Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.765
Filter
1.
Proc Natl Acad Sci U S A ; 121(28): e2320070121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968120

ABSTRACT

Hedgehog (Hh) signaling, an evolutionarily conserved pathway, plays an essential role in development and tumorigenesis, making it a promising drug target. Multiple negative regulators are known to govern Hh signaling; however, how activated Smoothened (SMO) participates in the activation of downstream GLI2 and GLI3 remains unclear. Herein, we identified the ciliary kinase DYRK2 as a positive regulator of the GLI2 and GLI3 transcription factors for Hh signaling. Transcriptome and interactome analyses demonstrated that DYRK2 phosphorylates GLI2 and GLI3 on evolutionarily conserved serine residues at the ciliary base, in response to activation of the Hh pathway. This phosphorylation induces the dissociation of GLI2/GLI3 from suppressor, SUFU, and their translocation into the nucleus. Loss of Dyrk2 in mice causes skeletal malformation, but neural tube development remains normal. Notably, DYRK2-mediated phosphorylation orchestrates limb development by controlling cell proliferation. Taken together, the ciliary kinase DYRK2 governs the activation of Hh signaling through the regulation of two processes: phosphorylation of GLI2 and GLI3 downstream of SMO and cilia formation. Thus, our findings of a unique regulatory mechanism of Hh signaling expand understanding of the control of Hh-associated diseases.


Subject(s)
Dyrk Kinases , Hedgehog Proteins , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Signal Transduction , Zinc Finger Protein Gli2 , Zinc Finger Protein Gli3 , Animals , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli2/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Mice , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Humans , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Cell Proliferation , Cilia/metabolism , Smoothened Receptor/metabolism , Smoothened Receptor/genetics , Nuclear Proteins , Repressor Proteins
2.
Stem Cell Res Ther ; 15(1): 198, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971766

ABSTRACT

BACKGROUND: Trans-sutural distraction osteogenesis (TSDO) involves the application of distraction force to facial sutures to stimulate osteogenesis. Gli1+ cells in the cranial sutures play an important role in bone growth. However, whether Gli1+ cells in facial sutures differentiate into bone under distraction force is unknown. METHODS: 4-week-old Gli1ER/Td and C57BL/6 mice were used to establish a TSDO model to explore osteogenesis of zygomaticomaxillary sutures. A Gli1+ cell lineage tracing model was used to observe the distribution of Gli1+ cells and explore the role of Gli1+ cells in facial bone remodeling. RESULTS: Distraction force promoted bone remodeling during TSDO. Fluorescence and two-photon scanning images revealed the distribution of Gli1+ cells. Under distraction force, Gli1-lineage cells proliferated significantly and co-localized with Runx2+ cells. Hedgehog signaling was upregulated in Gli1+ cells. Inhibition of Hedgehog signaling suppresses the proliferation and osteogenesis of Gli1+ cells induced by distraction force. Subsequently, the stem cell characteristics of Gli1+ cells were identified. Cell-stretching experiments verified that mechanical force promoted the osteogenic differentiation of Gli1+ cells through Hh signaling. Furthermore, immunofluorescence staining and RT-qPCR experiments demonstrated that the primary cilia in Gli1+ cells exhibit Hedgehog-independent mechanosensitivity, which was required for the osteogenic differentiation induced by mechanical force. CONCLUSIONS: Our study indicates that the primary cilia of Gli1+ cells sense mechanical stimuli, mediate Hedgehog signaling activation, and promote the osteogenic differentiation of Gli1+ cells in zygomaticomaxillary sutures.


Subject(s)
Cell Differentiation , Cilia , Cranial Sutures , Hedgehog Proteins , Osteogenesis , Signal Transduction , Zinc Finger Protein GLI1 , Animals , Mice , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Osteogenesis/physiology , Cilia/metabolism , Cranial Sutures/metabolism , Mice, Inbred C57BL , Osteogenesis, Distraction/methods , Cell Proliferation
3.
Transl Vis Sci Technol ; 13(7): 11, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39007834

ABSTRACT

Purpose: Microphthalmia is a rare developmental eye disease that affects 1 in 7000 births. Currently, there is no cure for this condition. This study aimed to construct a stable mouse model of microphthalmia, thus providing a new tool for the study of the etiology of microphthalmia. Methods: The Hedgehog signaling pathway plays a crucial role in eye development. One of the key mechanisms of the Sonic Hedgehog signaling is the strong transcriptional activation ability of GLI3, a major mediator of this pathway. This study used CRISPR/Cas9 system to construct a novel TgGli3Ki/Ki lens-specific over-expression mouse line. To identify the ocular characteristics of this line, quantitative PCR, Western blot, hematoxylin and eosin staining, immunofluorescent staining, and RNA-seq were performed on the ocular tissues of this line and normal mice. Results: The TgGli3Ki/Ki lens-specific over-expression mouse model exhibits the ocular phenotype of microphthalmia. In the TgGli3Ki/Ki mouse, Gli3 is over-expressed in the lens, and the size of the eyeball and lens is significantly smaller than the normal one. RNA-seq analysis using the lens and the retina samples from TgGli3Ki/Ki and normal mice indicates that the phototransduction pathway is ectopically activated in the lens. Immunofluorescent staining of the lens samples confirmed this activation. Conclusions: The TgGli3Ki/Ki mouse model consistently manifests the stereotypical microphthalmia phenotype across generations, making it an excellent tool for studying this severe eye disease. Translational Relevance: This study developed a novel animal model to facilitate clinical research on microphthalmia.


Subject(s)
Disease Models, Animal , Microphthalmos , Zinc Finger Protein Gli3 , Animals , Microphthalmos/genetics , Microphthalmos/pathology , Microphthalmos/metabolism , Mice , Zinc Finger Protein Gli3/genetics , Zinc Finger Protein Gli3/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Signal Transduction , CRISPR-Cas Systems , Mice, Inbred C57BL , Nerve Tissue Proteins
4.
Oncol Rep ; 52(3)2024 Sep.
Article in English | MEDLINE | ID: mdl-38994763

ABSTRACT

In years of research on classical pathways, the composition, information transmission mechanism, crosstalk with other pathways, and physiological and pathological effects of hedgehog (HH) pathway have been gradually clarified. HH also plays a critical role in tumor formation and development. According to the update of interpretation of tumor phenotypes, the latest relevant studies have been sorted out, to explore the specific mechanism of HH pathway in regulating different tumor phenotypes through gene mutation and signal regulation. The drugs and natural ingredients involved in regulating HH pathway were also reviewed; five approved drugs and drugs under research exert efficacy by blocking HH pathway, and at least 22 natural components have potential to treat tumors by HH pathway. Nevertheless, there is a deficiency of existing studies. The present review confirmed the great potential of HH pathway in future cancer treatment with factual basis.


Subject(s)
Hedgehog Proteins , Neoplasms , Signal Transduction , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Signal Transduction/drug effects , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Animals , Mutation
5.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000154

ABSTRACT

Putatively, tooth agenesis was attributed to the initiation failure of tooth germs, though little is known about the histological and molecular alterations. To address if constitutively active FGF signaling is associated with tooth agenesis, we activated Fgf8 in dental mesenchyme with Osr-cre knock-in allele in mice (Osr2-creKI; Rosa26R-Fgf8) and found incisor agenesis and molar microdontia. The cell survival assay showed tremendous apoptosis in both the Osr2-creKI; Rosa26R-Fgf8 incisor epithelium and mesenchyme, which initiated incisor regression from cap stage. In situ hybridization displayed vanished Shh transcription, and immunostaining exhibited reduced Runx2 expression and enlarged mesenchymal Lef1 domain in Osr2-creKI; Rosa26R-Fgf8 incisors, both of which were suggested to enhance apoptosis. In contrast, Osr2-creKI; Rosa26R-Fgf8 molar germs displayed mildly suppressed Shh transcription, and the increased expression of Ectodin, Runx2 and Lef1. Although mildly smaller than WT controls prenatally, the Osr2-creKI; Rosa26R-Fgf8 molar germs produced a miniature tooth with impaired mineralization after a 6-week sub-renal culture. Intriguingly, the implanted Osr2-creKI; Rosa26R-Fgf8 molar germs exhibited delayed odontoblast differentiation and accelerated ameloblast maturation. Collectively, the ectopically activated Fgf8 in dental mesenchyme caused incisor agenesis by triggering incisor regression and postnatal molar microdontia. Our findings reported tooth agenesis resulting from the regression from the early bell stage and implicated a correlation between tooth agenesis and microdontia.


Subject(s)
Fibroblast Growth Factor 8 , Incisor , Mesoderm , Molar , Animals , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Mice , Incisor/abnormalities , Incisor/metabolism , Mesoderm/metabolism , Mesoderm/pathology , Molar/abnormalities , Molar/metabolism , Anodontia/genetics , Anodontia/metabolism , Anodontia/pathology , Apoptosis , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Signal Transduction , Gene Expression Regulation, Developmental , Odontogenesis/genetics , Mice, Transgenic
6.
Int J Mol Sci ; 25(13)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39000427

ABSTRACT

The amyloid-beta peptide (Aß) is the neurotoxic component in senile plaques of Alzheimer's disease (AD) brains. Previously we have reported that Aß toxicity is mediated by the induction of sonic hedgehog (SHH) to trigger cell cycle re-entry (CCR) and apoptosis in post-mitotic neurons. Basella alba is a vegetable whose polysaccharides carry immunomodulatory and anti-cancer actions, but their protective effects against neurodegeneration have never been reported. Herein, we tested whether polysaccharides derived from Basella alba (PPV-6) may inhibit Aß toxicity and explored its underlying mechanisms. In differentiated rat cortical neurons, Aß25-35 reduced cell viability, damaged neuronal structure, and compromised mitochondrial bioenergetic functions, all of which were recovered by PPV-6. Immunocytochemistry and western blotting revealed that Aß25-35-mediated induction of cell cycle markers including cyclin D1, proliferating cell nuclear antigen (PCNA), and histone H3 phosphorylated at Ser-10 (p-Histone H3) in differentiated neurons was all suppressed by PPV-6, along with mitigation of caspase-3 cleavage. Further studies revealed that PPV-6 inhibited Aß25-35 induction of SHH; indeed, PPV-6 was capable of suppressing neuronal CCR and apoptosis triggered by the exogenous N-terminal fragment of sonic hedgehog (SHH-N). Our findings demonstrated that, in the fully differentiated neurons, PPV-6 exerts protective actions against Aß neurotoxicity via the downregulation of SHH to suppress neuronal CCR and apoptosis.


Subject(s)
Amyloid beta-Peptides , Apoptosis , Cell Cycle , Hedgehog Proteins , Neurons , Polysaccharides , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Hedgehog Proteins/metabolism , Animals , Neurons/drug effects , Neurons/metabolism , Apoptosis/drug effects , Rats , Polysaccharides/pharmacology , Polysaccharides/chemistry , Cell Cycle/drug effects , Peptide Fragments , Cell Survival/drug effects , Neuroprotective Agents/pharmacology
7.
Molecules ; 29(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38999049

ABSTRACT

Aberrant activation of hedgehog (Hh) signaling has been implicated in various cancers. Current FDA-approved inhibitors target the seven-transmembrane receptor Smoothened, but resistance to these drugs has been observed. It has been proposed that a more promising strategy to target this pathway is at the GLI1 transcription factor level. GANT61 was the first small molecule identified to directly suppress GLI-mediated activity; however, its development as a potential anti-cancer agent has been hindered by its modest activity and aqueous chemical instability. Our study aimed to identify novel GLI1 inhibitors. JChem searches identified fifty-two compounds similar to GANT61 and its active metabolite, GANT61-D. We combined high-throughput cell-based assays and molecular docking to evaluate these analogs. Five of the fifty-two GANT61 analogs inhibited activity in Hh-responsive C3H10T1/2 and Gli-reporter NIH3T3 cellular assays without cytotoxicity. Two of the GANT61 analogs, BAS 07019774 and Z27610715, reduced Gli1 mRNA expression in C3H10T1/2 cells. Treatment with BAS 07019774 significantly reduced cell viability in Hh-dependent glioblastoma and lung cancer cell lines. Molecular docking indicated that BAS 07019774 is predicted to bind to the ZF4 region of GLI1, potentially interfering with its ability to bind DNA. Our findings show promise in developing more effective and potent GLI inhibitors.


Subject(s)
Hedgehog Proteins , Molecular Docking Simulation , Pyridines , Pyrimidines , Zinc Finger Protein GLI1 , Pyridines/pharmacology , Pyridines/chemistry , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Pyrimidines/pharmacology , Pyrimidines/chemistry , Hedgehog Proteins/metabolism , Humans , Animals , Mice , Cell Line, Tumor , NIH 3T3 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Signal Transduction/drug effects , Cell Survival/drug effects
9.
Cell Chem Biol ; 31(7): 1231-1233, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39029450

ABSTRACT

In this issue of Cell Chemical Biology, Liu et al.1 report the identification of Q29, a synthetic diterpenoid that blocks covalent cholesterol modification of smoothened (SMO) and inhibits hedgehog signaling. Q29 is capable of suppressing tumor cell growth, both in vitro and in vivo, and overcoming resistance to SMO inhibitors.


Subject(s)
Cholesterol , Diterpenes , Drug Resistance, Neoplasm , Hedgehog Proteins , Smoothened Receptor , Humans , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/metabolism , Drug Resistance, Neoplasm/drug effects , Cholesterol/metabolism , Cholesterol/chemistry , Diterpenes/pharmacology , Diterpenes/chemistry , Hedgehog Proteins/metabolism , Hedgehog Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Animals , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Signal Transduction/drug effects
10.
Vitam Horm ; 126: 97-111, 2024.
Article in English | MEDLINE | ID: mdl-39029978

ABSTRACT

Sonic hedgehog (Shh) is a secreted glycopeptide belonging to the hedgehog family that is essential for morphogenesis during embryonic development. The Shh signal is mediated by two membrane proteins, Patched-1 (Ptch-1) and Smoothened (Smo), following the activation of transcription factors such as Gli. Shh decreases the permeability of the blood-brain barrier (BBB) and plays a key role in its function. In the damaged brain, BBB function is remarkably disrupted. The BBB disruption causes brain edema and neuroinflammation resulting from the extravasation of serum components and the infiltration of inflammatory cells into the cerebral parenchyma. Multiple studies have suggested that astrocyte is a source of Shh and that astrocytic Shh production is increased in the damaged brain. In various experimental animal models of acute brain injury, Shh or Shh signal activators alleviate BBB disruption by increasing tight junction proteins in endothelial cells. Furthermore, activation of astrocytic Shh signaling reduces reactive astrogliosis, neuroinflammation, and increases the production of vascular protective factors, which alleviates BBB disruption in the damaged brain. These findings suggest that astrocytic Shh and Shh signaling protect BBB function in the damaged brain and that target drugs for Shh signaling are expected to be novel therapeutic drugs for acute brain injuries.


Subject(s)
Astrocytes , Blood-Brain Barrier , Hedgehog Proteins , Signal Transduction , Hedgehog Proteins/metabolism , Blood-Brain Barrier/metabolism , Astrocytes/metabolism , Animals , Humans , Signal Transduction/physiology
11.
Sci Rep ; 14(1): 14669, 2024 06 25.
Article in English | MEDLINE | ID: mdl-38918575

ABSTRACT

Non-obese diabetes (NOD) mice are an established, spontaneous model of type 1 diabetes in which diabetes develops through insulitis. Using next-generation sequencing, coupled with pathway analysis, the molecular fingerprint of early insulitis was mapped in a cohort of mice ranging from 4 to 12 weeks of age. The resulting dynamic timeline revealed an initial decrease in proliferative capacity followed by the emergence of an inflammatory signature between 6 and 8 weeks that increased to a regulatory plateau between 10 and 12 weeks. The inflammatory signature is identified by the activation of central immunogenic factors such as Infg, Il1b, and Tnfa, and activation of canonical inflammatory signaling. Analysis of the regulatory landscape revealed the transcription factor Atf3 as a potential novel modulator of inflammatory signaling in the NOD islets. Furthermore, the Hedgehog signaling pathway correlated with Atf3 regulation, suggesting that the two play a role in regulating islet inflammation; however, further studies are needed to establish the nature of this connection.


Subject(s)
Activating Transcription Factor 3 , Diabetes Mellitus, Type 1 , Islets of Langerhans , Mice, Inbred NOD , Signal Transduction , Animals , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 3/genetics , Mice , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Female , Inflammation/genetics , Inflammation/pathology , Inflammation/metabolism , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Gene Expression Profiling , Disease Models, Animal
12.
Cells ; 13(12)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38920633

ABSTRACT

The primary cilium, an antenna-like sensory organelle that protrudes from the surface of most eukaryotic cell types, has become a signaling hub of growing interest given that defects in its structure and/or function are associated with human diseases and syndromes, known as ciliopathies. With the continuously expanding role of primary cilia in health and diseases, identifying new players in ciliogenesis will lead to a better understanding of the function of this organelle. It has been shown that the primary cilium shares similarities with the immune synapse, a highly organized structure at the interface between an antigen-presenting or target cell and a lymphocyte. Studies have demonstrated a role for known cilia regulators in immune synapse formation. However, whether immune synapse regulators modulate ciliogenesis remains elusive. Here, we find that programmed death ligand 1 (PD-L1), an immune checkpoint protein and regulator of immune synapse formation, plays a role in the regulation of ciliogenesis. We found that PD-L1 is enriched at the centrosome/basal body and Golgi apparatus of ciliated cells and depleting PD-L1 enhanced ciliogenesis and increased the accumulation of ciliary membrane trafficking proteins Rab8a, BBS5, and sensory receptor protein PC-2. Moreover, PD-L1 formed a complex with BBS5 and PC-2. In addition, we found that depletion of PD-L1 resulted in the ciliary accumulation of Gli3 and the downregulation of Gli1. Our results suggest that PD-L1 is a new player in ciliogenesis, contributing to PC-2-mediated sensory signaling and the Hh signaling cascade.


Subject(s)
B7-H1 Antigen , Cilia , Hedgehog Proteins , Signal Transduction , Cilia/metabolism , B7-H1 Antigen/metabolism , Hedgehog Proteins/metabolism , Humans , Animals , Mice , Centrosome/metabolism , Golgi Apparatus/metabolism
13.
PLoS One ; 19(6): e0301670, 2024.
Article in English | MEDLINE | ID: mdl-38917070

ABSTRACT

The Hedgehog (HH) pathway is crucial for embryonic development, and adult homeostasis. Its dysregulation is implicated in multiple diseases. Existing cellular models used to study HH signal regulation in mammals do not fully recapitulate the complexity of the pathway. Here we show that Spinal Cord Organoids (SCOs) can be applied to quantitively study the activity of the HH pathway. During SCO formation, the specification of different categories of neural progenitors (NPC) depends on the intensity of the HH signal, mirroring the process that occurs during neural tube development. By assessing the number of NPCs within these distinct subgroups, we are able to categorize and quantify the activation level of the HH pathway. We validate this system by measuring the effects of mutating the HH receptor PTCH1 and the impact of HH agonists and antagonists on NPC specification. SCOs represent an accessible and reliable in-vitro tool to quantify HH signaling and investigate the contribution of genetic and chemical cues in the HH pathway regulation.


Subject(s)
Hedgehog Proteins , Organoids , Signal Transduction , Spinal Cord , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Animals , Organoids/metabolism , Organoids/cytology , Spinal Cord/metabolism , Spinal Cord/cytology , Mice , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Patched-1 Receptor/metabolism , Patched-1 Receptor/genetics
14.
PLoS One ; 19(6): e0294835, 2024.
Article in English | MEDLINE | ID: mdl-38848388

ABSTRACT

The Hedgehog (HH) pathway regulates embryonic development of anterior tongue taste fungiform papilla (FP) and the posterior circumvallate (CVP) and foliate (FOP) taste papillae. HH signaling also mediates taste organ maintenance and regeneration in adults. However, there are knowledge gaps in HH pathway component expression during postnatal taste organ differentiation and maturation. Importantly, the HH transcriptional effectors GLI1, GLI2 and GLI3 have not been investigated in early postnatal stages; the HH receptors PTCH1, GAS1, CDON and HHIP, required to either drive HH pathway activation or antagonism, also remain unexplored. Using lacZ reporter mouse models, we mapped expression of the HH ligand SHH, HH receptors, and GLI transcription factors in FP, CVP and FOP in early and late postnatal and adult stages. In adults we also studied the soft palate, and the geniculate and trigeminal ganglia, which extend afferent fibers to the anterior tongue. Shh and Gas1 are the only components that were consistently expressed within taste buds of all three papillae and the soft palate. In the first postnatal week, we observed broad expression of HH signaling components in FP and adjacent, non-taste filiform (FILIF) papillae in epithelium or stroma and tongue muscles. Notably, we observed elimination of Gli1 in FILIF and Gas1 in muscles, and downregulation of Ptch1 in lingual epithelium and of Cdon, Gas1 and Hhip in stroma from late postnatal stages. Further, HH receptor expression patterns in CVP and FOP epithelium differed from anterior FP. Among all the components, only known positive regulators of HH signaling, SHH, Ptch1, Gli1 and Gli2, were expressed in the ganglia. Our studies emphasize differential regulation of HH signaling in distinct postnatal developmental periods and in anterior versus posterior taste organs, and lay the foundation for functional studies to understand the roles of numerous HH signaling components in postnatal tongue development.


Subject(s)
Hedgehog Proteins , Signal Transduction , Taste Buds , Tongue , Animals , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Tongue/metabolism , Tongue/growth & development , Mice , Taste Buds/metabolism , Taste Buds/growth & development , Gene Expression Regulation, Developmental , Homeostasis , Patched-1 Receptor/metabolism , Patched-1 Receptor/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Nerve Tissue Proteins , Cell Cycle Proteins , GPI-Linked Proteins
15.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891790

ABSTRACT

Derived from axial structures, Sonic Hedgehog (Shh) is secreted into the paraxial mesoderm, where it plays crucial roles in sclerotome induction and myotome differentiation. Through conditional loss-of-function in quail embryos, we investigate the timing and impact of Shh activity during early formation of sclerotome-derived vertebrae and ribs, and of lateral mesoderm-derived sternum. To this end, Hedgehog interacting protein (Hhip) was electroporated at various times between days 2 and 5. While the vertebral body and rib primordium showed consistent size reduction, rib expansion into the somatopleura remained unaffected, and the sternal bud developed normally. Additionally, we compared these effects with those of locally inhibiting BMP activity. Transfection of Noggin in the lateral mesoderm hindered sternal bud formation. Unlike Hhip, BMP inhibition via Noggin or Smad6 induced myogenic differentiation of the lateral dermomyotome lip, while impeding the growth of the myotome/rib complex into the somatic mesoderm, thus affirming the role of the lateral dermomyotome epithelium in rib guidance. Overall, these findings underscore the continuous requirement for opposing gradients of Shh and BMP activity in the morphogenesis of proximal and distal flank skeletal structures, respectively. Future research should address the implications of these early interactions to the later morphogenesis and function of the musculo-skeletal system and of possible associated malformations.


Subject(s)
Hedgehog Proteins , Ribs , Spine , Animals , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Ribs/metabolism , Ribs/embryology , Spine/metabolism , Spine/embryology , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Mesoderm/embryology , Quail , Somites/metabolism , Somites/embryology , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Carrier Proteins
16.
Drug Discov Today ; 29(7): 104053, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38849028

ABSTRACT

Pancreatic cancer (PC), a disease with high heterogeneity and a dense stromal microenvironment, presents significant challenges and a bleak prognosis. Recent breakthroughs have illuminated the crucial interplay among RAS, epidermal growth factor receptor (EGFR), and hedgehog pathways in PC progression. Small molecular inhibitors have emerged as a potential solution with their advantages of oral administration and the ability to target intracellular and extracellular sites effectively. However, despite the US FDA approving over 100 small-molecule targeted antitumor drugs, challenges such as low response rates and drug resistance persist. This review delves into the possibility of using small molecules to treat persistent or spreading PC, highlighting the challenges and the urgent need for a diverse selection of inhibitors to develop more effective treatment strategies.


Subject(s)
Antineoplastic Agents , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Animals , Drug Resistance, Neoplasm , Molecular Targeted Therapy , ErbB Receptors/antagonists & inhibitors , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
17.
Eur J Endocrinol ; 191(1): 38-46, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38917024

ABSTRACT

BACKGROUND: Heterozygous Indian Hedgehog gene (IHH) variants are associated with brachydactyly type A1 (BDA1). However, in recent years, numerous variants have been identified in patients with short stature and more variable forms of brachydactyly. Many are located in the C-terminal domain of IHH (IHH-C), which lacks signaling activity but is critical for auto-cleavage and activation of the N-terminal (IHH-N) peptide. The absence of functional studies of IHH variants, particularly for those located in IHH-C, has led to these variants being classified as variants of uncertain significance (VUS). OBJECTIVE: To establish a simple functional assay to determine the pathogenicity of IHH VUS and confirm that variants in the C-terminal domain affect protein function. DESIGN/METHODS: In vitro studies were performed for 9 IHH heterozygous variants, to test their effect on secretion and IHH intracellular processing by western blot of cells expressing each variant. RESULTS: IHH secretion was significantly reduced in all mutants, regardless of the location. Similarly, intracellular levels of N-terminal and C-terminal IHH peptides were severely reduced in comparison with the control. Two variants present at a relatively high frequency in the general population also reduced secretion but to a lesser degree in the heterozygous state. CONCLUSIONS: These studies provide the first evidence that variants in the C-terminal domain affect the secretion capacity of IHH and thus, reduce availability of IHH ligand, resulting in short stature and mild skeletal defects. The secretion assay permits a relatively easy test to determine the pathogenicity of IHH variants. All studied variants affected secretion and interestingly, more frequent population variants appear to have a deleterious effect and thus contribute to height variation.


Subject(s)
Hedgehog Proteins , Humans , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Protein Domains/genetics , Brachydactyly/genetics , Dwarfism/genetics , Mutation , Animals , Genetic Variation/genetics , Body Height/genetics , Heterozygote
18.
PLoS Genet ; 20(6): e1011326, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38857279

ABSTRACT

The development of ectodermal organs begins with the formation of a stratified epithelial placode that progressively invaginates into the underlying mesenchyme as the organ takes its shape. Signaling by secreted molecules is critical for epithelial morphogenesis, but how that information leads to cell rearrangement and tissue shape changes remains an open question. Using the mouse dentition as a model, we first establish that non-muscle myosin II is essential for dental epithelial invagination and show that it functions by promoting cell-cell adhesion and persistent convergent cell movements in the suprabasal layer. Shh signaling controls these processes by inducing myosin II activation via AKT. Pharmacological induction of AKT and myosin II can also rescue defects caused by the inhibition of Shh. Together, our results support a model in which the Shh signal is transmitted through myosin II to power effective cellular rearrangement for proper dental epithelial invagination.


Subject(s)
Cell Adhesion , Cell Movement , Hedgehog Proteins , Myosin Type II , Signal Transduction , Animals , Mice , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Cell Adhesion/genetics , Myosin Type II/metabolism , Myosin Type II/genetics , Cell Movement/genetics , Epithelium/metabolism , Morphogenesis/genetics , Tooth/metabolism , Tooth/growth & development , Epithelial Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Gene Expression Regulation, Developmental
19.
Neuroreport ; 35(11): 679-686, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38874950

ABSTRACT

Intracerebral hemorrhage (ICH) is a significant public health matter that has no effective treatment. ICH-induced destruction of the blood-brain barrier (BBB) leads to neurological deterioration. Astrocytic sonic hedgehog (SHH) alleviates brain injury by maintaining the integrity of the BBB after ICH. Silent information regulator 1 (SIRT1) is neuroprotective in several central nervous system diseases via BBB regulation. It is also a possible influential factor of the SHH signaling pathway. Nevertheless, the role of SIRT1 on BBB and the underlying pathological process associated with the SHH signaling pathway after ICH remain unclear. We established an intracerebral hemorrhagic mouse model by collagenase injection. SRT1720 (a selective agonist of SIRT1) was used to evaluate the effect of SIRT1 on BBB integrity after ICH. SIRT1 expression was reduced in the mouse brain after ICH. SRT1720 attenuated neurobehavioral impairments and brain edema of ICH mouse. After ICH induction, SRT1720 improved BBB integrity and tight junction expressions in the mouse brain. The SHH signaling pathway-related factors smoothened and glioma-associated oncogene homolog-1 were increased with the intervention of SRT1720, while cyclopamine (a specific inhibitor of the SHH signaling pathway) reversed these effects. These findings suggest that SIRT1 protects from ICH by altering BBB permeability and tight junction expression levels. This process is associated with the SHH signaling pathway, suggesting that SIRT1 may be a potential therapeutic target for ICH.


Subject(s)
Blood-Brain Barrier , Cerebral Hemorrhage , Heterocyclic Compounds, 4 or More Rings , Sirtuin 1 , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Sirtuin 1/metabolism , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/drug therapy , Heterocyclic Compounds, 4 or More Rings/pharmacology , Male , Mice , Disease Models, Animal , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Hedgehog Proteins/metabolism , Hedgehog Proteins/agonists , Brain Edema/drug therapy , Brain Edema/metabolism , Signal Transduction/drug effects
20.
Life Sci ; 351: 122791, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38848936

ABSTRACT

Sorafenib is a multikinase inhibitor employed for managing hepatocellular carcinoma (HCC). The emergence of sorafenib resistance presents an obstacle to its therapeutic efficacy. One notable approach to overcoming sorafenib resistance is the exploration of combination therapies. The role of hedgehog signaling in sorafenib resistance has been also examined in HCC. R51211, known as itraconazole, has been safely employed in clinical practice. Through in vitro and in vivo investigations, we assessed the potential of R51211 to enhance the therapeutic efficacy of sorafenib by inhibiting the hedgehog signaling. The zero-interaction potency synergy model demonstrated a synergistic interaction between R51211 and sorafenib, a phenomenon reversed by the action of a smoothened receptor agonist. This dual therapy exhibited an increased capacity to induce apoptosis, as evidenced by alterations in the Bax/BCL-2 ratio and caspase-3, along with a propensity to promote autophagy, as indicated by changes in BECN1, p62, and the LC3I/LC3II ratio. Furthermore, the combination therapy resulted in significant reductions in biomarkers associated with liver preneoplastic alterations, improved liver microstructure, and mitigated changes in liver function enzymes. The substantial decrease in hedgehog components (Shh, SMO, GLI1, and GLI2) following R51211 treatment appears to be a key factor contributing to the increased efficacy of sorafenib. In conclusion, our study highlights the potential of R51211 as an adjunct to sorafenib, introducing a new dimension to this combination therapy through the modulation of the hedgehog signaling pathway. Further investigations are essential to validate the therapeutic efficacy of this combined approach in inhibiting the development of liver cancer.


Subject(s)
Carcinoma, Hepatocellular , Hedgehog Proteins , Itraconazole , Liver Neoplasms , Signal Transduction , Sorafenib , Sorafenib/pharmacology , Sorafenib/therapeutic use , Hedgehog Proteins/metabolism , Humans , Animals , Signal Transduction/drug effects , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Mice , Itraconazole/pharmacology , Itraconazole/therapeutic use , Apoptosis/drug effects , Male , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Synergism , Cell Line, Tumor , Xenograft Model Antitumor Assays , Drug Resistance, Neoplasm/drug effects , Autophagy/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL