Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.395
Filtrer
1.
Oncol Rep ; 52(3)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38994763

RÉSUMÉ

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.


Sujet(s)
Protéines Hedgehog , Tumeurs , Transduction du signal , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Humains , Tumeurs/génétique , Tumeurs/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Animaux , Mutation
2.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39000154

RÉSUMÉ

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.


Sujet(s)
Facteur de croissance fibroblastique de type 8 , Incisive , Mésoderme , Molaire , Animaux , Facteur de croissance fibroblastique de type 8/génétique , Facteur de croissance fibroblastique de type 8/métabolisme , Souris , Incisive/malformations , Incisive/métabolisme , Mésoderme/métabolisme , Mésoderme/anatomopathologie , Molaire/malformations , Molaire/métabolisme , Anodontie/génétique , Anodontie/métabolisme , Anodontie/anatomopathologie , Apoptose , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Facteur de transcription LEF-1/métabolisme , Facteur de transcription LEF-1/génétique , Sous-unité alpha 1 du facteur CBF/génétique , Sous-unité alpha 1 du facteur CBF/métabolisme , Transduction du signal , Régulation de l'expression des gènes au cours du développement , Odontogenèse/génétique , Souris transgéniques
3.
Transl Vis Sci Technol ; 13(7): 11, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-39007834

RÉSUMÉ

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.


Sujet(s)
Modèles animaux de maladie humaine , Microphtalmie , Protéine à doigts de zinc Gli3 , Animaux , Microphtalmie/génétique , Microphtalmie/anatomopathologie , Microphtalmie/métabolisme , Souris , Protéine à doigts de zinc Gli3/génétique , Protéine à doigts de zinc Gli3/métabolisme , Protéines Hedgehog/génétique , Protéines Hedgehog/métabolisme , Cristallin/métabolisme , Cristallin/anatomopathologie , Transduction du signal , Systèmes CRISPR-Cas , Souris de lignée C57BL , Protéines de tissu nerveux
4.
Stem Cell Res Ther ; 15(1): 198, 2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38971766

RÉSUMÉ

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.


Sujet(s)
Différenciation cellulaire , Cils vibratiles , Sutures crâniennes , Protéines Hedgehog , Ostéogenèse , Transduction du signal , Protéine à doigt de zinc GLI1 , Animaux , Souris , Protéine à doigt de zinc GLI1/métabolisme , Protéine à doigt de zinc GLI1/génétique , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Ostéogenèse/physiologie , Cils vibratiles/métabolisme , Sutures crâniennes/métabolisme , Souris de lignée C57BL , Ostéogenèse par distraction/méthodes , Prolifération cellulaire
5.
Proc Natl Acad Sci U S A ; 121(28): e2320070121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38968120

RÉSUMÉ

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.


Sujet(s)
, Protéines Hedgehog , Protein-Serine-Threonine Kinases , Protein-tyrosine kinases , Transduction du signal , Protéine à doigts de zinc Gli2 , Protéine à doigts de zinc Gli3 , Animaux , Protéine à doigts de zinc Gli3/métabolisme , Protéine à doigts de zinc Gli3/génétique , Protéine à doigts de zinc Gli2/métabolisme , Protéine à doigts de zinc Gli2/génétique , Phosphorylation , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Souris , Protein-tyrosine kinases/métabolisme , Protein-tyrosine kinases/génétique , Humains , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/génétique , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Prolifération cellulaire , Cils vibratiles/métabolisme , Récepteur Smoothened/métabolisme , Récepteur Smoothened/génétique , Protéines nucléaires , Protéines de répression
6.
PLoS Genet ; 20(6): e1011326, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38857279

RÉSUMÉ

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.


Sujet(s)
Adhérence cellulaire , Mouvement cellulaire , Protéines Hedgehog , Myosine de type II , Transduction du signal , Animaux , Souris , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Adhérence cellulaire/génétique , Myosine de type II/métabolisme , Myosine de type II/génétique , Mouvement cellulaire/génétique , Épithélium/métabolisme , Morphogenèse/génétique , Dent/métabolisme , Dent/croissance et développement , Cellules épithéliales/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Régulation de l'expression des gènes au cours du développement
7.
Development ; 151(13)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38828852

RÉSUMÉ

The cellular and genetic networks that contribute to the development of the zeugopod (radius and ulna of the forearm, tibia and fibula of the leg) are not well understood, although these bones are susceptible to loss in congenital human syndromes and to the action of teratogens such as thalidomide. Using a new fate-mapping approach with the Chameleon transgenic chicken line, we show that there is a small contribution of SHH-expressing cells to the posterior ulna, posterior carpals and digit 3. We establish that although the majority of the ulna develops in response to paracrine SHH signalling in both the chicken and mouse, there are differences in the contribution of SHH-expressing cells between mouse and chicken as well as between the chicken ulna and fibula. This is evidence that, although zeugopod bones are clearly homologous according to the fossil record, the gene regulatory networks that contribute to their development and evolution are not fixed.


Sujet(s)
Animal génétiquement modifié , Poulets , Protéines Hedgehog , Animaux , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Poulets/génétique , Souris , Évolution biologique , Embryon de poulet , Ulna , Régulation de l'expression des gènes au cours du développement , Fibula/métabolisme , Radius/métabolisme , Humains , Membres/embryologie
8.
Viruses ; 16(6)2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38932210

RÉSUMÉ

Human cytomegalovirus (CMV) infection is the leading non-genetic cause of congenital malformation in developed countries, causing significant fetal injury, and in some cases fetal death. The pathogenetic mechanisms through which this host-specific virus infects then damages both the placenta and the fetal brain are currently ill-defined. We investigated the CMV modulation of key signaling pathway proteins for these organs including dual-specificity tyrosine phosphorylation-regulated kinases (DYRK) and Sonic Hedgehog (SHH) pathway proteins using human first trimester placental trophoblast (TEV-1) cells, primary human astrocyte (NHA) brain cells, and CMV-infected human placental tissue. Immunofluorescence demonstrated the accumulation and re-localization of SHH proteins in CMV-infected TEV-1 cells with Gli2, Ulk3, and Shh re-localizing to the CMV cytoplasmic virion assembly complex (VAC). In CMV-infected NHA cells, DYRK1A re-localized to the VAC and DYRK1B re-localized to the CMV nuclear replication compartments, and the SHH proteins re-localized with a similar pattern as was observed in TEV-1 cells. Western blot analysis in CMV-infected TEV-1 cells showed the upregulated expression of Rb, Ulk3, and Shh, but not Gli2. In CMV-infected NHA cells, there was an upregulation of DYRK1A, DYRK1B, Gli2, Rb, Ulk3, and Shh. These in vitro monoculture findings are consistent with patterns of protein upregulation and re-localization observed in naturally infected placental tissue and CMV-infected ex vivo placental explant histocultures. This study reveals CMV-induced changes in proteins critical for fetal development, and identifies new potential targets for CMV therapeutic development.


Sujet(s)
Astrocytes , Infections à cytomégalovirus , Cytomegalovirus , Protéines Hedgehog , Placenta , Protein-tyrosine kinases , Transduction du signal , Humains , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Cytomegalovirus/physiologie , Grossesse , Placenta/virologie , Placenta/métabolisme , Astrocytes/virologie , Astrocytes/métabolisme , Femelle , Protein-tyrosine kinases/métabolisme , Protein-tyrosine kinases/génétique , Infections à cytomégalovirus/virologie , Infections à cytomégalovirus/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Phosphorylation , Trophoblastes/virologie , Trophoblastes/métabolisme , , Lignée cellulaire , Cellules cultivées
9.
Neurosurg Rev ; 47(1): 283, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38904885

RÉSUMÉ

This study examined the risk factors for short-term outcomes, focusing particularly on the associations among molecular subgroups. The analysis focused on the data of pediatric patients with medulloblastoma between 2013 and 2023, as well as operative complications, length of stay from surgery to adjuvant treatment, 30-day unplanned reoperation, unplanned readmission, and mortality. 148 patients were included. Patients with the SHH TP53-wildtype exhibited a lower incidence of complications (45.2% vs. 66.0%, odds ratio [OR] 0.358, 95% confidence interval [CI] 0.160 - 0.802). Female sex (0.437, 0.207 - 0.919) was identified as an independent protective factor for complications, and brainstem involvement (1.900, 1.297 - 2.784) was identified as a risk factor. Surgical time was associated with an increased risk of complications (1.004, 1.001 - 1.008), duration of hospitalization (1.006, 1.003 - 1.010), and reoperation (1.003, 1.001 - 1.006). Age was found to be a predictor of improved outcomes, as each additional year was associated with a 14.1% decrease in the likelihood of experiencing a prolonged length of stay (0.859, 0.772 - 0.956). Patients without metastasis exhibited a reduced risk of reoperation (0.322, 0.133 - 0.784) and readmission (0.208, 0.074 - 0.581). There is a significant degree of variability in the occurrence of operative complications in pediatric patients with medulloblastoma. SHH TP53-wildtype medulloblastoma is commonly correlated with a decreased incidence of complications. The short-term outcomes of patients are influenced by various unmodifiable endogenous factors. These findings could enhance the knowledge of onconeurosurgeons and alleviate the challenges associated with patient/parent education through personalized risk communication. However, the importance of a dedicated center with expertise surgical team and experienced neurosurgeon in improving neurosurgical outcomes appears self-evident.


Sujet(s)
Tumeurs du cervelet , Médulloblastome , Procédures de neurochirurgie , Complications postopératoires , Humains , Médulloblastome/chirurgie , Femelle , Mâle , Enfant , Tumeurs du cervelet/chirurgie , Procédures de neurochirurgie/méthodes , Enfant d'âge préscolaire , Complications postopératoires/épidémiologie , Résultat thérapeutique , Adolescent , Études de cohortes , Durée du séjour , Réintervention , Protéines Hedgehog/génétique , Facteurs de risque , Protéine p53 suppresseur de tumeur/génétique
10.
Sci Rep ; 14(1): 14669, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38918575

RÉSUMÉ

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.


Sujet(s)
Facteur de transcription ATF-3 , Diabète de type 1 , Ilots pancréatiques , Souris de lignée NOD , Transduction du signal , Animaux , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Souris , Diabète de type 1/génétique , Diabète de type 1/métabolisme , Diabète de type 1/anatomopathologie , Femelle , Inflammation/génétique , Inflammation/anatomopathologie , Inflammation/métabolisme , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Analyse de profil d'expression de gènes , Modèles animaux de maladie humaine
11.
J Cell Biol ; 223(9)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-38856684

RÉSUMÉ

Sonic Hedgehog (SHH) is a driver of embryonic patterning that, when corrupted, triggers developmental disorders and cancers. SHH effector responses are organized through primary cilia (PC) that grow and retract with the cell cycle and in response to extracellular cues. Disruption of PC homeostasis corrupts SHH regulation, placing significant pressure on the pathway to maintain ciliary fitness. Mechanisms by which ciliary robustness is ensured in SHH-stimulated cells are not yet known. Herein, we reveal a crosstalk circuit induced by SHH activation of Phospholipase A2α that drives ciliary E-type prostanoid receptor 4 (EP4) signaling to ensure PC function and stabilize ciliary length. We demonstrate that blockade of SHH-EP4 crosstalk destabilizes PC cyclic AMP (cAMP) equilibrium, slows ciliary transport, reduces ciliary length, and attenuates SHH pathway induction. Accordingly, Ep4-/- mice display shortened neuroepithelial PC and altered SHH-dependent neuronal cell fate specification. Thus, SHH initiates coordination between distinct ciliary receptors to maintain PC function and length homeostasis for robust downstream signaling.


Sujet(s)
Cils vibratiles , Protéines Hedgehog , Prostaglandines , Transduction du signal , Animaux , Souris , Cils vibratiles/métabolisme , AMP cyclique/métabolisme , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Souris knockout , Prostaglandines/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/génétique
12.
Microb Pathog ; 192: 106723, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38823465

RÉSUMÉ

The Hedgehog (Hh) signaling pathway is involved in T cell differentiation and development and plays a major regulatory part in different stages of T cell development. A previous study by us suggested that prenatal exposure to staphylococcal enterotoxin B (SEB) changed the percentages of T cell subpopulation in the offspring thymus. However, it is unclear whether prenatal SEB exposure impacts the Hh signaling pathway in thymic T cells. In the present study, pregnant rats at gestational day 16 were intravenously injected once with 15 µg SEB, and the thymi of both neonatal and adult offspring rats were aseptically acquired to scrutinize the effects of SEB on the Hh signaling pathway. It firstly found that prenatal SEB exposure clearly caused the increased expression of Shh and Dhh ligands of the Hh signaling pathway in thymus tissue of both neonatal and adult offspring rats, but significantly decreased the expression levels of membrane receptors of Ptch1 and Smo, transcription factor Gli1, as well as target genes of CyclinD1, C-myc, and N-myc in Hh signaling pathway of thymic T cells. These data suggest that prenatal SEB exposure inhibits the Hh signaling pathway in thymic T lymphocytes of the neonatal offspring, and this effect can be maintained in adult offspring via the imprinting effect.


Sujet(s)
Entérotoxines , Protéines Hedgehog , Transduction du signal , Lymphocytes T , Thymus (glande) , Animaux , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Femelle , Grossesse , Rats , Thymus (glande)/métabolisme , Thymus (glande)/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Protéine à doigt de zinc GLI1/métabolisme , Protéine à doigt de zinc GLI1/génétique , Récepteur Patched-1/métabolisme , Récepteur Patched-1/génétique , Récepteur Smoothened/métabolisme , Récepteur Smoothened/génétique , Effets différés de l'exposition prénatale à des facteurs de risque/immunologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Mâle
13.
Mol Biol Rep ; 51(1): 740, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38874802

RÉSUMÉ

BACKGROUND: Sonic Hedgehog (SHH) is a fundamental signaling pathway that controls tissue reconstruction, stem cell biology, and differentiation and has a role in gut tissue homeostasis and development. Dysregulation of SHH leads to the development of HCC. METHODS, AND RESULTS: The present study was conducted to compare the effects of mesenchymal stem cells (MSCs) and curcumin on SHH molecular targets in an experimental model of HCC in rats. One hundred rats were divided equally into the following groups: control group, HCC group, HCC group received MSCs, HCC group received curcumin, and HCC group received MSCs and curcumin. Histopathological examinations were performed, and gene expression of SHH signaling target genes (SHH, PTCH1, SMOH, and GLI1) was assessed by real-time PCR in rat liver tissue. Results showed that SHH target genes were significantly upregulated in HCC-untreated rat groups and in MSC-treated groups, with no significant difference between them. Administration of curcumin with or without combined administration of MSCs led to a significant down-regulation of SHH target genes, with no significant differences between both groups. As regards the histopathological examination of liver tissues, both curcumin and MSCs, either through separate use or their combined use, led to a significant restoration of normal liver pathology. CONCLUSIONS: In conclusion, SHH signaling is upregulated in the HCC experimental model. MSCs do not inhibit the upregulated SHH target genes in HCC. Curcumin use with or without MSCs administration led to a significant down-regulation of SHH signaling in HCC and a significant restoration of normal liver pathology.


Sujet(s)
Carcinome hépatocellulaire , Curcumine , Protéines Hedgehog , Tumeurs du foie , Cellules souches mésenchymateuses , Transduction du signal , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Animaux , Curcumine/pharmacologie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Rats , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Transplantation de cellules souches mésenchymateuses/méthodes , Mâle , Modèles animaux de maladie humaine , Récepteur Patched-1/génétique , Récepteur Patched-1/métabolisme , Protéine à doigt de zinc GLI1/métabolisme , Protéine à doigt de zinc GLI1/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Foie/effets des médicaments et des substances chimiques
14.
Sci Rep ; 14(1): 13596, 2024 06 12.
Article de Anglais | MEDLINE | ID: mdl-38866867

RÉSUMÉ

The RE1 silencing transcription factor (REST) is a driver of sonic hedgehog (SHH) medulloblastoma genesis. Our previous studies showed that REST enhances cell proliferation, metastasis and vascular growth and blocks neuronal differentiation to drive progression of SHH medulloblastoma tumors. Here, we demonstrate that REST promotes autophagy, a pathway that is found to be significantly enriched in human medulloblastoma tumors relative to normal cerebella. In SHH medulloblastoma tumor xenografts, REST elevation is strongly correlated with increased expression of the hypoxia-inducible factor 1-alpha (HIF1α)-a positive regulator of autophagy, and with reduced expression of the von Hippel-Lindau (VHL) tumor suppressor protein - a component of an E3 ligase complex that ubiquitinates HIF1α. Human SHH-medulloblastoma tumors with higher REST expression exhibit nuclear localization of HIF1α, in contrast to its cytoplasmic localization in low-REST tumors. In vitro, REST knockdown promotes an increase in VHL levels and a decrease in cytoplasmic HIF1α protein levels, and autophagy flux. In contrast, REST elevation causes a decline in VHL levels, as well as its interaction with HIF1α, resulting in a reduction in HIF1α ubiquitination and an increase in autophagy flux. These data suggest that REST elevation promotes autophagy in SHH medulloblastoma cells by modulating HIF1α ubiquitination and stability in a VHL-dependent manner. Thus, our study is one of the first to connect VHL to REST-dependent control of autophagy in a subset of medulloblastomas.


Sujet(s)
Autophagie , Tumeurs du cervelet , Protéines Hedgehog , Sous-unité alpha du facteur-1 induit par l'hypoxie , Médulloblastome , Protéine Von Hippel-Lindau supresseur de tumeur , Médulloblastome/métabolisme , Médulloblastome/anatomopathologie , Médulloblastome/génétique , Humains , Protéine Von Hippel-Lindau supresseur de tumeur/métabolisme , Protéine Von Hippel-Lindau supresseur de tumeur/génétique , Autophagie/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Animaux , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Lignée cellulaire tumorale , Tumeurs du cervelet/métabolisme , Tumeurs du cervelet/anatomopathologie , Tumeurs du cervelet/génétique , Souris , Régulation négative , Régulation de l'expression des gènes tumoraux , Ubiquitination , Protéines de répression
15.
Int J Mol Sci ; 25(11)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38891790

RÉSUMÉ

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.


Sujet(s)
Protéines Hedgehog , Côtes , Rachis , Animaux , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Côtes/métabolisme , Côtes/embryologie , Rachis/métabolisme , Rachis/embryologie , Régulation de l'expression des gènes au cours du développement , Mésoderme/métabolisme , Mésoderme/embryologie , Caille , Somites/métabolisme , Somites/embryologie , Protéines morphogénétiques osseuses/métabolisme , Protéines morphogénétiques osseuses/génétique , Protéines de transport
16.
PLoS One ; 19(6): e0294835, 2024.
Article de Anglais | MEDLINE | ID: mdl-38848388

RÉSUMÉ

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.


Sujet(s)
Protéines Hedgehog , Transduction du signal , Calicules gustatifs , Langue , Animaux , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Langue/métabolisme , Langue/croissance et développement , Souris , Calicules gustatifs/métabolisme , Calicules gustatifs/croissance et développement , Régulation de l'expression des gènes au cours du développement , Homéostasie , Récepteur Patched-1/métabolisme , Récepteur Patched-1/génétique , Protéine à doigt de zinc GLI1/métabolisme , Protéine à doigt de zinc GLI1/génétique , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Protéine à doigts de zinc Gli2/métabolisme , Protéine à doigts de zinc Gli2/génétique , Protéine à doigts de zinc Gli3/métabolisme , Protéine à doigts de zinc Gli3/génétique , Protéines de tissu nerveux , Protéines du cycle cellulaire , Protéines liées au GPI
17.
Development ; 151(14)2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38940473

RÉSUMÉ

The direction of left-right visceral asymmetry is conserved in vertebrates. Deviations of the standard asymmetric pattern are rare, and the underlying mechanisms are not understood. Here, we use the teleost Astyanax mexicanus, consisting of surface fish with normal left-oriented heart asymmetry and cavefish with high levels of reversed right-oriented heart asymmetry, to explore natural changes in asymmetry determination. We show that Sonic Hedgehog (Shh) signaling is increased at the posterior midline, Kupffer's vesicle (the teleost left-right organizer) is enlarged and contains longer cilia, and the number of dorsal forerunner cells is increased in cavefish. Furthermore, Shh increase in surface fish embryos induces asymmetric changes resembling the cavefish phenotype. Asymmetric expression of the Nodal antagonist Dand5 is equalized or reversed in cavefish, and Shh increase in surface fish mimics changes in cavefish dand5 asymmetry. Shh decrease reduces the level of right-oriented heart asymmetry in cavefish. Thus, naturally occurring modifications in cavefish heart asymmetry are controlled by the effects of Shh signaling on left-right organizer function.


Sujet(s)
Plan d'organisation du corps , Coeur , Protéines Hedgehog , Transduction du signal , Animaux , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Plan d'organisation du corps/génétique , Coeur/embryologie , Characidae/embryologie , Characidae/génétique , Régulation de l'expression des gènes au cours du développement , Cils vibratiles/métabolisme , Embryon non mammalien/métabolisme
18.
Eur J Endocrinol ; 191(1): 38-46, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38917024

RÉSUMÉ

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.


Sujet(s)
Protéines Hedgehog , Humains , Protéines Hedgehog/génétique , Protéines Hedgehog/métabolisme , Domaines protéiques/génétique , Brachydactylie/génétique , Nanisme/génétique , Mutation , Animaux , Variation génétique/génétique , Taille/génétique , Hétérozygote
19.
PLoS One ; 19(6): e0301670, 2024.
Article de Anglais | MEDLINE | ID: mdl-38917070

RÉSUMÉ

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.


Sujet(s)
Protéines Hedgehog , Organoïdes , Transduction du signal , Moelle spinale , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Animaux , Organoïdes/métabolisme , Organoïdes/cytologie , Moelle spinale/métabolisme , Moelle spinale/cytologie , Souris , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Récepteur Patched-1/métabolisme , Récepteur Patched-1/génétique
20.
Genesis ; 62(3): e23602, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38721990

RÉSUMÉ

Cilia play a key role in the regulation of signaling pathways required for embryonic development, including the proper formation of the neural tube, the precursor to the brain and spinal cord. Forward genetic screens were used to generate mouse lines that display neural tube defects (NTD) and secondary phenotypes useful in interrogating function. We describe here the L3P mutant line that displays phenotypes of disrupted Sonic hedgehog signaling and affects the initiation of cilia formation. A point mutation was mapped in the L3P line to the gene Rsg1, which encodes a GTPase-like protein. The mutation lies within the GTP-binding pocket and disrupts the highly conserved G1 domain. The mutant protein and other centrosomal and IFT proteins still localize appropriately to the basal body of cilia, suggesting that RSG1 GTPase activity is not required for basal body maturation but is needed for a downstream step in axonemal elongation.


Sujet(s)
Cils vibratiles , Tube neural , Animaux , Souris , Cils vibratiles/métabolisme , Cils vibratiles/génétique , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Tube neural/embryologie , Tube neural/métabolisme , Anomalies du tube neural/génétique , Anomalies du tube neural/métabolisme , Mutation ponctuelle , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...