Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biomolecules ; 13(8)2023 08 03.
Article in English | MEDLINE | ID: mdl-37627279

ABSTRACT

Failure to properly form bone or integrate surgical implants can lead to morbidity and additional surgical interventions in a significant proportion of orthopedic surgeries. While the role of skeletal stem cells (SSCs) in bone formation and repair is well-established, very little is known about the factors that regulate the downstream Bone, Cartilage, Stromal, Progenitors (BCSPs). BCSPs, as transit amplifying progenitor cells, undergo multiple mitotic divisions to expand the pool of lineage committed progenitors allowing stem cells to preserve their self-renewal and stemness. Del1 is a protein widely expressed in the skeletal system, but its deletion led to minimal phenotype changes in the uninjured mouse. In this paper, we demonstrate that Del1 is a key regulator of BCSP expansion following injury. In Del1 knockout mice, there is a significant reduction in the number of BCSPs which leads to a smaller callus and decreased bone formation compared with wildtype (WT) littermates. Del1 serves to promote BCSP proliferation and prevent apoptosis in vivo and in vitro. Moreover, exogenous Del1 promotes proliferation of aged human BCSPs. Our results highlight the potential of Del1 as a therapeutic target for improving bone formation and implant success. Del1 injections may improve the success of orthopedic surgeries and fracture healing by enhancing the proliferation and survival of BCSPs, which are crucial for generating new bone tissue during the process of bone formation and repair.


Subject(s)
Bone and Bones , Osteogenesis , Humans , Animals , Mice , Aged , Fracture Healing , Intercellular Signaling Peptides and Proteins , Apoptosis , Mice, Knockout
2.
Nature ; 599(7884): 320-324, 2021 11.
Article in English | MEDLINE | ID: mdl-34707294

ABSTRACT

The Dispatched protein, which is related to the NPC1 and PTCH1 cholesterol transporters1,2 and to H+-driven transporters of the RND family3,4, enables tissue-patterning activity of the lipid-modified Hedgehog protein by releasing it from tightly -localized sites of embryonic expression5-10. Here we determine a cryo-electron microscopy structure of the mouse protein Dispatched homologue 1 (DISP1), revealing three Na+ ions coordinated within a channel that traverses its transmembrane domain. We find that the rate of Hedgehog export is dependent on the Na+ gradient across the plasma membrane. The transmembrane channel and Na+ binding are disrupted in DISP1-NNN, a variant with asparagine substitutions for three intramembrane aspartate residues that each coordinate and neutralize the charge of one of the three Na+ ions. DISP1-NNN and variants that disrupt single Na+ sites retain binding to, but are impaired in export of the lipid-modified Hedgehog protein to the SCUBE2 acceptor. Interaction of the amino-terminal signalling domain of the Sonic hedgehog protein (ShhN) with DISP1 occurs via an extensive buried surface area and contacts with an extended furin-cleaved DISP1 arm. Variability analysis reveals that ShhN binding is restricted to one extreme of a continuous series of DISP1 conformations. The bound and unbound DISP1 conformations display distinct Na+-site occupancies, which suggests a mechanism by which transmembrane Na+ flux may power extraction of the lipid-linked Hedgehog signal from the membrane. Na+-coordinating residues in DISP1 are conserved in PTCH1 and other metazoan RND family members, suggesting that Na+ flux powers their conformationally driven activities.


Subject(s)
Cryoelectron Microscopy , Hedgehog Proteins/chemistry , Hedgehog Proteins/metabolism , Lipid Metabolism , Membrane Proteins/metabolism , Sodium/metabolism , Animals , Binding Sites , Cell Membrane/chemistry , Cell Membrane/metabolism , Hedgehog Proteins/ultrastructure , Membrane Lipids/chemistry , Membrane Lipids/isolation & purification , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/ultrastructure , Mice , Models, Molecular , Mutation
3.
Proc Natl Acad Sci U S A ; 115(2): E200-E209, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29279401

ABSTRACT

How organs maintain and restore functional integrity during ordinary tissue turnover or following injury represents a central biological problem. The maintenance of taste sensory organs in the tongue was shown 140 years ago to depend on innervation from distant ganglion neurons, but the underlying mechanism has remained unknown. Here, we show that Sonic hedgehog (Shh), which encodes a secreted protein signal, is expressed in these sensory neurons, and that experimental ablation of neuronal Shh expression causes loss of taste receptor cells (TRCs). TRCs are also lost upon pharmacologic blockade of Hedgehog pathway response, accounting for the loss of taste sensation experienced by cancer patients undergoing Hedgehog inhibitor treatment. We find that TRC regeneration following such pharmacologic ablation requires neuronal expression of Shh and can be substantially enhanced by pharmacologic activation of Hedgehog response. Such pharmacologic enhancement of Hedgehog response, however, results in additional TRC formation at many ectopic sites, unlike the site-restricted regeneration specified by the projection pattern of Shh-expressing neurons. Stable regeneration of TRCs thus requires neuronal Shh, illustrating the principle that neuronal delivery of cues such as the Shh signal can pattern distant cellular responses to assure functional integrity during tissue maintenance and regeneration.


Subject(s)
Epithelium/metabolism , Hedgehog Proteins/metabolism , Taste Buds/metabolism , Tongue/metabolism , Animals , Epithelium/growth & development , Epithelium/physiology , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organogenesis/genetics , Regeneration/genetics , Signal Transduction/genetics , Taste/genetics , Taste Buds/cytology , Taste Buds/growth & development , Time Factors , Tongue/cytology , Tongue/growth & development
4.
Proc Natl Acad Sci U S A ; 113(21)2016 May 24.
Article in English | MEDLINE | ID: mdl-27162362

ABSTRACT

Cellular lipids are speculated to act as key intermediates in Hedgehog signal transduction, but their precise identity and function remain enigmatic. In an effort to identify such lipids, we pursued a Hedgehog pathway inhibitory activity that is particularly abundant in flagellar lipids of Chlamydomonas reinhardtii, resulting in the purification and identification of ergosterol endoperoxide, a B-ring oxysterol. A mammalian analog of ergosterol, 7-dehydrocholesterol (7-DHC), accumulates in Smith-Lemli-Opitz syndrome, a human genetic disease that phenocopies deficient Hedgehog signaling and is caused by genetic loss of 7-DHC reductase. We found that depleting endogenous 7-DHC with methyl-ß-cyclodextrin treatment enhances Hedgehog activation by a pathway agonist. Conversely, exogenous addition of 3ß,5α-dihydroxycholest-7-en-6-one, a naturally occurring B-ring oxysterol derived from 7-DHC that also accumulates in Smith-Lemli-Opitz syndrome, blocked Hedgehog signaling by inhibiting activation of the essential transduction component Smoothened, through a mechanism distinct from Smoothened modulation by other lipids.


Subject(s)
Dehydrocholesterols/metabolism , Hedgehog Proteins/metabolism , Signal Transduction , Smoothened Receptor/metabolism , Animals , Chlamydomonas reinhardtii/chemistry , Dehydrocholesterols/chemistry , Dehydrocholesterols/pharmacology , Flagella/chemistry , HEK293 Cells , Hedgehog Proteins/genetics , Humans , Mice , NIH 3T3 Cells , Smith-Lemli-Opitz Syndrome/genetics , Smith-Lemli-Opitz Syndrome/metabolism , Smoothened Receptor/genetics , Veratrum Alkaloids/pharmacology , beta-Cyclodextrins/pharmacology
5.
Genes Dev ; 29(3): 262-76, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25644602

ABSTRACT

Binding of the Hedgehog (Hh) protein signal to its receptor, Patched, induces accumulation of the seven-pass transmembrane protein Smoothened (Smo) within the primary cilium and of the zinc finger transcription factor Gli2 at the ciliary tip, resulting ultimately in Gli-mediated changes in nuclear gene expression. However, the mechanism by which pathway activation is communicated from Smo to Gli2 is not known. In an effort to elucidate this mechanism, we identified Dlg5 (Discs large, homolog 5) in a biochemical screen for proteins that preferentially interact with activated Smo. We found that disruption of Smo-Dlg5 interactions or depletion of endogenous Dlg5 leads to diminished Hh pathway response without a significant impact on Smo ciliary accumulation. We also found that Dlg5 is localized at the basal body, where it associates with another pathway component, Kif7. We show that Dlg5 is required for Hh-induced enrichment of Kif7 and Gli2 at the tip of the cilium but is dispensable for Gpr161 exit from the cilium and the consequent suppression of Gli3 processing into its repressor form. Our findings suggest a bifurcation of Smo activity in Hh response, with a Dlg5-independent arm for suppression of Gli repressor formation and a second arm involving Smo interaction with Dlg5 for Gli activation.


Subject(s)
Guanylate Kinases/metabolism , Hedgehog Proteins/metabolism , Membrane Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Amino Acid Motifs , Animals , Basal Bodies/metabolism , Cell Line , Cilia/metabolism , Guanylate Kinases/genetics , HEK293 Cells , Humans , Kinesins/metabolism , Kruppel-Like Transcription Factors/metabolism , Membrane Proteins/genetics , Mice , NIH 3T3 Cells , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Transport , Receptors, G-Protein-Coupled/genetics , Smoothened Receptor , Zinc Finger Protein Gli2 , Zinc Finger Protein Gli3
6.
Genes Dev ; 26(12): 1312-25, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22677548

ABSTRACT

Owing to their covalent modification by cholesterol and palmitate, Hedgehog (Hh) signaling proteins are localized predominantly to the plasma membrane of expressing cells. Yet Hh proteins are also capable of mobilizing to and eliciting direct responses from distant cells. The zebrafish you gene, identified genetically >15 years ago, was more recently shown to encode a secreted glycoprotein that acts cell-nonautonomously in the Hh signaling pathway by an unknown mechanism. We investigated the function of the protein encoded by murine Scube2, an ortholog of you, and found that it mediates release in soluble form of the mature, cholesterol- and palmitate-modified Sonic hedgehog protein signal (ShhNp) when added to cultured cells or purified detergent-resistant membrane microdomains containing ShhNp. The efficiency of Scube2-mediated release of ShhNp is enhanced by the palmitate adduct of ShhNp and by coexpression in ShhNp-producing cells of mDispatchedA (mDispA), a transporter-like protein with a previously defined role in the release of lipid-modified Hh signals. The structural determinants of Scube2 required for its activity in cultured cell assays match those required for rescue of you mutant zebrafish embryos, and we thus conclude that the role of Scube/You proteins in Hh signaling in vivo is to facilitate the release and mobilization of Hh proteins for distant action.


Subject(s)
Hedgehog Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cell-Free System , Cells, Cultured , Cholesterol/metabolism , Culture Media/pharmacology , Detergents/pharmacology , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Lipid Metabolism/drug effects , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Mice , Palmitates/pharmacology , Protein Binding/drug effects , Protein Stability/drug effects , Signal Transduction/drug effects , Solubility/drug effects , Structure-Activity Relationship , Zebrafish
7.
Genes Dev ; 24(1): 57-71, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20048000

ABSTRACT

Although the transporter-like protein Patched (Ptc) is genetically implicated in reception of the extracellular Hedgehog (Hh) protein signal, a clear definition of the Hh receptor is complicated by the existence of additional Hh-binding proteins and, in Drosophila, by the lack of physical evidence for direct binding of Hh to Ptc. Here we show that activity of Ihog (Interference hedgehog), or of its close relative Boi (Brother of Ihog), is absolutely required for Hh biological response and for sequestration of the Hh protein to limit long-range signaling. We demonstrate that Ihog interacts directly with Ptc, is required for presentation of Ptc on the cell surface, and that Ihog and Ptc are both required for high-affinity Hh binding. On the basis of their joint roles in ligand binding, signal transduction, and receptor trafficking, we conclude that Ihog and Ptc together constitute the Drosophila Hh receptor.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Animals , Body Patterning/genetics , Cells, Cultured , Gene Expression Regulation, Developmental , Mutation , Protein Binding , Protein Structure, Tertiary
8.
Annu Rev Biochem ; 73: 891-923, 2004.
Article in English | MEDLINE | ID: mdl-15189162

ABSTRACT

Secreted signaling proteins function in a diverse array of essential patterning events during metazoan development, ranging from embryonic segmentation in insects to neural tube differentiation in vertebrates. These proteins generally are expressed in a localized manner, and they may elicit distinct concentration-dependent responses in the cells of surrounding tissues and structures, thus functioning as morphogens that specify the pattern of cellular responses by their tissue distribution. Given the importance of signal distribution, it is notable that the Hedgehog (Hh) and Wnt proteins, two of the most important families of such signals, are known to be covalently modified by lipid moieties, the membrane-anchoring properties of which are not consistent with passive models of protein mobilization within tissues. This review focuses on the mechanisms underlying biogenesis of the mature Hh proteins, which are dually modified by cholesteryl and palmitoyl adducts, as well as on the relationship between Hh proteins and the self-splicing proteins (i.e., proteins containing inteins) and the Hh-like proteins of nematodes. We further discuss the cellular mechanisms that have evolved to handle lipidated Hh proteins in the spatial deployment of the signal in developing tissues and the more recent findings that implicate palmitate modification as an important feature of Wnt signaling proteins.


Subject(s)
Drosophila Proteins/chemistry , Trans-Activators/chemistry , Amino Acid Sequence , Animals , Body Patterning , Cholesterol/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Embryonic Induction , Hedgehog Proteins , Humans , Lipids/chemistry , Models, Molecular , Molecular Sequence Data , Palmitic Acids/chemistry , Protein Processing, Post-Translational , Protein Sorting Signals/genetics , Protein Sorting Signals/physiology , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/physiology , Sequence Homology, Amino Acid , Signal Transduction , Tissue Distribution , Trans-Activators/genetics , Trans-Activators/physiology , Wnt Proteins
9.
Mol Cell ; 12(5): 1261-74, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14636583

ABSTRACT

The seven-transmembrane protein Smoothened (Smo) transduces extracellular activation of the Hedgehog (Hh) pathway by an unknown mechanism to increase transcriptional activity of the latent cytoplasmic transcription factor Ci (Cubitus interruptus). Here, we present evidence that Smo associates directly with a Ci-containing complex that is scaffolded and stabilized by the atypical kinesin, Costal-2 (Cos2). This complex constitutively suppresses pathway activity, but Hh signaling reverses its regulatory effect to promote Ci-mediated transcription. In response to Hh activation of Smo, Cos2 mediates accumulation and phosphorylation of Smo at the membrane as well as phosphorylation of the cytoplasmic components Fu and Su(fu). Positive response of Cos2 to Hh stimulation requires a portion of the Smo cytoplasmic tail and the Cos2 cargo domain, which interacts directly with Smo.


Subject(s)
Drosophila Proteins/metabolism , Kinesins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Cell Line , Cytoplasm/chemistry , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Embryonic Structures/anatomy & histology , Embryonic Structures/physiology , Gene Expression Regulation, Developmental , Genes, Reporter , Hedgehog Proteins , Humans , Kinesins/genetics , Macromolecular Substances , Molecular Sequence Data , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Alignment , Smoothened Receptor , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...