 2/metabolism , Gene Silencing , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Knockout , Retina/metabolism , Retina/pathology , Retinal Vessels/metabolism , Retinal Vessels/pathology , Semaphorins/deficiency , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
13.
J Immunol ; 188(10): 4858-65, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22491253

ABSTRACT

Multiple sclerosis (MS) is a demyelinating autoimmune disease of the CNS and a leading cause of lasting neurologic disabilities in young adults. Although the precise mechanism remains incompletely understood, Ag presentation and subsequent myelin-reactive CD4(+) T cell activation/differentiation are essential for the pathogenesis of MS. Although semaphorins were initially identified as axon guidance cues during neural development, several semaphorins are crucially involved in various phases of immune responses. Sema4A is one of the membrane-type class IV semaphorins, which we originally identified from the cDNA library of dendritic cell (DC). Sema4A plays critical roles in T cell activation and Th1 differentiation during the course of experimental autoimmune encephalomyelitis, an animal model of MS; however, its pathological involvement in human MS has not been determined. In this study, we report that Sema4A is increased in the sera of patients with MS. The expression of Sema4A is increased on DCs in MS patients and shed from these cells in a metalloproteinase-dependent manner. DC-derived Sema4A is not only critical for Th1 but also for Th17 cell differentiation, and MS patients with high Sema4A levels exhibit Th17 skewing. Furthermore, patients with high Sema4A levels have more severe disabilities and are unresponsive to IFN-ß treatment. Taken together, our results suggest that Sema4A is involved in the pathogenesis of MS by promoting Th17 skewing.


Subject(s)
Cell Differentiation/immunology , Interferon-beta/therapeutic use , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Semaphorins/biosynthesis , Th17 Cells/immunology , Up-Regulation/immunology , Amino Acid Sequence , Animals , Cell Differentiation/genetics , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Multiple Sclerosis/pathology , Rats , Semaphorins/blood , Semaphorins/deficiency , Semaphorins/metabolism , Th17 Cells/metabolism , Th17 Cells/pathology , Up-Regulation/genetics
14.
Hum Mol Genet ; 20(24): 4759-74, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21903667

ABSTRACT

Reproduction in mammals is dependent on the function of specific neurons that secrete gonadotropin-releasing hormone-1 (GnRH-1). These neurons originate prenatally in the nasal placode and migrate into the forebrain along the olfactory-vomeronasal nerves. Alterations in this migratory process lead to defective GnRH-1 secretion, resulting in heterogeneous genetic disorders such as idiopathic hypogonadotropic hypogonadism (IHH), and other reproductive diseases characterized by the reduction or failure of sexual competence. Combining mouse genetics with in vitro models, we demonstrate that Semaphorin 7A (Sema7A) is essential for the development of the GnRH-1 neuronal system. Loss of Sema7A signaling alters the migration of GnRH-1 neurons, resulting in significantly reduced numbers of these neurons in the adult brain as well as in reduced gonadal size and subfertility. We also show that GnRH-1 cells differentially express the Sema7 receptors ß1-integrin and Plexin C1 as a function of their migratory stage, whereas the ligand is robustly expressed along developing olfactory/vomeronasal fibers. Disruption of Sema7A function in vitro inhibits ß1-integrin-mediated migration. Analysis of Plexin C1(-/-) mice did not reveal any difference in the migratory process of GnRH-1 neurons, indicating that Sema7A mainly signals through ß1-integrin to regulate GnRH-1 cell motility. In conclusion, we have identified Sema7A as a gene implicated in the normal development of the GnRH-1 system in mice and as a genetic marker for the elucidation of some forms of GnRH-1 deficiency in humans.


Subject(s)
Antigens, CD/metabolism , Cell Movement , Fertility , Gonadotropin-Releasing Hormone/metabolism , Gonads/embryology , Integrin beta1/metabolism , Protein Precursors/metabolism , Semaphorins/metabolism , Signal Transduction , Animals , Axons/metabolism , Brain/embryology , Brain/pathology , Cell Count , Gonads/abnormalities , Gonads/metabolism , Gonads/pathology , Humans , Male , Mice , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Olfactory Bulb/embryology , Olfactory Bulb/metabolism , Receptors, Cell Surface/metabolism , Semaphorins/deficiency , Testis/embryology , Testis/metabolism , Testis/pathology , Vomeronasal Organ/embryology , Vomeronasal Organ/metabolism
15.
J Clin Immunol ; 33(1): 200-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23007237

ABSTRACT

PURPOSE: The class IV semaphorin Sema4A is critical for efficient Th1 differentiation and Sema4a (-/-) mice exhibit impaired Th1 immune responses. However, the role of Sema4A in Th2 cell-mediated allergic diseases has not been fully studied. The aim of this study was to clarify the regulatory role possessed by Sema4A in mouse models of allergic diseases, particularly allergic asthma. METHODS: Sema4a (-/-) mice on a BALB/c background were examined for the development of allergic diseases. To induce experimental asthma, mice were sensitized with ovalbumin (OVA) followed by intranasal challenges with OVA. After challenge, airway hyperreactivity (AHR) and airway inflammation were evaluated. The role of Sema4A in asthma was examined using Sema4a (-/-) mice and Sema4A-Fc fusion proteins. The direct effects of Sema4A-Fc on antigen-specific effector CD4(+) T cells were also examined. RESULTS: A fraction of Sema4a (-/-) BALB/c mice spontaneously developed skin lesions that resembled atopic dermatitis (AD) in humans. Furthermore, AHR, airway inflammation, and Th2-type immune responses were enhanced in Sema4a (-/-) mice compared to wild type (WT) mice when immunized and challenged with OVA. In vivo systemic administration of Sema4A-Fc during the challenge period ameliorated AHR and lung inflammation and reduced the production of Th2-type cytokines in WT mice. The inhibitory effects of Sema4A on airway inflammation were also observed in mice deficient in Tim-2, a Sema4A receptor. Finally, we showed that Sema4A-Fc directly inhibited IL-4-producing OVA-specific CD4(+) T cells. CONCLUSION: These results demonstrate that Sema4A plays an inhibitory role in Th2-type allergic diseases, such as allergic asthma.


Subject(s)
Allergens/immunology , Asthma/immunology , Epitopes/immunology , Semaphorins/physiology , Allergens/administration & dosage , Animals , Asthma/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Cells, Cultured , Dermatitis, Atopic/immunology , Disease Models, Animal , Female , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Semaphorins/deficiency
16.
J Immunol ; 186(5): 2881-8, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21270411

ABSTRACT

Basophils are strong mediators of Th2 responses during helminthic infections. Recently, basophils were shown to function as APCs and promote both Th2 skewing and humoral memory responses. However, the mechanisms that regulate basophils are still unclear. In this article, we show that a class IV semaphorin, Sema4B, negatively regulates basophil functions through T cell-basophil contacts. In a screen to identify semaphorins that function in the immune system, we determined that Sema4B is expressed in T and B cells. Interestingly, Sema4B(-/-) mice had considerably increased serum IgE levels despite normal lymphocyte and dendritic cell functions. Recombinant Sema4B significantly inhibited IL-4 and IL-6 production from basophils in response to various stimuli, including IL-3, papain, and FcεRI cross-linking. In addition, T cell-derived Sema4B, which accumulated at contact sites between basophils and CD4(+) T cells, suppressed basophil-mediated Th2 skewing, suggesting that Sema4B regulates basophil responses through cognate cell-cell contacts. Furthermore, Sema4B(-/-) mice had enhanced basophil-mediated memory IgE production, which was abolished by treating with an anti-FcεRIα Ab. Collectively, these results indicate that Sema4B negatively regulates basophil-mediated Th2 and humoral memory responses.


Subject(s)
Basophils/immunology , Basophils/metabolism , Down-Regulation/immunology , Immune Tolerance , Semaphorins/physiology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Down-Regulation/genetics , Gene Expression Regulation/immunology , Immune Tolerance/genetics , Immunoglobulin E/biosynthesis , Immunoglobulin E/blood , Immunologic Memory/genetics , Interleukin-4/antagonists & inhibitors , Interleukin-4/biosynthesis , Interleukin-6/antagonists & inhibitors , Interleukin-6/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Mice, Transgenic , Semaphorins/deficiency , Semaphorins/genetics , Semaphorins/isolation & purification , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Th2 Cells/cytology , Th2 Cells/immunology
17.
Nature ; 446(7136): 680-4, 2007 Apr 05.
Article in English | MEDLINE | ID: mdl-17377534

ABSTRACT

Semaphorins are axon guidance factors that assist growing axons in finding appropriate targets and forming synapses. Emerging evidence suggests that semaphorins are involved not only in embryonic development but also in immune responses. Semaphorin 7A (Sema7A; also known as CD108), which is a glycosylphosphatidylinositol-anchored semaphorin, promotes axon outgrowth through beta1-integrin receptors and contributes to the formation of the lateral olfactory tract. Although Sema7A has been shown to stimulate human monocytes, its function as a negative regulator of T-cell responses has also been reported. Thus, the precise function of Sema7A in the immune system remains unclear. Here we show that Sema7A, which is expressed on activated T cells, stimulates cytokine production in monocytes and macrophages through alpha1beta1 integrin (also known as very late antigen-1) as a component of the immunological synapse, and is critical for the effector phase of the inflammatory immune response. Sema7A-deficient (Sema7a-/-) mice are defective in cell-mediated immune responses such as contact hypersensitivity and experimental autoimmune encephalomyelitis. Although antigen-specific and cytokine-producing effector T cells can develop and migrate into antigen-challenged sites in Sema7a-/- mice, Sema7a-/- T cells fail to induce contact hypersensitivity even when directly injected into the antigen-challenged sites. Thus, the interaction between Sema7A and alpha1beta1 integrin is crucial at the site of inflammation. These findings not only identify a function of Sema7A as an effector molecule in T-cell-mediated inflammation, but also reveal a mechanism of integrin-mediated immune regulation.


Subject(s)
Antigens, CD/metabolism , Inflammation/immunology , Integrin alpha1beta1/metabolism , Semaphorins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigens, CD/genetics , Cytokines/metabolism , Immunity/immunology , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/metabolism , Semaphorins/deficiency , Semaphorins/genetics , Signal Transduction
18.
Glia ; 60(10): 1590-604, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22777942

ABSTRACT

Myelination is regulated by extracellular proteins, which control interactions between oligodendrocytes and axons. Semaphorins are repulsive axon guidance molecules, which control the migration of oligodendrocyte precursors during normal development and possibly in demyelinating diseases. We show here that the transmembrane semaphorin 6A (Sema6A) is highly expressed by myelinating oligodendrocytes in the postnatal mouse brain. In adult mice, Sema6A expression is upregulated in demyelinating lesions in cuprizone-treated mice. The analysis of the optic nerve and anterior commissure of Sema6A-deficient mice revealed a marked delay of oligodendrocyte differentiation. Accordingly, the development of the nodes of Ranvier is also transiently delayed. We also observed an arrest in the in vitro differentiation of purified oligodendrocytes lacking Sema6A, with a reduction of the expression level of Myelin Basic Protein. Their morphology is also abnormal, with less complex and ramified processes than wild-type oligodendrocytes. In myelinating co-cultures of dorsal root ganglion neurons and purified oligodendrocytes we found that myelination is perturbed in absence of Sema6A. These results suggest that Sema6A might have a role in myelination by controlling oligodendrocyte differentiation.


Subject(s)
Cell Differentiation/physiology , Demyelinating Diseases/pathology , Gene Expression Regulation, Developmental/genetics , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Semaphorins/metabolism , Age Factors , Animals , Animals, Newborn , Antigens, Differentiation/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/cytology , Bromodeoxyuridine/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Coculture Techniques , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/physiopathology , Disease Models, Animal , Embryo, Mammalian , Female , Ganglia, Spinal/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monoamine Oxidase Inhibitors/toxicity , Mutation/physiology , Myelin Basic Protein/metabolism , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Neurons/physiology , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/drug effects , Pregnancy , RNA, Messenger/metabolism , Ranvier's Nodes/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Cell Surface/deficiency , Semaphorins/deficiency , Stem Cells/physiology , Time Factors , Transcription Factors/metabolism
19.
Arterioscler Thromb Vasc Biol ; 31(1): 151-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20947821

ABSTRACT

OBJECTIVE: To characterize the role of a vascular-expressed class 3 semaphorin (semaphorin 3G [Sema3G]). METHODS AND RESULTS: Semaphorins have been identified as axon guidance molecules. Yet, they have more recently also been characterized as attractive and repulsive regulators of angiogenesis. Through a transcriptomic screen, we identified Sema3G as a molecule of angiogenic endothelial cells. Sema3G-deficient mice are viable and exhibit no overt vascular phenotype. Yet, LacZ expression in the Sema3G locus revealed intense arterial vascular staining in the angiogenic vasculature, starting at E9.5, which was detectable throughout adolescence and downregulated in adult vasculature. Sema3G is expressed as a full-length 100-kDa secreted molecule that is processed by furin proteases to yield 95- and a 65-kDa Sema domain-containing subunits. Full-length Sema3G binds to NP2, whereas processed Sema3G binds to NP1 and NP2. Expression profiling and cellular experiments identified autocrine effects of Sema3G on endothelial cells and paracrine effects on smooth muscle cells. CONCLUSIONS: Although the mouse knockout phenotype suggests compensatory mechanisms, the experiments identify Sema3G as a primarily endothelial cell-expressed class 3 semaphorin that controls endothelial and smooth muscle cell functions in autocrine and paracrine manners, respectively.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Gene Expression Profiling , Semaphorins/metabolism , Animals , Autocrine Communication , C-Reactive Protein/metabolism , Cells, Cultured , Coculture Techniques , Endothelium, Vascular/embryology , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Genotype , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/embryology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Paracrine Communication , Phenotype , Protein Binding , Protein Processing, Post-Translational , RNA Interference , Recombinant Proteins/metabolism , Semaphorins/deficiency , Semaphorins/genetics , Transfection , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
20.
J Immunol ; 184(3): 1499-506, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20038643

ABSTRACT

Although semaphorins were originally identified as axonal guidance molecules during neuronal development, it is emerging that several semaphorins play crucial roles in various phases of immune responses. Sema4D/CD100, a class IV semaphorin, has been shown to be involved in the nervous and immune systems through its receptors plexin-B1 and CD72, respectively. However, the involvement of Sema4D in neuroinflammation still remains unclear. We found that Sema4D promoted inducible NO synthase expression by primary mouse microglia, the effects of which were abolished in plexin-B1-deficient but not in CD72-deficient microglia. In addition, during the development of experimental autoimmune encephalomyelitis (EAE), which was induced by immunization with myelin oligodendrocyte glycoprotein-derived peptides, we observed that the expression of Sema4D and plexin-B1 was induced in infiltrating mononuclear cells and microglia, respectively. Consistent with these expression profiles, when myelin oligodendrocyte glycoprotein-specific T cells derived from wild-type mice were adoptively transferred into plexin-B1-deficient mice or bone marrow chimera mice with plexin-B1-deficient CNS resident cells, the development of EAE was considerably attenuated. Furthermore, blocking Abs against Sema4D significantly inhibited neuroinflammation during EAE development. Collectively, our findings demonstrate the role of Sema4D-plexin-B1 interactions in the activation of microglia and provide their pathologic significance in neuroinflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Microglia/immunology , Microglia/metabolism , Nerve Tissue Proteins/physiology , Receptors, Cell Surface/physiology , Semaphorins/physiology , Amino Acid Sequence , Animals , Bone Marrow Cells/enzymology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/enzymology , Microglia/pathology , Molecular Sequence Data , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Radiation Chimera/immunology , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/metabolism , Semaphorins/deficiency , Semaphorins/metabolism , Up-Regulation/genetics , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL