Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.368
Filter
1.
Life Sci Alliance ; 7(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38719753

ABSTRACT

We recently reported that growth/differentiation factor 15 (GDF15) and its receptor GDNF family receptor alpha-like (GFRAL) are expressed in the periventricular germinal epithelium thereby regulating apical progenitor proliferation. However, the mechanisms are unknown. We now found GFRAL in primary cilia and altered cilia morphology upon GDF15 ablation. Mutant progenitors also displayed increased histone deacetylase 6 (Hdac6) and ciliary adenylate cyclase 3 (Adcy3) transcript levels. Consistently, microtubule acetylation, endogenous sonic hedgehog (SHH) activation and ciliary ADCY3 were all affected in this group. Application of exogenous GDF15 or pharmacological antagonists of either HDAC6 or ADCY3 similarly normalized ciliary morphology, proliferation and SHH signalling. Notably, Gdf15 ablation affected Hdac6 expression and cilia length only in the mutant periventricular niche, in concomitance with ciliary localization of GFRAL. In contrast, in the hippocampus, where GFRAL was not expressed in the cilium, progenitors displayed altered Adcy3 expression and SHH signalling, but Hdac6 expression, cilia morphology and ciliary ADCY3 levels remained unchanged. Thus, ciliary signalling underlies the effect of GDF15 on primary cilia elongation and proliferation in apical progenitors.


Subject(s)
Adenylyl Cyclases , Cell Proliferation , Cilia , Hedgehog Proteins , Histone Deacetylase 6 , Signal Transduction , Animals , Mice , Acetylation , Adenylyl Cyclases/metabolism , Adenylyl Cyclases/genetics , Cell Proliferation/genetics , Cilia/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Histone Deacetylase 6/metabolism , Histone Deacetylase 6/genetics , Mice, Knockout , Stem Cells/metabolism , Stem Cells/cytology
2.
Genesis ; 62(3): e23602, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38721990

ABSTRACT

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.


Subject(s)
Cilia , Neural Tube Defects , Neural Tube , Animals , Cilia/metabolism , Cilia/genetics , Mice , Neural Tube/metabolism , Neural Tube/embryology , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Signal Transduction , Point Mutation
3.
Dev Biol ; 512: 57-69, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38750688

ABSTRACT

Understanding the developmental processes and signaling pathways involved in larval myogenesis and metamorphosis is crucial for comprehending the life history and adaptive strategies of marine organisms. In this study, we investigated the temporal and spatial patterns of myogenesis in the mussel Mytilus coruscus (Mc), focusing on the emergence and transformation of major muscle groups during different larval stages. We also explored the role of the Hedgehog (Hh) signaling pathway in regulating myogenesis and larval metamorphosis. The results revealed distinct developmental stages characterized by the emergence of specific muscular components, such as velum retractor muscles and anterior adductor muscles, in D-veliger and umbo larvae, which are responsible for the planktonic stage. In the pediveliger stage, posterior ventral, posterior adductor, and foot muscles appeared. After larval metamorphosis, the velum structure and its corresponding retractor muscles degenerate, indicating the transition from planktonic to benthic life. We observed a conserved pattern of larval musculature development and revealed a high degree of conservation across bivalve species, with comparable emergence times during myogenesis. Furthermore, exposure to the Hh signaling inhibitor cyclopamine impaired larval muscle development, reduced larval swimming activity, and inhibited larval metamorphosis in M. coruscus. Cyclopamine-mediated inhibition of Hh signaling led to reduced expression of four key genes within the Hh signaling pathway (McHh, McPtc, McSmo, and McGli) and the striated myosin heavy chain gene (McMHC). It is hypothesised that the abnormal larval muscle development in cyclopamine-treated groups may be an indirect effect due to disrupted McMHC expression. We provide evidence for the first time that cyclopamine treatment inhibited larval metamorphosis in bivalves, highlighting the potential involvement of Hh signaling in mediating larval muscle development and metamorphosis in M. coruscus. The present study provides insights into the dynamic nature of myogenesis and the regulatory role of the Hh signaling pathway during larval development and metamorphosis in M. coruscus. The results obtained in this study contribute to a better understanding of the evolutionary significance of Hh signaling in bivalves and shed light on the mechanisms underlying larval muscle development and metamorphosis in marine invertebrates.


Subject(s)
Gene Expression Regulation, Developmental , Hedgehog Proteins , Larva , Metamorphosis, Biological , Muscle Development , Mytilus , Signal Transduction , Animals , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Larva/growth & development , Larva/metabolism , Mytilus/growth & development , Mytilus/metabolism , Veratrum Alkaloids/pharmacology , Muscles/metabolism
4.
Int J Mol Sci ; 25(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731849

ABSTRACT

Tumors of the head and neck, more specifically the squamous cell carcinoma, often show upregulation of the Hedgehog signaling pathway. However, almost nothing is known about its role in the sinonasal adenocarcinoma, either in intestinal or non-intestinal subtypes. In this work, we have analyzed immunohistochemical staining of six Hedgehog pathway proteins, sonic Hedgehog (SHH), Indian Hedgehog (IHH), Patched1 (PTCH1), Gli family zinc finger 1 (GLI1), Gli family zinc finger 2 (GLI2), and Gli family zinc finger 3 (GLI3), on 21 samples of sinonasal adenocarcinoma and compared them with six colon adenocarcinoma and three salivary gland tumors, as well as with matching healthy tissue, where available. We have detected GLI2 and PTCH1 in the majority of samples and also GLI1 in a subset of samples, while GLI3 and the ligands SHH and IHH were generally not detected. PTCH1 pattern of staining shows an interesting pattern, where healthy samples are mostly positive in the stromal compartment, while the signal shifts to the tumor compartment in tumors. This, taken together with a stronger signal of GLI2 in tumors compared to non-tumor tissues, suggests that the Hedgehog pathway is indeed activated in sinonasal adenocarcinoma. As Hedgehog pathway inhibitors are being tested in combination with other therapies for head and neck squamous cell carcinoma, this could provide a therapeutic option for patients with sinonasal adenocarcinoma as well.


Subject(s)
Adenocarcinoma , Hedgehog Proteins , Immunohistochemistry , Signal Transduction , Zinc Finger Protein Gli2 , Humans , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Male , Female , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli2/genetics , Middle Aged , Pilot Projects , Aged , Patched-1 Receptor/metabolism , Patched-1 Receptor/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Paranasal Sinus Neoplasms/metabolism , Paranasal Sinus Neoplasms/pathology , Adult , Gene Expression Regulation, Neoplastic , Nerve Tissue Proteins , Nuclear Proteins
5.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 190-197, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38814217

ABSTRACT

Circular RNA hsa_circ_0001322 (circ1322) was demonstrated to be significantly reduced in expression in gastric cancer patients in our previous study, and changes in its expression were significantly correlated with lymph node metastasis. However, the underlying workings of circ1322 in gastric cancer are still not fully understood. Therefore, to confirm the effect of circ1322 on gastric cancer, we examined the expression of circ1322 in gastric cancer cells and tissues. The results showed that circ1322 was lowly expressed in GC tissues and cells. Subsequently, we further performed cellular assays and animal experiments, which showed that Circ1322 upregulation inhibited GC cell proliferation, migration and invasion. while promoting GC cell apoptosis, and inhibited tumor growth in mice. The direct targeting of circ1322 to miR-1264 was confirmed by bioinformatics prediction and validation of luciferase reporter gene assay. Circ1322 can act as a miR-1264 sponge to alleviate the inhibitory effect of miR-1264 on its target gene, QKI. miR-1264 regulates the expression of QKI and the activity of the hedgehog pathway. That is, circ1322 may act as a competing endogenous RNA (ceRNA) to inhibit the hedgehog pathway by targeting the miR-1264/QKI axis, which in turn promotes GC progression.


Subject(s)
Cell Movement , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Hedgehog Proteins , MicroRNAs , RNA, Circular , Signal Transduction , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Signal Transduction/genetics , Cell Movement/genetics , Mice, Nude , Apoptosis/genetics , Mice , Male , Mice, Inbred BALB C , Female , Base Sequence , Middle Aged
6.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 238-242, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38814213

ABSTRACT

Patients with sepsis are often complicated by acute kidney injury (AKI), which greatly increases mortality. In this study, our purpose was to explore the expression and function of CDGSH iron sulfur domain 2 (CISD2) in septic AKI, and the underlying molecular mechanism. Western blot and quantitative real-time polymerase chain reaction (RT-PCR) were employed to detect protein and mRNA levels in cells. The inflammation level of cells was evaluated by detecting the content of inflammatory factors (TNF-α, IL-1ß, IL-6). Apoptosis of cells was evaluated by Caspase-3 activity assay, flow cytometry and terminal deoxynucleotidyl transferase-mediated dUTP Nick-End Labeling (TUNEL) staining. CISD2 was down-regulated in HK-2 cells treated with lipopolysaccharide (LPS). LPS treatment increased the level of inflammatory factors, the activity of Caspase-3, and the rate of apoptosis in HK-2 cells. However, overexpression of CISD2 significantly suppressed these effects. Moreover, overexpression of CISD2 activated the Sonic Hedgehog (SHH) signaling pathway. The use of cyclopamine (Cyc), a SHH signaling pathway inhibitor, eliminated the effect of overexpressing CISD2, that is, inhibiting LPS-induced inflammation and apoptosis of HK-2 cells. LPS treatment down-regulated CISD2 in HK-2 cells, and overexpression of CISD2 could inhibit LPS-induced inflammation and apoptosis of HK-2 cells by activating the SHH signaling pathway.


Subject(s)
Acute Kidney Injury , Apoptosis , Hedgehog Proteins , Lipopolysaccharides , Sepsis , Signal Transduction , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Humans , Acute Kidney Injury/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Signal Transduction/drug effects , Apoptosis/drug effects , Sepsis/metabolism , Sepsis/complications , Cell Line , Caspase 3/metabolism , Caspase 3/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics
7.
Neuropathol Appl Neurobiol ; 50(3): e12983, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38708554

ABSTRACT

We describe a 46-year-old patient with an IDH-wildtype diffusely infiltrating atypical teratoid/rhabdoid tumour (AT/RT), SHH-1B molecular subtype. The unusual histology and subsequent diagnosis in an adult patient will be discussed.


Subject(s)
Brain Neoplasms , Rhabdoid Tumor , Teratoma , Humans , Rhabdoid Tumor/pathology , Rhabdoid Tumor/genetics , Teratoma/pathology , Teratoma/genetics , Middle Aged , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Male , Hedgehog Proteins/genetics
8.
Proc Natl Acad Sci U S A ; 121(20): e2321711121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38713624

ABSTRACT

During development, neural stem cells in the cerebral cortex, also known as radial glial cells (RGCs), generate excitatory neurons, followed by production of cortical macroglia and inhibitory neurons that migrate to the olfactory bulb (OB). Understanding the mechanisms for this lineage switch is fundamental for unraveling how proper numbers of diverse neuronal and glial cell types are controlled. We and others recently showed that Sonic Hedgehog (Shh) signaling promotes the cortical RGC lineage switch to generate cortical oligodendrocytes and OB interneurons. During this process, cortical RGCs generate intermediate progenitor cells that express critical gliogenesis genes Ascl1, Egfr, and Olig2. The increased Ascl1 expression and appearance of Egfr+ and Olig2+ cortical progenitors are concurrent with the switch from excitatory neurogenesis to gliogenesis and OB interneuron neurogenesis in the cortex. While Shh signaling promotes Olig2 expression in the developing spinal cord, the exact mechanism for this transcriptional regulation is not known. Furthermore, the transcriptional regulation of Olig2 and Egfr has not been explored. Here, we show that in cortical progenitor cells, multiple regulatory programs, including Pax6 and Gli3, prevent precocious expression of Olig2, a gene essential for production of cortical oligodendrocytes and astrocytes. We identify multiple enhancers that control Olig2 expression in cortical progenitors and show that the mechanisms for regulating Olig2 expression are conserved between the mouse and human. Our study reveals evolutionarily conserved regulatory logic controlling the lineage switch of cortical neural stem cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Cerebral Cortex , ErbB Receptors , Hedgehog Proteins , Nerve Tissue Proteins , Neural Stem Cells , Neurogenesis , Oligodendrocyte Transcription Factor 2 , PAX6 Transcription Factor , Animals , Neurogenesis/physiology , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , ErbB Receptors/metabolism , ErbB Receptors/genetics , Mice , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendrocyte Transcription Factor 2/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , PAX6 Transcription Factor/metabolism , PAX6 Transcription Factor/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Eye Proteins/metabolism , Eye Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Paired Box Transcription Factors/metabolism , Paired Box Transcription Factors/genetics , Neuroglia/metabolism , Neuroglia/cytology , Gene Expression Regulation, Developmental , Signal Transduction , Olfactory Bulb/metabolism , Olfactory Bulb/cytology , Cell Lineage , Humans
9.
Clin Exp Dent Res ; 10(2): e861, 2024 04.
Article in English | MEDLINE | ID: mdl-38558491

ABSTRACT

OBJECTIVES: The main objective of this study was to evaluate how an apparently minor anomaly of the sphenoid bone, observed in a haploinsufficient mouse model for Sonic Hedgehog (Shh), affects the growth of the adult craniofacial region. This study aims to provide valuable information to orthodontists when making decisions regarding individuals carrying SHH mutation. MATERIALS AND METHODS: The skulls of embryonic, juvenile and adult mice of two genotypes (Shh heterozygous and wild type) were examined and measured using landmark-based linear dimensions. Additionally, we analysed the clinical characteristics of a group of patients and their relatives with SHH gene mutations. RESULTS: In the viable Shh+/ - mouse model, bred on a C57BL/6J background, we noted the presence of a persistent foramen at the midline of the basisphenoid bone. This particular anomaly was attributed to the existence of an ectopic pituitary gland. We discovered that this anomaly led to premature closure of the intrasphenoidal synchondrosis and contributed to craniofacial deformities in adult mice, including a longitudinally shortened skull base. This developmental anomaly is reminiscent of that commonly observed in human holoprosencephaly, a disorder resulting from a deficiency in SHH activity. However, sphenoid morphogenesis is not currently monitored in individuals carrying SHH mutations. CONCLUSION: Haploinsufficiency of Shh leads to isolated craniofacial skeletal hypoplasia in adult mouse. This finding highlights the importance of radiographic monitoring of the skull base in all individuals with SHH gene mutations.


Subject(s)
Hedgehog Proteins , Holoprosencephaly , Adult , Animals , Humans , Mice , Hedgehog Proteins/genetics , Holoprosencephaly/genetics , Mice, Inbred C57BL , Mutation , Sphenoid Bone
10.
Bone Res ; 12(1): 21, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561387

ABSTRACT

Syndactyly type V (SDTY5) is an autosomal dominant extremity malformation characterized by fusion of the fourth and fifth metacarpals. In the previous publication, we first identified a heterozygous missense mutation Q50R in homeobox domain (HD) of HOXD13 in a large Chinese family with SDTY5. In order to substantiate the pathogenicity of the variant and elucidate the underlying pathogenic mechanism causing limb malformation, transcription-activator-like effector nucleases (TALEN) was employed to generate a Hoxd13Q50R mutant mouse. The mutant mice exhibited obvious limb malformations including slight brachydactyly and partial syndactyly between digits 2-4 in the heterozygotes, and severe syndactyly, brachydactyly and polydactyly in homozygotes. Focusing on BMP2 and SHH/GREM1/AER-FGF epithelial mesenchymal (e-m) feedback, a crucial signal pathway for limb development, we found the ectopically expressed Shh, Grem1 and Fgf8 and down-regulated Bmp2 in the embryonic limb bud at E10.5 to E12.5. A transcriptome sequencing analysis was conducted on limb buds (LBs) at E11.5, revealing 31 genes that exhibited notable disparities in mRNA level between the Hoxd13Q50R homozygotes and the wild-type. These genes are known to be involved in various processes such as limb development, cell proliferation, migration, and apoptosis. Our findings indicate that the ectopic expression of Shh and Fgf8, in conjunction with the down-regulation of Bmp2, results in a failure of patterning along both the anterior-posterior and proximal-distal axes, as well as a decrease in interdigital programmed cell death (PCD). This cascade ultimately leads to the development of syndactyly and brachydactyly in heterozygous mice, and severe limb malformations in homozygous mice. These findings suggest that abnormal expression of SHH, FGF8, and BMP2 induced by HOXD13Q50R may be responsible for the manifestation of human SDTY5.


Subject(s)
Brachydactyly , Limb Deformities, Congenital , Syndactyly , Mice , Humans , Animals , Hedgehog Proteins/genetics , Transcription Factors/genetics , Syndactyly/genetics
11.
Cells ; 13(7)2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38607081

ABSTRACT

Increased activation of ovarian primordial follicles in Erß knockout (ErßKO) rats becomes evident as early as postnatal day 8.5. To identify the ERß-regulated genes that may control ovarian primordial follicle activation, we analyzed the transcriptome profiles of ErßKO rat ovaries collected on postnatal days 4.5, 6.5, and 8.5. Compared to wildtype ovaries, ErßKO ovaries displayed dramatic downregulation of Indian hedgehog (Ihh) expression. IHH-regulated genes, including Hhip, Gli1, and Ptch1, were also downregulated in ErßKO ovaries. This was associated with a downregulation of steroidogenic enzymes Cyp11a1, Cyp19a1, and Hsd17b1. The expression of Ihh remained very low in ErßKO ovaries despite the high levels of Gdf9 and Bmp15, which are known upregulators of Ihh expression in the granulosa cells of activated ovarian follicles. Strikingly, the downregulation of the Ihh gene in ErßKO ovaries began to disappear on postnatal day 16.5 and recovered on postnatal day 21.5. In rat ovaries, the first wave of primordial follicles is rapidly activated after their formation, whereas the second wave of primordial follicles remains dormant in the ovarian cortex and slowly starts activating after postnatal day 12.5. We localized the expression of Ihh mRNA in postnatal day 8.5 wildtype rat ovaries but not in the age-matched ErßKO ovaries. In postnatal day 21.5 ErßKO rat ovaries, we detected Ihh mRNA mainly in the activated follicles in the ovaries' peripheral regions. Our findings indicate that the expression of Ihh in the granulosa cells of the activated first wave of ovarian follicles depends on ERß.


Subject(s)
Estrogen Receptor beta , Hedgehog Proteins , Animals , Female , Rats , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Ovarian Follicle/metabolism , Ovary/metabolism , RNA, Messenger/metabolism
12.
Cell Rep ; 43(4): 114083, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38602877

ABSTRACT

A common cause of deafness in humans is dysregulation of the endocochlear potential generated by the stria vascularis (SV). Thus, proper formation of the SV is critical for hearing. Using single-cell transcriptomics and a series of Shh signaling mutants, we discovered that the Shh receptor Patched1 (Ptch1) is essential for marginal cell (MC) differentiation and SV formation. Single-cell RNA sequencing analyses revealed that the cochlear roof epithelium is already specified into discrete domains with distinctive gene expression profiles at embryonic day 14, with Gsc as a marker gene of the MC lineage. Ptch1 deficiency leads to defective specification of MC precursors along the cochlear basal-apical regions. We demonstrated that elevated Gli2 levels impede MC differentiation through sustaining Otx2 expression and maintaining the progenitor state of MC precursors. Our results uncover an early specification of cochlear non-sensory epithelial cells and establish a crucial role of the Ptch1-Gli2 axis in regulating the development of SV.


Subject(s)
Cell Differentiation , Cochlea , Patched-1 Receptor , Stria Vascularis , Patched-1 Receptor/metabolism , Patched-1 Receptor/genetics , Animals , Mice , Stria Vascularis/metabolism , Stria Vascularis/cytology , Cochlea/metabolism , Cochlea/embryology , Cochlea/cytology , Signal Transduction , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli2/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics
13.
Sci Adv ; 10(17): eade1650, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38669326

ABSTRACT

While the kinesin-2 motors KIF3A and KIF3B have essential roles in ciliogenesis and Hedgehog (HH) signal transduction, potential role(s) for another kinesin-2 motor, KIF17, in HH signaling have yet to be explored. Here, we investigated the contribution of KIF17 to HH-dependent cerebellar development, where Kif17 is expressed in both HH-producing Purkinje cells and HH-responding cerebellar granule neuron progenitors (CGNPs). Germline Kif17 deletion in mice results in cerebellar hypoplasia due to reduced CGNP proliferation, a consequence of decreased HH pathway activity mediated through decreased Sonic HH (SHH) protein. Notably, Purkinje cell-specific Kif17 deletion partially phenocopies Kif17 germline mutants. Unexpectedly, CGNP-specific Kif17 deletion results in the opposite phenotype-increased CGNP proliferation and HH target gene expression due to altered GLI transcription factor processing. Together, these data identify KIF17 as a key regulator of HH-dependent cerebellar development, with dual and opposing roles in HH-producing Purkinje cells and HH-responding CGNPs.


Subject(s)
Cerebellum , Cerebellum/abnormalities , Hedgehog Proteins , Kinesins , Nervous System Malformations , Purkinje Cells , Animals , Kinesins/metabolism , Kinesins/genetics , Cerebellum/metabolism , Cerebellum/growth & development , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Mice , Purkinje Cells/metabolism , Signal Transduction , Cell Proliferation , Mice, Knockout , Gene Expression Regulation, Developmental , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Developmental Disabilities
14.
J Exp Clin Cancer Res ; 43(1): 130, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689348

ABSTRACT

BACKGROUND: Medulloblastomas (MBs) are one of the most common malignant brain tumor types in children. MB prognosis, despite improvement in recent years, still depends on clinical and biological risk factors. Metastasis is the leading cause of MB-related deaths, which highlights an unmet need for risk stratification and targeted therapy to improve clinical outcomes. Among the four molecular subgroups, sonic-hedgehog (SHH)-MB harbors clinical and genetic heterogeneity with a subset of high-risk cases. Recently, long non-coding (lnc)RNAs were implied to contribute to cancer malignant progression, but their role in MB remains unclear. This study aimed to identify pro-malignant lncRNAs that have prognostic and therapeutic significance in SHH-MB. METHODS: The Daoy SHH-MB cell line was engineered for ectopic expression of MYCN, a genetic signature of SHH-MB. MYCN-associated lncRNA genes were identified using RNA-sequencing data and were validated in SHH-MB cell lines, MB tissue samples, and patient cohort datasets. SHH-MB cells with genetic manipulation of the candidate lncRNA were evaluated for metastatic phenotypes in vitro, including cell migration, invasion, sphere formation, and expressions of stemness markers. An orthotopic xenograft mouse model was used to evaluate metastasis occurrence and survival. Finally, bioinformatic screening and in vitro assays were performed to explore downstream mechanisms. RESULTS: Elevated lncRNA LOXL1-AS1 expression was identified in MYCN-expressing Daoy cells and MYCN-amplified SHH-MB tumors, and was significantly associated with lower survival in SHH-MB patients. Functionally, LOXL1-AS1 promoted SHH-MB cell migration and cancer stemness in vitro. In mice, MYCN-expressing Daoy cells exhibited a high metastatic rate and adverse effects on survival, both of which were suppressed under LOLX1-AS1 perturbation. Integrative bioinformatic analyses revealed associations of LOXL1-AS1 with processes of cancer stemness, cell differentiation, and the epithelial-mesenchymal transition. LOXL1-AS1 positively regulated the expression of transforming growth factor (TGF)-ß2. Knockdown of TGF-ß2 in SHH-MB cells significantly abrogated their LOXL1-AS1-mediated prometastatic functions. CONCLUSIONS: This study proved the functional significance of LOXL1-AS1 in SHH-MB metastasis by its promotion of TGF-ß2-mediated cancer stem-like phenotypes, providing both prognostic and therapeutic potentials for targeting SHH-MB metastasis.


Subject(s)
Hedgehog Proteins , Medulloblastoma , Neoplastic Stem Cells , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , Medulloblastoma/metabolism , Animals , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Neoplasm Metastasis , Phenotype , Female , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cell Line, Tumor , Male , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/metabolism , Prognosis , Cell Movement
15.
Stem Cell Res Ther ; 15(1): 98, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38581019

ABSTRACT

BACKGROUND: In vitro chondrogenesis of mesenchymal stromal cells (MSCs) driven by the essential chondro-inducer transforming growth factor (TGF)-ß is instable and yields undesired hypertrophic cartilage predisposed to bone formation in vivo. TGF-ß can non-canonically activate bone morphogenetic protein-associated ALK1/2/3 receptors. These have been accused of driving hypertrophic MSC misdifferentiation, but data remained conflicting. We here tested the antihypertrophic capacity of two highly specific ALK1/2/3 inhibitors - compound A (CompA) and LDN-212854 (LDN21) - in order to reveal potential prohypertrophic contributions of these BMP/non-canonical TGF-ß receptors during MSC in vitro chondrogenesis. METHODS: Standard chondrogenic pellet cultures of human bone marrow-derived MSCs were treated with TGF-ß and CompA (500 nM) or LDN21 (500 nM). Daily 6-hour pulses of parathyroid hormone-related peptide (PTHrP[1-34], 2.5 nM, from day 7) served as potent antihypertrophic control treatment. Day 28 samples were subcutaneously implanted into immunodeficient mice. RESULTS: All groups underwent strong chondrogenesis, but GAG/DNA deposition and ACAN expression were slightly but significantly reduced by ALK inhibition compared to solvent controls along with a mild decrease of the hypertrophy markers IHH-, SPP1-mRNA, and Alkaline phosphatase (ALP) activity. When corrected for the degree of chondrogenesis (COL2A1 expression), only pulsed PTHrP but not ALK1/2/3 inhibition qualified as antihypertrophic treatment. In vivo, all subcutaneous cartilaginous implants mineralized within 8 weeks, but PTHrP pretreated samples formed less bone and attracted significantly less haematopoietic marrow than ALK1/2/3 inhibitor groups. CONCLUSIONS: Overall, our data show that BMP-ALK1/2/3 inhibition cannot program mesenchymal stromal cells toward stable chondrogenesis. BMP-ALK1/2/3 signalling is no driver of hypertrophic MSC misdifferentiation and BMP receptor induction is not an adverse prohypertrophic side effect of TGF-ß that leads to endochondral MSC misdifferentiation. Instead, the prohypertrophic network comprises misregulated PTHrP/hedgehog signalling and WNT activity, and a potential contribution of TGF-ß-ALK4/5-mediated SMAD1/5/9 signalling should be further investigated to decide about its postulated prohypertrophic activity. This will help to successfully engineer cartilage replacement tissues from MSCs in vitro and translate these into clinical cartilage regenerative therapies.


Subject(s)
Mesenchymal Stem Cells , Parathyroid Hormone-Related Protein , Animals , Humans , Mice , Cells, Cultured , Chondrocytes/metabolism , Chondrogenesis , Hedgehog Proteins/genetics , Hypertrophy/metabolism , Mesenchymal Stem Cells/metabolism , Parathyroid Hormone-Related Protein/pharmacology , Transforming Growth Factor beta/metabolism
16.
Genes Chromosomes Cancer ; 63(4): e23233, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38607297

ABSTRACT

Medulloblastomas, the most common malignant pediatric brain tumors, can be classified into the wingless, sonic hedgehog (SHH), group 3, and group 4 subgroups. Among them, the SHH subgroup with the TP53 mutation and group 3 generally present with the worst patient outcomes due to their high rates of recurrence and metastasis. A novel and effective treatment for refractory medulloblastomas is urgently needed. To date, the tumor microenvironment (TME) has been shown to influence tumor growth, recurrence, and metastasis through immunosuppression, angiogenesis, and chronic inflammation. Treatments targeting TME components have emerged as promising approaches to the treatment of solid tumors. In this review, we summarize progress in research on medulloblastoma microenvironment components and their interactions. We also discuss challenges and future research directions for TME-targeting medulloblastoma therapy.


Subject(s)
Brain Neoplasms , Cerebellar Neoplasms , Medulloblastoma , Humans , Child , Hedgehog Proteins/genetics , Medulloblastoma/genetics , Tumor Microenvironment/genetics , Cerebellar Neoplasms/genetics
17.
Nat Commun ; 15(1): 3365, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664376

ABSTRACT

Hedgehog (Hh) signaling relies on the primary cilium, a cell surface organelle that serves as a signaling hub for the cell. Using proximity labeling and quantitative proteomics, we identify Numb as a ciliary protein that positively regulates Hh signaling. Numb localizes to the ciliary pocket and acts as an endocytic adaptor to incorporate Ptch1 into clathrin-coated vesicles, thereby promoting Ptch1 exit from the cilium, a key step in Hh signaling activation. Numb loss impedes Sonic hedgehog (Shh)-induced Ptch1 exit from the cilium, resulting in reduced Hh signaling. Numb loss in spinal neural progenitors reduces Shh-induced differentiation into cell fates reliant on high Hh activity. Genetic ablation of Numb in the developing cerebellum impairs the proliferation of granule cell precursors, a Hh-dependent process, resulting in reduced cerebellar size. This study highlights Numb as a regulator of ciliary Ptch1 levels during Hh signal activation and demonstrates the key role of ciliary pocket-mediated endocytosis in cell signaling.


Subject(s)
Cerebellum , Cilia , Hedgehog Proteins , Nerve Tissue Proteins , Patched-1 Receptor , Signal Transduction , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Cilia/metabolism , Animals , Patched-1 Receptor/metabolism , Patched-1 Receptor/genetics , Mice , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Cerebellum/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Humans , Endocytosis , Cell Differentiation , Cell Proliferation , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Mice, Knockout
18.
Ecotoxicol Environ Saf ; 276: 116311, 2024 May.
Article in English | MEDLINE | ID: mdl-38615639

ABSTRACT

Prenatal environmental exposure could be an essential health risk factor associated with neurodevelopmental disorders in offspring. However, the exact mechanisms underlying the impact of prenatal PM2.5 exposure on offspring cognition remain unclear. In our recent study using a PM2.5 exposed pregnant mouse model, we observed significant synaptic dysfunction in the hippocampi of the offspring. Concurrently, the epigenetic regulator of KDM5A and the Shh signaling pathway exhibited decreased activities. Significantly, changes in hippocampal KDM5A and Shh levels directly correlated with PM2.5 exposure intensity. Subsequent experiments revealed a marked reduction in the expression of Shh signaling and related synaptic proteins when KDM5A was silenced in cells. Notably, the effects of KDM5A deficiency were reversed significantly with the supplementation of a Shh activator. Furthermore, our findings indicate that Shh activation significantly attenuates PM2.5-induced synaptic impairments in hippocampal neurons. We further demonstrated that EGR1, a transcriptional inhibitor, plays a direct role in KDM5A's regulation of the Shh pathway under conditions of PM2.5 exposure. Our results suggest that the KDM5A's inhibitory regulation on the Shh pathway through the EGR1 gene is a crucial epigenetic mechanism underlying the synaptic dysfunction in hippocampal neurons caused by maternal PM2.5 exposure. This emphasizes the role of epigenetic regulations in neurodevelopmental disorders caused by environmental factors.


Subject(s)
Epigenesis, Genetic , Hedgehog Proteins , Hippocampus , Particulate Matter , Prenatal Exposure Delayed Effects , Signal Transduction , Hippocampus/drug effects , Animals , Female , Pregnancy , Signal Transduction/drug effects , Epigenesis, Genetic/drug effects , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mice , Particulate Matter/toxicity , Retinoblastoma-Binding Protein 2/genetics , Maternal Exposure/adverse effects , Synapses/drug effects , Air Pollutants/toxicity
19.
J Oral Pathol Med ; 53(5): 277-293, 2024 May.
Article in English | MEDLINE | ID: mdl-38664938

ABSTRACT

Ameloblastoma (AM) is a benign, although aggressive, epithelial odontogenic tumour originating from tooth-forming tissues or remnants. Its aetiopathogenesis remains unclear; however, molecular analysis techniques have allowed researchers to progress in understanding its genetic basis. The high frequency of BRAF p.V600E as a main driver mutation in AM is well established; nevertheless, it is insufficient to explain its tumourigenesis. In this review, we aimed to integrate the current knowledge about the biology of AM and to describe the main genetic alterations reported, focusing on the findings of large-scale sequencing and gene expression profiling techniques. Current evidence shows that besides BRAF mutation and activation of the MAPK pathway, alterations in Hedgehog and Wnt/ß-catenin pathway-related genes are also involved in AM pathogenesis. Recently, a tumour suppressor gene, KMT2D, has been reported as mutated by different research groups. The biological impact of these mutations in the pathogenesis of AM has yet to be elucidated. Further studies are needed to clarify the impact of these findings in the identification of novel biomarkers that could be useful for diagnosing, classifying, and molecular targeting this neoplasm.


Subject(s)
Ameloblastoma , Mutation , Proto-Oncogene Proteins B-raf , Ameloblastoma/genetics , Ameloblastoma/pathology , Humans , Proto-Oncogene Proteins B-raf/genetics , Jaw Neoplasms/genetics , Wnt Signaling Pathway/genetics , Hedgehog Proteins/genetics , Gene Expression Profiling
20.
Int J Mol Sci ; 25(7)2024 Apr 07.
Article in English | MEDLINE | ID: mdl-38612913

ABSTRACT

Salt is frequently introduced in ecosystems, where it acts as a pollutant. This study examined how changes in salinity affect the survival and development of zebrafish from the two-cell to the blastocyst stage and from the blastocyst to the larval stage. Control zebrafish embryos were cultured in E3 medium containing 5 mM Sodium Chloride (NaCl), 0.17 mM Potassium Chloride (KCL), 0.33 mM Calcium Chloride (CaCl2), and 0.33 mM Magnesium Sulfade (MgSO4). Experiments were conducted using increasing concentrations of each individual salt at 5×, 10×, 50×, and 100× the concentration found in E3 medium. KCL, CaCl2, and MgSO4 did not result in lethal abnormalities and did not affect early embryo growth at any of the concentrations tested. Concentrations of 50× and 100× NaCl caused embryonic death in both stages of development. Concentrations of 5× and 10× NaCl resulted in uninflated swim bladders in 12% and 65% of larvae, compared to 4.2% of controls, and caused 1654 and 2628 genes to be differentially expressed in blastocysts, respectively. The ATM signaling pathway was affected, and the Sonic Hedgehog pathway genes Shh and Ptc1 implicated in swim bladder development were downregulated. Our findings suggest that increased NaCl concentrations may alter gene expression and cause developmental abnormalities in animals found in affected ecosystems.


Subject(s)
Hedgehog Proteins , Perciformes , Animals , Hedgehog Proteins/genetics , Sodium Chloride/pharmacology , Water , Zebrafish/genetics , Calcium Chloride , Ecosystem , Sodium Chloride, Dietary , Larva/genetics , Gene Expression
SELECTION OF CITATIONS
SEARCH DETAIL
...