Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Development ; 149(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35112129

ABSTRACT

The tracheal epithelium is a primary target for pulmonary diseases as it provides a conduit for air flow between the environment and the lung lobes. The cellular and molecular mechanisms underlying airway epithelial cell proliferation and differentiation remain poorly understood. Hedgehog (HH) signaling orchestrates communication between epithelial and mesenchymal cells in the lung, where it modulates stromal cell proliferation, differentiation and signaling back to the epithelium. Here, we reveal a previously unreported autocrine function of HH signaling in airway epithelial cells. Epithelial cell depletion of the ligand sonic hedgehog (SHH) or its effector smoothened (SMO) causes defects in both epithelial cell proliferation and differentiation. In cultured primary human airway epithelial cells, HH signaling inhibition also hampers cell proliferation and differentiation. Epithelial HH function is mediated, at least in part, through transcriptional activation, as HH signaling inhibition leads to downregulation of cell type-specific transcription factor genes in both the mouse trachea and human airway epithelial cells. These results provide new insights into the role of HH signaling in epithelial cell proliferation and differentiation during airway development.


Subject(s)
Autocrine Communication/physiology , Cell Differentiation , Cell Proliferation , Hedgehog Proteins/metabolism , Signal Transduction/genetics , Animals , Cells, Cultured , Down-Regulation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Humans , Lung/growth & development , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Smoothened Receptor/deficiency , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Trachea/cytology , Trachea/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Mol Genet Genomics ; 296(1): 33-40, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32944789

ABSTRACT

Joubert syndrome (JBTS), a rare genetic disorder resulted from primary cilium defects or basal-body dysfunction, is characterized by agenesis of cerebellar vermis and abnormal brain stem. Both genotypes and phenotypes of JBTS are highly heterogeneous. The identification of pathogenic gene variation is essential for making a definite diagnosis on JBTS. Here, we found that hypoplasia of cerebellar vermis occurred in three male members in a Chinese family. Then, we performed whole exome sequencing to identify a novel missense mutation c.599T > C (p. L200P) in the OFD1 gene which is the candidate gene of X-linked JBTS (JBST10). The following analysis showed that the variant was absent in the 1000 Genomes, ExAC and the 200 female controls; the position 200 Leucine residue was highly conserved across species; the missense variant was predicted to be deleterious using PolyPhen-2, PROVEAN, SIFT and Mutation Taster. The OFD1 expression was heavily lower in the proband and an induced male fetus compared with a healthy male with a wild-type OFD1 gene. The in vitro expression analysis of transiently transfecting c.599T or c.599C plasmids into HEK-293T cells confirmed that the missense mutation caused OFD1 reduction at the protein level. And further the mutated OFD1 decreased the level of Gli1 protein, a read-out of Sonic hedgehog (SHH) signaling essential for development of central neural system. A known pathogenic variant c.515T > C (p. L172P) showed the similar results. All of these observations suggested that the missense mutation causes the loss function of OFD1, resulting in SHH signaling impairs and brain development abnormality. In addition, the three patients have Dandy-Walker malformation, macrogyria and tetralogy of Fallot, respectively, the latter two of which are firstly found in JBTS10 patients. In conclusion, our findings expand the context of genotype and phenotype in the JBTS10 patients.


Subject(s)
Abnormalities, Multiple/genetics , Cerebellum/abnormalities , Dandy-Walker Syndrome/genetics , Eye Abnormalities/genetics , Kidney Diseases, Cystic/genetics , Lissencephaly/genetics , Mutation, Missense , Proteins/genetics , Retina/abnormalities , Tetralogy of Fallot/genetics , Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Amino Acid Sequence , Brain Stem/abnormalities , Brain Stem/diagnostic imaging , Brain Stem/metabolism , Cerebellar Vermis/abnormalities , Cerebellar Vermis/diagnostic imaging , Cerebellar Vermis/metabolism , Cerebellum/diagnostic imaging , Cerebellum/metabolism , Cerebellum/pathology , Child, Preschool , Dandy-Walker Syndrome/diagnostic imaging , Dandy-Walker Syndrome/metabolism , Dandy-Walker Syndrome/pathology , Eye Abnormalities/diagnostic imaging , Eye Abnormalities/metabolism , Eye Abnormalities/pathology , Family , Female , Gene Expression , Genotype , HEK293 Cells , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Humans , Kidney Diseases, Cystic/diagnostic imaging , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/pathology , Lissencephaly/diagnostic imaging , Lissencephaly/metabolism , Lissencephaly/pathology , Male , Pedigree , Phenotype , Proteins/metabolism , Retina/diagnostic imaging , Retina/metabolism , Retina/pathology , Sequence Alignment , Sequence Homology, Amino Acid , Sex Factors , Signal Transduction , Tetralogy of Fallot/diagnostic imaging , Tetralogy of Fallot/metabolism , Tetralogy of Fallot/pathology , Zinc Finger Protein GLI1/deficiency , Zinc Finger Protein GLI1/genetics
3.
PLoS Biol ; 18(11): e3000946, 2020 11.
Article in English | MEDLINE | ID: mdl-33253145

ABSTRACT

Inflammation of the central nervous system (CNS) induces endothelial blood-brain barrier (BBB) opening as well as the formation of a tight junction barrier between reactive astrocytes at the Glia Limitans. We hypothesized that the CNS parenchyma may acquire protection from the reactive astrocytic Glia Limitans not only during neuroinflammation but also when BBB integrity is compromised in the resting state. Previous studies found that astrocyte-derived Sonic hedgehog (SHH) stabilizes the BBB during CNS inflammatory disease, while endothelial-derived desert hedgehog (DHH) is expressed at the BBB under resting conditions. Here, we investigated the effects of endothelial Dhh on the integrity of the BBB and Glia Limitans. We first characterized DHH expression within endothelial cells at the BBB, then demonstrated that DHH is down-regulated during experimental autoimmune encephalomyelitis (EAE). Using a mouse model in which endothelial Dhh is inducibly deleted, we found that endothelial Dhh both opens the BBB via the modulation of forkhead box O1 (FoxO1) transcriptional activity and induces a tight junctional barrier at the Glia Limitans. We confirmed the relevance of this glial barrier system in human multiple sclerosis active lesions. These results provide evidence for the novel concept of "chronic neuroinflammatory tolerance" in which BBB opening in the resting state is sufficient to stimulate a protective barrier at the Glia Limitans that limits the severity of subsequent neuroinflammatory disease. In summary, genetic disruption of the BBB generates endothelial signals that drive the formation under resting conditions of a secondary barrier at the Glia Limitans with protective effects against subsequent CNS inflammation. The concept of a reciprocally regulated CNS double barrier system has implications for treatment strategies in both the acute and chronic phases of multiple sclerosis pathophysiology.


Subject(s)
Blood-Brain Barrier/physiology , Blood-Brain Barrier/physiopathology , Adherens Junctions/pathology , Adherens Junctions/physiology , Animals , Antigens, CD/genetics , Antigens, CD/physiology , Astrocytes/pathology , Astrocytes/physiology , Cadherins/genetics , Cadherins/physiology , Capillary Permeability/genetics , Capillary Permeability/physiology , Claudin-5/genetics , Claudin-5/physiology , Down-Regulation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Endothelial Cells/pathology , Endothelial Cells/physiology , Female , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Hedgehog Proteins/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Neuroglia/pathology , Neuroglia/physiology , Tight Junctions/pathology , Tight Junctions/physiology
4.
Commun Biol ; 3(1): 144, 2020 03 25.
Article in English | MEDLINE | ID: mdl-32214226

ABSTRACT

The Creeper (Cp) chicken is characterized by chondrodystrophy in Cp/+ heterozygotes and embryonic lethality in Cp/Cp homozygotes. However, the genes underlying the phenotypes have not been fully known. Here, we show that a 25 kb deletion on chromosome 7, which contains the Indian hedgehog (IHH) and non-homologous end-joining factor 1 (NHEJ1) genes, is responsible for the Cp trait in Japanese bantam chickens. IHH is essential for chondrocyte maturation and is downregulated in the Cp/+ embryos and completely lost in the Cp/Cp embryos. This indicates that chondrodystrophy is caused by the loss of IHH and that chondrocyte maturation is delayed in Cp/+ heterozygotes. The Cp/Cp homozygotes exhibit impaired DNA double-strand break (DSB) repair due to the loss of NHEJ1, resulting in DSB accumulation in the vascular and nervous systems, which leads to apoptosis and early embryonic death.


Subject(s)
Bone Diseases, Developmental/veterinary , Bone and Bones/embryology , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Gene Deletion , Hedgehog Proteins/genetics , Poultry Diseases/genetics , Animals , Apoptosis , Bone Diseases, Developmental/embryology , Bone Diseases, Developmental/genetics , Bone Diseases, Developmental/metabolism , Bone and Bones/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chick Embryo , DNA Repair Enzymes/deficiency , DNA-Binding Proteins/deficiency , Embryonic Development , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Hedgehog Proteins/deficiency , Heterozygote , Homozygote , Phenotype , Poultry Diseases/embryology , Poultry Diseases/metabolism
5.
Toxicol Lett ; 320: 1-8, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-31756458

ABSTRACT

With the spread of hexavalent chromium [Cr(VI)] contamination, risk of exposure in non-occupational populations is increasing. The liver is the main target organ for Cr(VI) accumulation; however, the effect of long-term Cr(VI) exposure on liver toxicity is largely unknown. In this study, we investigated the effect of chronic Cr(VI) exposure on liver fibrosis and its possible mechanism. Mice were injected with Cr(VI) for two months, and our results showed Cr(VI) treatment caused liver toxicity characterized by liver structure disorganization, liver dysfunction, and antioxidant enzyme system inhibition. The development of liver fibrosis was also found via the emergence of collagen fibril deposition, increased expression of extracellular matrix-related genes, activation of hepatic stellate cells (HSCs) and increase the expression levels of Hedgehog (Hh) signaling pathway-related molecules. To demonstrate the role of Hh signaling in the regulation of Cr(VI)-induced liver fibrosis, genetically modified mice with heterozygous deficiency of Shh (Shh+/-) were used. In the Shh+/- mice, Hh signaling, HSCs activation and liver fibrosis development were all ameliorated. In conclusion, we demonstrated that Cr(VI)-induced liver fibrosis development resulted from Hh pathway-mediated HSCs activation. Our findings strongly suggest that inhibition of Hh pathway may help in the development of new strategies for Cr(VI)-associated liver fibrosis.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Chromium , Hedgehog Proteins/metabolism , Hepatic Stellate Cells/metabolism , Liver Cirrhosis, Experimental/metabolism , Liver/metabolism , Potassium Dichromate , Signal Transduction , Animals , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Hepatic Stellate Cells/pathology , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/pathology , Male , Mice, Inbred C57BL , Mice, Knockout
6.
Circ Res ; 123(9): 1053-1065, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30355159

ABSTRACT

RATIONALE: Klf (kruppel-like factor) 2 is critical to establish and maintain endothelial integrity. OBJECTIVE: Therefore, determining upstream and downstream mediators of Klf2 would lead to alternative therapeutic targets in cardiovascular disease management. METHODS AND RESULTS: Here we identify Dhh (desert hedgehog) as a downstream effector of Klf2, whose expression in endothelial cells (ECs) is upregulated by shear stress and decreased by inflammatory cytokines. Consequently, we show that Dhh knockdown in ECs promotes endothelial permeability and EC activation and that Dhh agonist prevents TNF-α (tumor necrosis factor alpha) or glucose-induced EC dysfunction. Moreover, we demonstrate that human critical limb ischemia, a pathological condition linked to diabetes mellitus and inflammation, is associated to major EC dysfunction. By recreating a complex model of critical limb ischemia in diabetic mice, we found that Dhh-signaling agonist significantly improved EC function without promoting angiogenesis, which subsequently improved muscle perfusion. CONCLUSION: Restoring EC function leads to significant critical limb ischemia recovery. Dhh appears to be a promising target, downstream of Klf2, to prevent the endothelial dysfunction involved in ischemic vascular diseases.


Subject(s)
Endothelial Cells/metabolism , Hedgehog Proteins/metabolism , Ischemia/metabolism , Kruppel-Like Transcription Factors/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Animals , Autocrine Communication , Capillary Permeability , Cells, Cultured , Critical Illness , Cyclohexylamines/pharmacology , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Gene Expression Regulation , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Hindlimb , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Ischemia/drug therapy , Ischemia/genetics , Ischemia/physiopathology , Kruppel-Like Transcription Factors/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/drug effects , Regional Blood Flow , Signal Transduction , Stress, Mechanical , Thiophenes/pharmacology
7.
FASEB J ; 32(10): 5703-5715, 2018 10.
Article in English | MEDLINE | ID: mdl-29768039

ABSTRACT

Sonic hedgehog ( Shh) is crucial for organogenesis in the foregut. This study investigated the function of Shh at the late-gestational stage; during which, the esophagus continues to differentiate. We established cytokeratin 14 ( CK14)-Cre;Shhfl/fl mice in which the down-regulation of Shh in the epithelium occurred at approximately the same time as esophageal muscle conversion. Hematoxylin and eosin and immunohistochemical staining, with antibodies against keratin 14, Shh, patched 1 (Ptch1), Gli1, proliferating cell nuclear antigen (PCNA), α-smooth muscle actin (αSMA), high-molecular-weight caldesmon (hCD), myogenin, paired box 7 (Pax7), ß3-tubulin, and protein gene product 9.5 (PGP9.5), was performed to detect specific tissue dysplasia. Organ culture was conducted in vitro, and total mRNA was extracted to determine the transcriptional dysregulation. The esophagus of CK14-Cre;Shhfl/fl mice developed into an independent tube with an obvious dilatation at postnatal d 0.5. The number of cell layers and the expression of PCNA were decreased in mutant mice, compared with those in wild-type mice. The expression of hCD declined progressively in the middle, distal, and lower esophageal sphincter levels of the mutant esophagus from embryonic d 17.5, compared with the expression in wild-type littermates. Pax7 accumulation and myogenin reduction in mutant mice indicated that esophageal skeletal-myoblast progression was blocked. RNA sequencing analysis revealed a significant down-regulation of genes involved in proliferation and muscular motivation in CK14-Cre;Shhfl/fl mice. Thus, loss of Shh at the late-gestational stage leads to megaesophagus with reduced proliferation and a muscle development disorder in mice.-Jia, X., Min, L., Zhu, S., Zhang, S., Huang, X. Loss of sonic hedgehog gene leads to muscle development disorder and megaesophagus in mice.


Subject(s)
Esophageal Achalasia , Hedgehog Proteins/deficiency , Muscle Proteins , Muscle, Skeletal , Muscular Diseases , Mutation , Animals , Animals, Genetically Modified , Esophageal Achalasia/embryology , Esophageal Achalasia/genetics , Esophageal Achalasia/metabolism , Esophageal Achalasia/pathology , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/epidemiology , Muscular Diseases/genetics , Muscular Diseases/metabolism , Muscular Diseases/pathology
8.
Endocrinology ; 159(7): 2563-2575, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29788357

ABSTRACT

Ovarian development requires coordinate communications among oocytes, granulosa cells, and theca cells. Two Hedgehog (Hh) pathway ligands, Desert hedgehog (Dhh) and Indian hedgehog (Ihh), are produced by the granulosa cells and work together to regulate theca cell specification and development. Mice lacking both Dhh and Ihh had loss of normal ovarian function, which raised the question of which biological actions are specifically controlled by each ligand during folliculogenesis. By comparing the reproductive fitness, hormonal profiles, and ovarian transcriptomes among control, Dhh single-knockout (KO), Ihh KO, and Dhh/Ihh double-knockout (DKO) mice, we examined the specific roles of Dhh and Ihh in these processes. Dhh/Ihh DKO female mice were infertile because of a lack of theca cells and their steroid product androgen. Although Dhh and Ihh KO mice were fertile with normal folliculogenesis, they had decreased androgen production and alterations in their ovarian transcriptomes. Absence of Ihh led to aberrant steroidogenesis and elevated inflammation responses, which were not found in Dhh KO mouse ovaries, implicating that IHH has a greater impact than DHH on the activation of the Hh signaling pathway in the ovary. Our findings provide insight into not only how the Hh pathway influences folliculogenesis but also the distinct and overlapping roles of Dhh and Ihh in supporting ovarian development.


Subject(s)
Hedgehog Proteins/deficiency , Hedgehog Proteins/metabolism , Animals , Female , Mice , Mice, Knockout , Ovary/metabolism , Reproduction/genetics , Reproduction/physiology , Signal Transduction/genetics , Signal Transduction/physiology
9.
PLoS One ; 13(5): e0197535, 2018.
Article in English | MEDLINE | ID: mdl-29771958

ABSTRACT

The elaborate anatomy of hands and feet is shaped by coordinated formation of digits and regression of the interdigital mesenchyme (IM). A failure of this process causes persistence of interdigital webbing and consequently cutaneous syndactyly. Bone morphogenetic proteins (BMPs) are key inductive factors for interdigital cell death (ICD) in vivo. NOGGIN (NOG) is a major BMP antagonist that can interfere with BMP-induced ICD when applied exogenously, but its in vivo role in this process is unknown. We investigated the physiological role of NOG in ICD and found that Noggin null mice display cutaneous syndactyly and impaired interdigital mesenchyme specification. Failure of webbing regression was caused by lack of cell cycle exit and interdigital apoptosis. Unexpectedly, Noggin null mutants also exhibit increased Indian hedgehog (Ihh) expression within cartilage condensations that leads to aberrant extension of IHH downstream signaling into the interdigital mesenchyme. A converse phenotype with increased apoptosis and reduced cell proliferation was found in the interdigital mesenchyme of Ihh mutant embryos. Our data point towards a novel role for NOG in balancing Ihh expression in the digits impinging on digit-interdigit cross talk. This suggests a so far unrecognized physiological role for IHH in interdigital webbing biology.


Subject(s)
Apoptosis/physiology , Bone Morphogenetic Proteins/physiology , Carrier Proteins/physiology , Hedgehog Proteins/physiology , Mesoderm/embryology , Signal Transduction/physiology , Syndactyly/physiopathology , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cartilage/embryology , Cell Cycle , Ectoderm/physiology , Gene Expression Regulation, Developmental , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Mesoderm/cytology , Mesoderm/pathology , Mice , Mice, Knockout , Signal Transduction/genetics , Specific Pathogen-Free Organisms , Syndactyly/embryology , Syndactyly/pathology , Toes/embryology
10.
Cardiovasc Res ; 114(5): 759-770, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29365079

ABSTRACT

Aims: Hedgehog (Hh) signalling has been shown to be re-activated in ischaemic tissues and participate in ischaemia-induced angiogenesis. Sonic Hedgehog (Shh) is upregulated by more than 80-fold in the ischaemic skeletal muscle, however its specific role in ischaemia-induced angiogenesis has not yet been fully investigated. The purpose of the present study was to investigate the role of endogenous Shh in ischaemia-induced angiogenesis. Methods and results: To this aim, we used inducible Shh knock-out (KO) mice and unexpectedly found that capillary density was significantly increased in re-generating muscle of Shh deficient mice 5 days after hind limb ischaemia was induced, demonstrating that endogenous Shh does not promote angiogenesis but more likely limits it. Myosin and MyoD expression were equivalent in Shh deficient mice and control mice, indicating that endogenous Shh is not required for ischaemia-induced myogenesis. Additionally, we observed a significant increase in macrophage infiltration in the ischaemic muscle of Shh deficient mice. Our data indicate that this was due to an increase in chemokine expression by myoblasts in the setting of impaired Hh signalling, using tissue specific Smoothened conditional KO mice. The increased macrophage infiltration in mice deficient for Hh signalling in myocytes was associated with increased VEGFA expression and a transiently increased angiogenesis, demonstrating that Shh limits inflammation and angiogenesis indirectly by signalling to myocytes. Conclusion: Although ectopic administration of Shh has previously been shown to promote ischaemia-induced angiogenesis, the present study reveals that endogenous Shh does not promote ischaemia-induced angiogenesis. On the contrary, the absence of Shh leads to aberrant ischaemic tissue inflammation and a transiently increased angiogenesis.


Subject(s)
Hedgehog Proteins/metabolism , Inflammation/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Neovascularization, Physiologic , Animals , Blood Flow Velocity , Chemokines/metabolism , Chemotaxis , Disease Models, Animal , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Hindlimb , Inflammation/genetics , Inflammation/physiopathology , Inflammation/prevention & control , Ischemia/genetics , Ischemia/physiopathology , Macrophages/metabolism , Mice, Knockout , Myoblasts, Skeletal/metabolism , Regional Blood Flow , Signal Transduction , Time Factors , Vascular Endothelial Growth Factor A/metabolism
11.
Nat Genet ; 49(10): 1539-1545, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28846100

ABSTRACT

Copy number variations (CNVs) often include noncoding sequences and putative enhancers, but how these rearrangements induce disease is poorly understood. Here we investigate CNVs involving the regulatory landscape of IHH (encoding Indian hedgehog), which cause multiple, highly localized phenotypes including craniosynostosis and synpolydactyly. We show through transgenic reporter and genome-editing studies in mice that Ihh is regulated by a constellation of at least nine enhancers with individual tissue specificities in the digit anlagen, growth plates, skull sutures and fingertips. Consecutive deletions, resulting in growth defects of the skull and long bones, showed that these enhancers function in an additive manner. Duplications, in contrast, caused not only dose-dependent upregulation but also misexpression of Ihh, leading to abnormal phalanges, fusion of sutures and syndactyly. Thus, precise spatiotemporal control of developmental gene expression is achieved by complex multipartite enhancer ensembles. Alterations in the composition of such clusters can result in gene misexpression and disease.


Subject(s)
Bone Diseases, Developmental/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Hedgehog Proteins/physiology , Osteogenesis/genetics , Animals , Base Sequence , DNA Copy Number Variations , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Foot Deformities, Congenital/genetics , Gene Deletion , Gene Dosage , Gene Duplication , Gene Knockout Techniques , Genes, Reporter , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Mice , Mice, Inbred C57BL , Polydactyly/genetics , Regulatory Sequences, Nucleic Acid , Sequence Analysis, DNA , Skull/abnormalities , Transcription, Genetic
12.
J Immunol ; 199(2): 501-509, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28576978

ABSTRACT

Allergic asthma is a significant health burden in western countries, and continues to increase in prevalence. Th2 cells contribute to the development of disease through release of the cytokines IL-4, IL-5, and IL-13, resulting in increased airway eosinophils and mucus hypersecretion. The molecular mechanisms behind the disease pathology remain largely unknown. In this study we investigated a potential regulatory role for the Hox5 gene family, Hoxa5, Hoxb5, and Hoxc5, genes known to be important in lung development within mesenchymal cell populations. We found that Hox5-mutant mice show exacerbated pathology compared with wild-type controls in a chronic allergen model, with an increased Th2 response and exacerbated lung tissue pathology. Bone marrow chimera experiments indicated that the observed enhanced pathology was mediated by immune cell function independent of mesenchymal cell Hox5 family function. Examination of T cells grown in Th2 polarizing conditions showed increased proliferation, enhanced Gata3 expression, and elevated production of IL-4, IL-5, and IL-13 in Hox5-deficient T cells compared with wild-type controls. Overexpression of FLAG-tagged HOX5 proteins in Jurkat cells demonstrated HOX5 binding to the Gata3 locus and decreased Gata3 and IL-4 expression, supporting a role for HOX5 proteins in direct transcriptional control of Th2 development. These results reveal a novel role for Hox5 genes as developmental regulators of Th2 immune cell function that demonstrates a redeployment of mesenchyme-associated developmental genes.


Subject(s)
Allergens/immunology , GATA3 Transcription Factor/genetics , Gene Expression Regulation , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Inflammation/immunology , Th2 Cells/immunology , Animals , Cell Proliferation , GATA3 Transcription Factor/metabolism , Hedgehog Proteins/deficiency , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Interleukin-13/biosynthesis , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/biosynthesis , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-5/biosynthesis , Interleukin-5/genetics , Interleukin-5/immunology , Jurkat Cells , Lung/immunology , Lung/pathology , Lung/physiology , Mesoderm/cytology , Mice , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Th2 Cells/metabolism , Transcription Factors
13.
Cell Rep ; 19(4): 809-821, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28445731

ABSTRACT

During development and regeneration, matrix progenitors undergo terminal differentiation to form the concentric layers of the hair follicle. These differentiation events are thought to require signals from the mesenchymal dermal papilla (DP); however, it remains unclear how DP-progenitor cell interactions govern specific cell fate decisions. Here, we show that the hair follicle differentiated layers are specified asynchronously, with early matrix progenitors initiating differentiation before surrounding the DP. Furthermore, these early matrix cells can undergo terminal differentiation in the absence of Shh, BMP signaling, and DP maturation. Whereas early matrix progenitors form the hair follicle companion layer, later matrix populations progressively form the inner root sheath and hair shaft. Altogether, our findings characterize some of the earliest terminal differentiation events in the hair follicle and reveal that the matrix progenitor pool can be divided into early and late phases based on distinct temporal, molecular, and functional characteristics.


Subject(s)
Cell Differentiation/physiology , Hair Follicle/cytology , Mesenchymal Stem Cells/cytology , Animals , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/metabolism , Dermis/cytology , GATA3 Transcription Factor/metabolism , Hair Follicle/metabolism , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Keratin-6/metabolism , Keratins/genetics , Keratins/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
14.
PLoS One ; 11(2): e0150263, 2016.
Article in English | MEDLINE | ID: mdl-26918743

ABSTRACT

The developing long bone is a model of endochondral ossification that displays the morphological layers of chondrocytes toward the ossification center of the diaphysis. Indian hedgehog (Ihh), a member of the hedgehog family of secreted molecules, regulates chondrocyte proliferation and differentiation, as well as osteoblast differentiation, through the process of endochondral ossification. Here, we report that the basic helix-loop-helix transcription factor Hand1, which is expressed in the cartilage primordia, is involved in proper osteogenesis of the bone collar via its control of Ihh production. Genetic overexpression of Hand1 in the osteochondral progenitors resulted in prenatal hypoplastic or aplastic ossification in the diaphyses, mimicking an Ihh loss-of-function phenotype. Ihh expression was downregulated in femur epiphyses of Hand1-overexpressing mice. We also confirmed that Hand1 downregulated Ihh gene expression in vitro by inhibiting Runx2 transactivation of the Ihh proximal promoter. These results demonstrate that Hand1 in chondrocytes regulates endochondral ossification, at least in part through the Runx2-Ihh axis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Hedgehog Proteins/physiology , Osteogenesis/physiology , Transcriptional Activation , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone and Bones/embryology , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Line , Core Binding Factor Alpha 1 Subunit/physiology , Diaphyses , Down-Regulation , Female , Genes, Reporter , Growth Plate/metabolism , Hedgehog Proteins/biosynthesis , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Limb Deformities, Congenital/genetics , Male , Mice , Mice, Transgenic , Osteogenesis/genetics , Osteopontin/biosynthesis , Osteopontin/genetics , Phenotype , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Signal Transduction/physiology , Transfection , Twist-Related Protein 1/genetics
15.
Cardiovasc Res ; 109(2): 217-27, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26645982

ABSTRACT

AIMS: Microangiopathy, i.e. endothelial dysfunction, has long been suggested to contribute to the development of diabetic neuropathy, although this has never been fully verified. In the present paper, we have identified the role of Hedgehog (Hh) signalling in endoneurial microvessel integrity and evaluated the impact of impaired Hh signalling in endothelial cells (ECs) on nerve function. METHODS AND RESULTS: By using Desert Hedgehog (Dhh)-deficient mice, we have revealed, that in the absence of Dhh, endoneurial capillaries are abnormally dense and permeable. Furthermore, Smoothened (Smo) conditional KO mice clarified that this increased vessel permeability is specifically due to impaired Hh signalling in ECs and is associated with a down-regulation of Claudin5 (Cldn5). Moreover, impairment of Hh signalling in ECs was sufficient to induce hypoalgesia and neuropathic pain. Finally in Lepr(db/db) type 2 diabetic mice, the loss of Dhh expression observed in the nerve was shown to be associated with increased endoneurial capillary permeability and decreased Cldn5 expression. Conversely, systemic administration of the Smo agonist SAG increased Cldn5 expression, decreased endoneurial capillary permeability, and restored thermal algesia to diabetic mice, demonstrating that loss of Dhh expression is crucial in the development of diabetic neuropathy. CONCLUSION: The present work demonstrates the critical role of Dhh in maintaining blood nerve barrier integrity and demonstrates for the first time that endothelial dysfunction is sufficient to induce neuropathy.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Endothelial Cells/metabolism , Endothelium/physiopathology , Hedgehog Proteins/metabolism , Signal Transduction , Animals , Capillaries/metabolism , Down-Regulation , Hedgehog Proteins/deficiency , Mice, Inbred C57BL , Signal Transduction/physiology , Smoothened Receptor/metabolism
16.
Nature ; 526(7574): 578-82, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26436454

ABSTRACT

Postnatal tissue quiescence is thought to be a default state in the absence of a proliferative stimulus such as injury. Although previous studies have demonstrated that certain embryonic developmental programs are reactivated aberrantly in adult organs to drive repair and regeneration, it is not well understood how quiescence is maintained in organs such as the lung, which displays a remarkably low level of cellular turnover. Here we demonstrate that quiescence in the adult lung is an actively maintained state and is regulated by hedgehog signalling. Epithelial-specific deletion of sonic hedgehog (Shh) during postnatal homeostasis in the murine lung results in a proliferative expansion of the adjacent lung mesenchyme. Hedgehog signalling is initially downregulated during the acute phase of epithelial injury as the mesenchyme proliferates in response, but returns to baseline during injury resolution as quiescence is restored. Activation of hedgehog during acute epithelial injury attenuates the proliferative expansion of the lung mesenchyme, whereas inactivation of hedgehog signalling prevents the restoration of quiescence during injury resolution. Finally, we show that hedgehog also regulates epithelial quiescence and regeneration in response to injury via a mesenchymal feedback mechanism. These results demonstrate that epithelial-mesenchymal interactions coordinated by hedgehog actively maintain postnatal tissue homeostasis, and deregulation of hedgehog during injury leads to aberrant repair and regeneration in the lung.


Subject(s)
Hedgehog Proteins/metabolism , Lung Injury/metabolism , Lung Injury/pathology , Lung/cytology , Lung/metabolism , Regeneration , Wound Healing , Animals , Cell Proliferation , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Feedback, Physiological , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Homeostasis , Lung/pathology , Male , Mesoderm/cytology , Mesoderm/metabolism , Mice , Paracrine Communication
17.
Am J Med Genet A ; 167A(10): 2451-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26080100

ABSTRACT

Solitary Median Maxillary Central Incisor occurs in 1 of 50,000 live births. It is the mildest manifestation of the holoprosencephaly spectrum and is genetically heterogeneous. Here we report six patients with solitary median maxillary central incisor, and a range of other phenotypic anomalies with different degrees of severity, varying from mild signs of holoprosencephaly to associated intellectual disability, and with different genetic background. Using array comparative genomic hybridization, pathogenic copy number variants were found in three of the six patients. Two patients had a deletion at the 18p11 chromosomal region that includes TGIF1 while the other patient had a deletion at 7q36, including the SHH gene. In one patient, a mutation in SIX3 was detected with exome sequencing, while in the two remaining patients all known holoprosencephaly genes were excluded using multiplex ligation-dependent probe amplification and sequencing, and remain unsolved. One of the two latter patients had isolated solitary median maxillary central incisor without other visible dentofacial anomalies, while the other had clinical features not part of the known holoprosencephaly spectrum.


Subject(s)
Anodontia/genetics , Chromosome Deletion , Chromosomes, Human, Pair 18 , Chromosomes, Human, Pair 7 , Genetic Association Studies , Genetic Heterogeneity , Incisor/abnormalities , Adolescent , Anodontia/metabolism , Anodontia/pathology , Child , Comparative Genomic Hybridization , DNA Copy Number Variations , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Genotype , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Holoprosencephaly , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Incisor/metabolism , Incisor/pathology , Male , Maxilla/abnormalities , Maxilla/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phenotype , Repressor Proteins/deficiency , Repressor Proteins/genetics , Young Adult , Homeobox Protein SIX3
18.
Exp Neurol ; 271: 84-94, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25936873

ABSTRACT

The secreted protein desert hedgehog (dhh) controls the formation of the nerve perineurium during development and is a key component of Schwann cells that ensures peripheral nerve survival. We postulated that dhh may play a critical role in maintaining myelination and investigated its role in demyelination-induced compression neuropathies by using a post-natal model of a chronic nerve injury in wildtype and dhh(-/-) mice. We evaluated demyelination using electrophysiological, morphological, and molecular approaches. dhh transcripts and protein are down-regulated early after injury in wild-type mice, suggesting an intimate relationship between the hedgehog pathway and demyelination. In dhh(-/-) mice, nerve injury induced more prominent and severe demyelination relative to their wild-type counterparts, suggesting a protective role of dhh. Alterations in nerve fiber characteristics included significant decreases in nerve conduction velocity, increased myelin debris, and substantial decreases in internodal length. Furthermore, in vitro studies showed that dhh blockade via either adenovirus-mediated (shRNA) or pharmacological inhibition both resulted in severe demyelination, which could be rescued by exogenous Dhh. Exogenous Dhh was protective against this demyelination and maintained myelination at baseline levels in a custom in vitro bioreactor to applied biophysical forces to myelinated DRG/Schwann cell co-cultures. Together, these results demonstrate a pivotal role for dhh in maintaining myelination. Furthermore, dhh signaling reveals a potential target for therapeutic intervention to prevent and treat demyelination of peripheral nerves in compression neuropathies.


Subject(s)
Arthrogryposis/complications , Demyelinating Diseases/etiology , Demyelinating Diseases/metabolism , Gene Expression Regulation/physiology , Hedgehog Proteins/deficiency , Hedgehog Proteins/metabolism , Hereditary Sensory and Motor Neuropathy/complications , Animals , Animals, Newborn , Axons/pathology , Cell Culture Techniques , Disease Models, Animal , Embryo, Mammalian , Functional Laterality/genetics , Ganglia, Spinal/cytology , Hedgehog Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/metabolism , Myelin Sheath/pathology , Neural Conduction/genetics , Neurons/physiology , Peripheral Nerves/pathology , Peripheral Nerves/ultrastructure , Rats , Rats, Sprague-Dawley , Schwann Cells/physiology
19.
PLoS Biol ; 13(3): e1002119, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25826604

ABSTRACT

During nervous system development, gradients of Sonic Hedgehog (Shh) and Netrin-1 attract growth cones of commissural axons toward the floor plate of the embryonic spinal cord. Mice defective for either Shh or Netrin-1 signaling have commissural axon guidance defects, suggesting that both Shh and Netrin-1 are required for correct axon guidance. However, how Shh and Netrin-1 collaborate to guide axons is not known. We first quantified the steepness of the Shh gradient in the spinal cord and found that it is mostly very shallow. We then developed an in vitro microfluidic guidance assay to simulate these shallow gradients. We found that axons of dissociated commissural neurons respond to steep but not shallow gradients of Shh or Netrin-1. However, when we presented axons with combined Shh and Netrin-1 gradients, they had heightened sensitivity to the guidance cues, turning in response to shallower gradients that were unable to guide axons when only one cue was present. Furthermore, these shallow gradients polarized growth cone Src-family kinase (SFK) activity only when Shh and Netrin-1 were combined, indicating that SFKs can integrate the two guidance cues. Together, our results indicate that Shh and Netrin-1 synergize to enable growth cones to sense shallow gradients in regions of the spinal cord where the steepness of a single guidance cue is insufficient to guide axons, and we identify a novel type of synergy that occurs when the steepness (and not the concentration) of a guidance cue is limiting.


Subject(s)
Growth Cones/drug effects , Hedgehog Proteins/pharmacology , Nerve Growth Factors/pharmacology , Spinal Cord/drug effects , Tumor Suppressor Proteins/pharmacology , src-Family Kinases/genetics , Animals , Chemotaxis/physiology , Embryo, Mammalian , Gene Expression Regulation, Developmental , Growth Cones/metabolism , Growth Cones/ultrastructure , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Lab-On-A-Chip Devices , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Imaging , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Netrin-1 , Primary Cell Culture , Signal Transduction , Spinal Cord/growth & development , Spinal Cord/metabolism , Spinal Cord/ultrastructure , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , src-Family Kinases/metabolism
20.
Med Sci (Paris) ; 30(11): 1024-33, 2014 Nov.
Article in French | MEDLINE | ID: mdl-25388585

ABSTRACT

Advances in genomics, bioinformatics and the creation of model organisms have identified many genes associated with polycystic kidney diseases. Historically, these genes were not necessarily associated with ciliopathies, but it appeared that many connections can be made between the cystic kidney disease and function of the primary cilium. Indeed, the proteins encoded by these genes are localized to the cilium itself, to the basal body or are known to regulate the expression and localization of ciliary proteins. The goal of this article is to describe the multiple cellular processes that may lead to the development of renal cysts if they are deregulated. These include changes in proliferation rate, cell polarity or signaling pathways involved in embryonic kidney development. To highlight the role of the primary cilium in cystogenesis, I will discuss several studies investigating the function of ciliary genes and cilia in the kidneys of different model organisms.


Subject(s)
Cilia/physiology , Ciliary Motility Disorders/pathology , Kidney Diseases, Cystic/pathology , Animals , Cell Division , Cell Movement , Cell Polarity , Ciliary Motility Disorders/genetics , Disease Models, Animal , Epithelial Cells/ultrastructure , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Hedgehog Proteins/physiology , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Kidney/embryology , Kidney/ultrastructure , Kidney Diseases, Cystic/classification , Kidney Diseases, Cystic/genetics , Mice , Mitosis , Models, Biological , Morphogenesis , Rats , Signal Transduction/physiology , Wnt Proteins/deficiency , Wnt Proteins/genetics , Wnt Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL