Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Development ; 147(13)2020 07 13.
Article in English | MEDLINE | ID: mdl-32661019

ABSTRACT

New neurons are generated in the postnatal rodent hypothalamus, with a subset of tanycytes in the third ventricular (3V) wall serving as neural stem/progenitor cells. However, the precise stem cell niche organization, the intermediate steps and the endogenous regulators of postnatal hypothalamic neurogenesis remain elusive. Quantitative lineage-tracing in vivo revealed that conditional deletion of fibroblast growth factor 10 (Fgf10) from Fgf10-expressing ß-tanycytes at postnatal days (P)4-5 results in the generation of significantly more parenchymal cells by P28, composed mostly of ventromedial and dorsomedial neurons and some glial cells, which persist into adulthood. A closer scrutiny in vivo and ex vivo revealed that the 3V wall is not static and is amenable to cell movements. Furthermore, normally ß-tanycytes give rise to parenchymal cells via an intermediate population of α-tanycytes with transient amplifying cell characteristics. Loss of Fgf10 temporarily attenuates the amplification of ß-tanycytes but also appears to delay the exit of their α-tanycyte descendants from the germinal 3V wall. Our findings suggest that transience of cells through the α-tanycyte domain is a key feature, and Fgf10 is a negative regulator of postnatal hypothalamic neurogenesis.


Subject(s)
Fibroblast Growth Factor 10/metabolism , Hypothalamus/cytology , Hypothalamus/metabolism , Neurogenesis/physiology , Animals , Cell Movement/physiology , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Female , Fibroblast Growth Factor 10/genetics , Male , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism
2.
J Anat ; 238(6): 1371-1385, 2021 06.
Article in English | MEDLINE | ID: mdl-33455001

ABSTRACT

A common question in organ regeneration is the extent to which regeneration recapitulates embryonic development. To investigate this concept, we compared the expression of two highly interlinked and essential genes for salivary gland development, Sox9 and Fgf10, during submandibular gland development, homeostasis and regeneration. Salivary gland duct ligation/deligation model was used as a regenerative model. Fgf10 and Sox9 expression changed during regeneration compared to homeostasis, suggesting that these key developmental genes play important roles during regeneration, however, significantly both displayed different patterns of expression in the regenerating gland compared to the developing gland. Regenerating glands, which during homeostasis had very few weakly expressing Sox9-positive cells in the striated/granular ducts, displayed elevated expression of Sox9 within these ducts. This pattern is in contrast to embryonic development, where Sox9 expression was absent in the proximally developing ducts. However, similar to the elevated expression at the distal tip of the epithelium in developing salivary glands, regenerating glands displayed elevated expression in a subpopulation of acinar cells, which during homeostasis expressed Sox9 at lower levels. A shift in expression of Fgf10 was observed from a widespread mesenchymal pattern during organogenesis to a more limited and predominantly epithelial pattern during homeostasis in the adult. This restricted expression in epithelial cells was maintained during regeneration, with no clear upregulation in the surrounding mesenchyme, as might be expected if regeneration recapitulated development. As both Fgf10 and Sox9 were upregulated in proximal ducts during regeneration, this suggests that the positive regulation of Sox9 by Fgf10, essential during development, is partially reawakened during regeneration using this model. Together these data suggest that developmentally important genes play a key role in salivary gland regeneration but do not precisely mimic the roles observed during development.


Subject(s)
Organogenesis/physiology , Regeneration/physiology , Submandibular Gland/physiology , Animals , Female , Fibroblast Growth Factor 10/metabolism , Gene Expression Regulation, Developmental , Male , Mice , SOX9 Transcription Factor/metabolism , Submandibular Gland/embryology
3.
Dev Dyn ; 248(9): 882-893, 2019 09.
Article in English | MEDLINE | ID: mdl-31290205

ABSTRACT

BACKGROUND: Fibroblast growth factor 18 (FGF18) functions in the development of several tissues, including the lung, limb bud, palate, skeleton, central nervous system, and hair follicle. Mice containing a germline knockout of Fgf18 (Fgf18 -/- ) die shortly after birth. Postnatally, FGF18 is being evaluated for pathogenic roles in fibrosis and several types of cancer. The specific cell types that express FGF18 have been difficult to identify, and the function of FGF18 in postnatal development and tissue homeostasis has been hampered by the perinatal lethality of Fgf18 null mice. RESULTS: We engineered a floxed allele of Fgf18 (Fgf18 flox ) that allows conditional gene inactivation and a CreERT2 knockin allele (Fgf18 CreERT2 ) that allows the precise identification of cells that express Fgf18 and their lineage. We validated the Fgf18 flox allele by targeting it in mesenchymal tissue and primary mesoderm during embryonic development, resulting in similar phenotypes to those observed in Fgf18 null mice. We also use the Fgf18 CreERT2 allele, in combination with a conditional fluorescent reporter to confirm known and identify new sites of Fgf18 expression. CONCLUSION: These alleles will be useful to investigate FGF18 function during organogenesis and tissue homeostasis, and to target specific cell lineages at embryonic and postnatal time points.


Subject(s)
Alleles , Fibroblast Growth Factors/metabolism , Integrases/genetics , Protein Engineering/methods , Animals , Cell Lineage , Embryonic Development , Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/physiology , Homeostasis , Mesoderm , Mice , Organogenesis
4.
Biochem J ; 473(24): 4593-4607, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27742760

ABSTRACT

Heterozygous mutations in the gene encoding fibroblast growth factor 10 (FGF10) or its cognate receptor, FGF-receptor 2 IIIb result in two human syndromes - LADD (lacrimo-auriculo-dento-digital) and ALSG (aplasia of lacrimal and salivary glands). To date, the partial loss-of-FGF10 function in these patients has been attributed solely to perturbed paracrine signalling functions between FGF10-producing mesenchymal cells and FGF10-responsive epithelial cells. However, the functioning of a LADD-causing G138E FGF10 mutation, which falls outside its receptor interaction interface, has remained enigmatic. In the present study, we interrogated this mutation in the context of FGF10's protein sequence and three-dimensional structure, and followed the subcellular fate of tagged proteins containing this or other combinatorial FGF10 mutations, in vitro We report that FGF10 harbours two putative nuclear localization sequences (NLSs), termed NLS1 and NLS2, which individually or co-operatively promote nuclear translocation of FGF10. Furthermore, FGF10 localizes to a subset of dense fibrillar components of the nucleolus. G138E falls within NLS1 and abrogates FGF10's nuclear translocation whilst attenuating its progression along the secretory pathway. Our findings suggest that in addition to its paracrine roles, FGF10 may normally play intracrine role/s within FGF10-producing cells. Thus, G138E may disrupt both paracrine and intracrine function/s of FGF10 through attenuated secretion and nuclear translocation, respectively.


Subject(s)
Abnormalities, Multiple/metabolism , Cell Nucleus/metabolism , Fibroblast Growth Factor 10/chemistry , Fibroblast Growth Factor 10/metabolism , Hearing Loss/metabolism , Lacrimal Apparatus Diseases/metabolism , Syndactyly/metabolism , Tooth Abnormalities/metabolism , Abnormalities, Multiple/genetics , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/physiology , Amino Acid Sequence , Blotting, Western , Cell Proliferation/genetics , Cell Proliferation/physiology , Fibroblast Growth Factor 10/genetics , HEK293 Cells , Hearing Loss/genetics , Humans , Immunohistochemistry , Lacrimal Apparatus Diseases/genetics , Molecular Sequence Data , Mutation , Protein Transport/genetics , Protein Transport/physiology , Syndactyly/genetics , Tooth Abnormalities/genetics
5.
J Neurosci ; 33(14): 6170-80, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23554498

ABSTRACT

Increasing evidence suggests that neurogenesis occurs in the postnatal and adult mammalian hypothalamus. However, the identity and location of the putative progenitor cells is under much debate, and little is known about the dynamics of neurogenesis in unchallenged brain. Previously, we postulated that Fibroblast growth factor 10-expressing (Fgf10(+)) tanycytes constitute a population of progenitor cells in the mouse hypothalamus. Here, we show that Fgf10(+) tanycytes express markers of neural stem/progenitor cells, divide late into postnatal life, and can generate both neurons and astrocytes in vivo. Stage-specific lineage-tracing of Fgf10(+) tanycytes using Fgf10-creERT2 mice, reveals robust neurogenesis at postnatal day 28 (P28), lasting as late as P60. Furthermore, we present evidence for amplification of Fgf10-lineage traced neural cells within the hypothalamic parenchyma itself. The neuronal descendants of Fgf10(+) tanycytes predominantly populate the arcuate nucleus, a subset of which express the orexigenic neuronal marker, Neuropeptide-Y, and respond to fasting and leptin-induced signaling. These studies provide direct evidence in support of hypothalamic neurogenesis during late postnatal and adult life, and identify Fgf10(+) tanycytes as a source of parenchymal neurons with putative roles in appetite and energy balance.


Subject(s)
Appetite/physiology , Cytosol/metabolism , Energy Metabolism/physiology , Fibroblast Growth Factor 10/metabolism , Hypothalamus/metabolism , Neurogenesis/physiology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Appetite/drug effects , Bacterial Proteins/genetics , Bromodeoxyuridine , Energy Metabolism/drug effects , Estrogen Antagonists/pharmacology , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins/metabolism , Fibroblast Growth Factor 10/genetics , Food Deprivation/physiology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Hypothalamus/drug effects , Hypothalamus/growth & development , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Phenylurea Compounds/administration & dosage , Proteins/genetics , RNA, Untranslated , Tamoxifen/pharmacology , beta-Galactosidase/metabolism
6.
Arthritis Rheum ; 64(6): 1909-19, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22143896

ABSTRACT

OBJECTIVE: To use an in vitro model of chondrogenesis to identify microRNAs (miRNAs) with a functional role in cartilage homeostasis. METHODS: The expression of miRNAs was measured in the ATDC5 cell model of chondrogenesis using microarray and was verified using quantitative reverse transcription-polymerase chain reaction. MicroRNA expression was localized by in situ hybridization. Predicted miRNA target genes were validated using 3'-untranslated region-Luc reporter plasmids containing either wild-type sequences or mutants of the miRNA target sequence. Signaling through the Smad pathway was measured using a (CAGA)(12) -Luc reporter. RESULTS: The expression of several miRNAs was regulated during chondrogenesis. These included 39 miRNAs that are coexpressed with miRNA-140 (miR-140), which is known to be involved in cartilage homeostasis and osteoarthritis (OA). Of these miRNAs, miR-455 resides within an intron of COL27A1 that encodes a cartilage collagen. When human OA cartilage was compared with cartilage obtained from patients with femoral neck fractures, the expression of both miR-140-5p and miR-455-3p was increased in OA cartilage. In situ hybridization showed miR-455-3p expression in the developing limbs of chicks and mice and in human OA cartilage. The expression of miR-455-3p was regulated by transforming growth factor ß (TGFß) ligands, and miRNA regulated TGFß signaling. ACVR2B, SMAD2, and CHRDL1 were direct targets of miR-455-3p and may mediate its functional impact on TGFß signaling. CONCLUSION: MicroRNA-455 is expressed during chondrogenesis and in adult articular cartilage, where it can regulate TGFß signaling, suppressing the Smad2/3 pathway. Diminished signaling through this pathway during the aging process and in OA chondrocytes is known to contribute to cartilage destruction. We propose that the increased expression of miR-455 in OA exacerbates this process and contributes to disease pathology.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Chondrogenesis/physiology , Hip Joint/metabolism , MicroRNAs/metabolism , Osteoarthritis, Hip/metabolism , 3T3 Cells , Adult , Aged , Aged, 80 and over , Animals , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/pathology , Female , Hip Joint/pathology , Humans , Male , Mice , MicroRNAs/genetics , Middle Aged , Osteoarthritis, Hip/genetics , Osteoarthritis, Hip/pathology
7.
Biochem J ; 436(1): 71-81, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21355848

ABSTRACT

AS (Apert syndrome) is a congenital disease composed of skeletal, visceral and neural abnormalities, caused by dominant-acting mutations in FGFR2 [FGF (fibroblast growth factor) receptor 2]. Multiple FGFR2 splice variants are generated through alternative splicing, including PTC (premature termination codon)-containing transcripts that are normally eliminated via the NMD (nonsense-mediated decay) pathway. We have discovered that a soluble truncated FGFR2 molecule encoded by a PTC-containing transcript is up-regulated and persists in tissues of an AS mouse model. We have termed this IIIa-TM as it arises from aberrant splicing of FGFR2 exon 7 (IIIa) into exon 10 [TM (transmembrane domain)]. IIIa-TM is glycosylated and can modulate the binding of FGF1 to FGFR2 molecules in BIAcore-binding assays. We also show that IIIa-TM can negatively regulate FGF signalling in vitro and in vivo. AS phenotypes are thought to result from gain-of-FGFR2 signalling, but our findings suggest that IIIa-TM can contribute to these through a loss-of-FGFR2 function mechanism. Moreover, our findings raise the interesting possibility that FGFR2 signalling may be a regulator of the NMD pathway.


Subject(s)
Acrocephalosyndactylia/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Up-Regulation , Acrocephalosyndactylia/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Exons , Humans , Mice , Mice, Inbred Strains , Models, Animal , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism
8.
Cell Rep ; 39(2): 110663, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35417692

ABSTRACT

Fibroblast growth factor 10 (FGF10) is well established as a mesenchyme-derived growth factor and a critical regulator of fetal organ development in mice and humans. Using a single-cell RNA sequencing (RNA-seq) atlas of salivary gland (SG) and a tamoxifen inducible Fgf10CreERT2:R26-tdTomato mouse, we show that FGF10pos cells are exclusively mesenchymal until postnatal day 5 (P5) but, after P7, there is a switch in expression and only epithelial FGF10pos cells are observed after P15. Further RNA-seq analysis of sorted mesenchymal and epithelial FGF10pos cells shows that the epithelial FGF10pos population express the hallmarks of ancient ionocyte signature Forkhead box i1 and 2 (Foxi1, Foxi2), Achaete-scute homolog 3 (Ascl3), and the cystic fibrosis transmembrane conductance regulator (Cftr). We propose that epithelial FGF10pos cells are specialized SG ionocytes located in ducts and important for the ionic modification of saliva. In addition, they maintain FGF10-dependent gland homeostasis via communication with FGFR2bpos ductal and myoepithelial cells.


Subject(s)
Fibroblast Growth Factor 10 , Receptor, Fibroblast Growth Factor, Type 2 , Salivary Glands , Animals , Epithelial Cells/metabolism , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/metabolism , Forkhead Transcription Factors/metabolism , Mice , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Salivary Glands/cytology , Salivary Glands/metabolism , Signal Transduction
9.
Gut Microbes ; 14(1): 2073784, 2022.
Article in English | MEDLINE | ID: mdl-35579971

ABSTRACT

Ruminococcus gnavus is a prevalent member of the human gut microbiota, which is over-represented in inflammatory bowel disease and neurological disorders. We previously showed that the ability of R. gnavus to forage on mucins is strain-dependent and associated with sialic acid metabolism. Here, we showed that mice monocolonized with R. gnavus ATCC 29149 (Rg-mice) display changes in major sialic acid derivatives in their cecum content, blood, and brain, which is accompanied by a significant decrease in the percentage of sialylated residues in intestinal mucins relative to germ-free (GF) mice. Changes in metabolites associated with brain function such as tryptamine, indolacetate, and trimethylamine N-oxide were also detected in the cecal content of Rg-mice when compared to GF mice. Next, we investigated the effect of R. gnavus monocolonization on hippocampus cell proliferation and behavior. We observed a significant decrease of PSA-NCAM immunoreactive granule cells in the dentate gyrus (DG) of Rg-mice as compared to GF mice and recruitment of phagocytic microglia in the vicinity. Behavioral assessments suggested an improvement of the spatial working memory in Rg-mice but no change in other cognitive functions. These results were also supported by a significant upregulation of genes involved in proliferation and neuroplasticity. Collectively, these data provide first insights into how R. gnavus metabolites may influence brain regulation and function through modulation of granule cell development and synaptic plasticity in the adult hippocampus. This work has implications for further understanding the mechanisms underpinning the role of R. gnavus in neurological disorders.


Subject(s)
Brain , Clostridiales , Gastrointestinal Microbiome , Mucins , Animals , Brain/metabolism , Mice , Mucins/metabolism , N-Acetylneuraminic Acid/metabolism , Polysaccharides/metabolism
10.
Dev Biol ; 335(1): 143-55, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19716814

ABSTRACT

Formation of the vertebrate nervous system requires coordinated cell-cell interactions, intracellular signalling events, gene transcription, and morphogenetic cell movements. Wnt signalling has been involved in regulating a wide variety of biological processes such as embryonic patterning, cell proliferation, cell polarity, motility, and the specification of cell fate. Wnt ligands associate with their receptors, members of the frizzled family (Fz). In Xenopus, five members of the frizzled family are expressed in the early nervous system. We have investigated the role of Xenopus frizzled-10 (Fz10) in neural development. We show that Fz10 is expressed in the dorsal neural ectoderm and neural folds in the region where primary sensory neurons develop. Fz10 mediates canonical Wnt signalling and interacts with Wnt1 and Wnt8 but not Wnt3a as shown in synergy assays. We find that Fz10 is required for the late stages of sensory neuron differentiation. Overexpression of Fz10 in Xenopus leads to an increase in the number of sensory neurons. Loss of Fz10 function using morpholinos inhibits the development of sensory neurons in Xenopus at later stages of neurogenesis and this can be rescued by co-injection of modified Fz10B and beta-catenin. In mouse P19 cells induced by retinoic acid to undergo neural differentiation, overexpression of Xenopus Fz10 leads to an increase in the number of neurons generated while siRNA knockdown of endogenous mouse Fz10 inhibits neurogenesis. Thus we propose Fz10 mediates Wnt1 signalling to determine sensory neural differentiation in Xenopus in vivo and in mouse cell culture.


Subject(s)
Cell Differentiation/physiology , Frizzled Receptors/metabolism , Neurogenesis/physiology , Sensory Receptor Cells/physiology , Signal Transduction/physiology , Xenopus Proteins/metabolism , Xenopus laevis , Animals , Biomarkers/metabolism , Cell Line , Frizzled Receptors/genetics , Gene Knockdown Techniques , Humans , Mice , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sensory Receptor Cells/cytology , Wnt Proteins/genetics , Wnt Proteins/metabolism , Xenopus Proteins/genetics , Xenopus laevis/anatomy & histology , Xenopus laevis/embryology , Xenopus laevis/metabolism , beta Catenin/genetics , beta Catenin/metabolism
11.
Front Cell Dev Biol ; 8: 609643, 2020.
Article in English | MEDLINE | ID: mdl-33363172

ABSTRACT

The pinna (or auricle) is part of the external ear, acting to capture and funnel sound toward the middle ear. The pinna is defective in a number of craniofacial syndromes, including Lacrimo-auriculo-dento-digital (LADD) syndrome, which is caused by mutations in FGF10 or its receptor FGFR2b. Here we study pinna defects in the Fgf10 knockout mouse. We show that Fgf10 is expressed in both the muscles and forming cartilage of the developing external ear, with loss of signaling leading to a failure in the normal extension of the pinna over the ear canal. Conditional knockout of Fgf10 in the neural crest fails to recapitulate this phenotype, suggesting that the defect is due to loss of Fgf10 from the muscles, or that this source of Fgf10 can compensate for loss in the forming cartilage. The defect in the Fgf10 null mouse is driven by a reduction in proliferation, rather than an increase in cell death, which can be partially phenocopied by inhibiting cell proliferation in explant culture. Overall, we highlight the mechanisms that could lead to the phenotype observed in LADD syndrome patients and potentially explain the formation of similar low-set and cup shaped ears observed in other syndromes.

12.
Pediatr Res ; 66(4): 386-90, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19581825

ABSTRACT

Patients with Apert syndrome (AS) display a wide range of congenital malformations including tracheal stenosis, which is a disease characterized by a uniform cartilaginous sleeve in place of a normally ribbed cartilagenous trachea. We have studied the cellular and molecular basis of this phenotype in a mouse model of AS (Fgfr2c(+/Delta) mice), which shows ectopic expression of Fgfr2b in mesenchymal tissues. Here we report that tracheal stenosis is associated with increased proliferation of mesenchymal cells, where the expression of Fgf10 and its upstream regulators Tbx4 and Tbx5 are abnormally elevated. We show that Fgf10 has a critical inductive role in tracheal stenosis, as genetic knockdown of Fgf10 in Fgfr2c(+/Delta) mice rescues this phenotype. These novel findings demonstrate a regulatory role for Fgf10 in tracheal development and shed more light on the underlying cause of tracheal defects in AS.


Subject(s)
Acrocephalosyndactylia/pathology , Cartilage/abnormalities , Fibroblast Growth Factor 10/metabolism , Trachea/anatomy & histology , Acrocephalosyndactylia/metabolism , Animals , Body Patterning , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Fibroblast Growth Factor 10/genetics , Humans , Mice , Mice, Transgenic , Phenotype , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction/physiology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
13.
Mol Cell Neurosci ; 37(4): 857-68, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18329286

ABSTRACT

We used Fgf10-lacZ reporter mice to investigate the distribution and fate of Fgf10-expressing cells in the developing and adult mouse brain. We find that the domain of Fgf10 expression expands post-natally and new niches emerge in the adult brain. Fgf10 is expressed in the adult cerebellum, thalamic, mid- and hindbrain nuclei and hippocampal CA fields, as previously reported in the rat brain. In addition though, we have discovered expression in: the hippocampal dentate gyrus; a discrete trail linking the ventral telencephalon with the olfactory bulbs; ventral ependyma of the third ventricle from where cells appear to disperse into the hypothalamus; and in the pituitary gland. Most Fgf10-expressing cells or their immediate descendants appear immature but a subset differentiates into neurons and glial cells. The manner in which Fgf10 is expressed in these active and quiescent neurogenic niches implicates it in control of neurogenesis and/or conservation of neurogenic potential.


Subject(s)
Brain/growth & development , Cell Differentiation/physiology , Fibroblast Growth Factor 10/genetics , Gene Expression Regulation, Developmental/physiology , Neurons/cytology , Neurons/physiology , Age Factors , Animals , Brain/cytology , Fibroblast Growth Factor 10/biosynthesis , Fibroblast Growth Factor 10/physiology , Mice , Mice, Transgenic , Neurons/chemistry
14.
J Neuroendocrinol ; 31(8): e12750, 2019 08.
Article in English | MEDLINE | ID: mdl-31111569

ABSTRACT

The mammalian hypothalamus regulates key homeostatic and neuroendocrine functions ranging from circadian rhythm and energy balance to growth and reproductive cycles via the hypothalamic-pituitary and hypothalamic-thyroid axes. In addition to its neurones, tanycytes are taking centre stage in the short- and long-term augmentation and integration of diverse hypothalamic functions, although the genetic regulators and mediators of their involvement are poorly understood. Exogenous interventions have implicated fibroblast growth factor (FGF) signalling, although the focal point of the action of FGF and any role for putative endogenous players also remains elusive. We carried out a comprehensive high-resolution screen of FGF signalling pathway mediators and modifiers using a combination of in situ hybridisation, immunolabelling and transgenic reporter mice, aiming to map their spatial distribution in the adult hypothalamus. Our findings suggest that ß-tanycytes are the likely focal point of exogenous and endogenous action of FGF in the third ventricular wall, utilising FGF receptor (FGFR)1 and FGFR2 IIIc isoforms, but not FGFR3. Key IIIc-activating endogenous ligands include FGF1, 2, 9 and 18, which are expressed by a subset of ependymal and parenchymal cells. In the parenchymal compartment, FGFR1-3 show divergent patterns, with FGFR1 being predominant in neuronal nuclei and expression of FGFR3 being associated with glial cell function. Intracrine FGFs are also present, suggestive of multiple modes of FGF function. Our findings provide a testable framework for understanding the complex role of FGFs with respect to regulating the metabolic endocrine and neurogenic functions of hypothalamus in vivo.


Subject(s)
Appetite Regulation/genetics , Energy Metabolism/genetics , Ependymoglial Cells/physiology , Fibroblast Growth Factors/physiology , Hypothalamus/cytology , Animals , Appetite Regulation/drug effects , Energy Metabolism/drug effects , Ependymoglial Cells/drug effects , Female , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
15.
Histol Histopathol ; 33(12): 1253-1270, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29749591

ABSTRACT

The Blood-Brain Barrier (BBB) is a highly specialised interface separating the Central Nervous System (CNS) from circulating blood. Dysregulation of the BBB is a key early event in pathological conditions such as inflammation, in which the entry of activated leukocytes into the CNS is facilitated by BBB breakdown. The metzincin family of metalloproteinases (MPs) is one of the major contributors to BBB permeability as they cleave endothelial cell-cell contacts and underlying basal lamina components. However, the mechanisms by which MPs regulate BBB integrity has not yet been fully elucidated. The aim of this review is to provide an overview of pathways by which MPs could regulate the BBB in the context of neuroinflammation.


Subject(s)
Blood-Brain Barrier/pathology , Capillary Permeability/physiology , Metalloproteases/metabolism , Signal Transduction/physiology , Animals , Blood-Brain Barrier/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology
16.
FEBS Lett ; 580(17): 4214-7, 2006 Jul 24.
Article in English | MEDLINE | ID: mdl-16828749

ABSTRACT

MicroRNAs (miRNA) are short RNA molecules regulating the expression of specific mRNAs. We investigated the expression pattern and potential targets of mouse miR-140 and found that miR-140 is specifically expressed in cartilage tissues of mouse embryos during both long and flat bone development. MiR-140 expression was detected in the limbs of E11.5 embryos in the primorida of future bones both in the fore and hindlimb and across autopod, zeugopod and stylopod. All digits of E14.5 fore- and hindlimbs showed accumulation of miR-140, except the first digit of the hindlimb. MiR-140 expression was also detected in the cartilagenous base of E17.5 skulls and in the sternum, the proximal rib heads and the developing vertebral column of E15.5 embryos. A potential target of miR-140, histone deacetylase 4, was validated experimentally and the possible role of miR-140 in long bone development is discussed.


Subject(s)
Cartilage/embryology , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Histone Deacetylases/biosynthesis , MicroRNAs/biosynthesis , Osteogenesis/physiology , Animals , Cartilage/cytology , Hindlimb/cytology , Hindlimb/embryology , In Situ Hybridization , Mice , Skull Base/cytology , Skull Base/embryology
17.
BMC Dev Biol ; 5: 11, 2005 Jun 22.
Article in English | MEDLINE | ID: mdl-15972105

ABSTRACT

BACKGROUND: Analyses of Fgf10 and Fgfr2b mutant mice, as well as human studies, suggest that FGF10/FGFR2b signaling may play an essential, nonredundant role during embryonic SMG development. To address this question, we have analyzed the SMG phenotype in Fgf10 and Fgfr2b heterozygous and null mutant mice. In addition, although previous studies suggest that the FGF10/FGFR2b and FGF8/FGFR2c signaling pathways are functionally interrelated, little is known about the functional relationship between these two pathways during SMG development. We have designed in vivo and in vitro experiments to address this question. RESULTS: We analyzed Fgf10 and Fgfr2b heterozygous mutant and null mice and demonstrate dose-dependent SMG phenotypic differences. Hypoplastic SMGs are seen in Fgf10 and Fgfr2b heterozygotes whereas SMG aplasia is seen in Fgf10 and Fgfr2b null embryos. Complementary in vitro studies further indicate that FGF10/FGFR2b signaling regulates SMG epithelial branching and cell proliferation. To delineate the functional relationship between the FGF10/FGFR2b and FGF8/FGFR2c pathways, we compared the SMG phenotype in Fgfr2c+/Delta/Fgf10+/- double heterozygous mice to that seen in wildtype, Fgf10+/- (Fgfr2c+/+/Fgf10+/-) and Fgfr2c+/Delta (Fgfr2c+/Delta/Fgf10+/+) single heterozygous mutant littermates and demonstrate genotype-specific SMG phenotypes. In addition, exogenous FGF8 was able to rescue the abnormal SMG phenotype associated with abrogated FGFR2b signaling in vitro and restore branching to normal levels. CONCLUSION: Our data indicates that FGF10/FGFR2b signaling is essential for the SMG epithelial branching and histodifferentiation, but not earliest initial bud formation. The functional presence of other endogenous signaling pathways could not prevent complete death of embryonic SMG cells in Fgf10 and Fgfr2b null mice. Though we were able to rescue the abnormal phenotype associated with reduced in vitro FGF10/FGFR2b signaling with exogenous FGF8 supplementation, our results indicate that the FGF10/FGFR2b and FGF8/FGFR2c are nonredundant signaling pathways essential for in vivo embryonic SMG development. What remains to be determined is the in vivo functional relationship between the FGF10/FGFR2b signal transduction pathway and other key signaling pathways, and how these pathways are integrated during embryonic SMG development to compose the functional epigenome.


Subject(s)
Fibroblast Growth Factor 10/physiology , Morphogenesis , Receptor, Fibroblast Growth Factor, Type 2/physiology , Signal Transduction/physiology , Submandibular Gland/embryology , Animals , Cell Proliferation , Embryo, Mammalian , Epithelial Cells/cytology , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 8/physiology , Genotype , Mice , Mice, Knockout , Receptor, Fibroblast Growth Factor, Type 2/genetics , Submandibular Gland/cytology , Submandibular Gland/growth & development
18.
Mech Dev ; 113(1): 79-83, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11900978

ABSTRACT

The normal development of calvarial bones and sutures critically relies on proper signalling through Fgf receptors, but the source and identity of cognate ligands have remained unknown. Reverse transcriptase polymerase chain reaction analysis in this study shows that a broad range of Fgf ligands are expressed in the coronal sutures separating the parietal and frontal bones. Analysis by whole mount in situ hybridization further reveals distinct expression patterns for Fgf-18, Fgf-20, and by comparison, Fgf-9, in the calvaria, and Fgfs-20 and -9 in the developing limbs, suggestive of their role in proliferation, differentiation and apoptosis.


Subject(s)
Extremities/embryology , Fibroblast Growth Factors/biosynthesis , Skull/embryology , Animals , Apoptosis , Cell Differentiation , Cell Division , Fibroblast Growth Factor 9 , Gene Expression Regulation, Developmental , In Situ Hybridization , Ligands , Mice , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Tissue Distribution
19.
Front Neurosci ; 9: 387, 2015.
Article in English | MEDLINE | ID: mdl-26578855

ABSTRACT

There is a resurgent interest in tanycytes, a radial glial-like cell population occupying the floor and ventro-lateral walls of the third ventricle (3V). Tanycytes reside in close proximity to hypothalamic neuronal nuclei that regulate appetite and energy expenditure, with a subset sending projections into these nuclei. Moreover, tanycytes are exposed to 3V cerebrospinal fluid and have privileged access to plasma metabolites and hormones, through fenestrated capillaries. Indeed, some tanycytes act as conduits for trafficking of these molecules into the brain parenchyma. Tanycytes can also act as neural stem/progenitor cells, supplying the postnatal and adult hypothalamus with new neurons. Collectively, these findings suggest that tanycytes regulate and integrate important trophic and metabolic processes and possibly endow functional malleability to neuronal circuits of the hypothalamus. Hence, manipulation of tanycyte biology could provide a valuable tool for modulating hypothalamic functions such as energy uptake and expenditure in order to tackle prevalent eating disorders such as obesity and anorexia.

20.
Anat Embryol (Berl) ; 208(6): 479-86, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15340846

ABSTRACT

Members of the fibroblast growth factor (FGF) family play diverse roles during the development and patterning of various organs. In human and mice, 22 FGFs and four receptors derived from several splice variants are present. Redundant expression and function of FGF genes in organogenesis have been reported, but their roles in embryonic external genitalia, genital tubercle (GT), development have not been studied in detail. To address the role of FGF during external genitalia development, we have analyzed the expression of FGF genes (Fgf8, 9, 10) and receptor genes (Fgfr1, r2IIIb, r2IIIc) in GT of mice. Furthermore, Fgf10 and Fgfr2IIIb mutant mice were analyzed to elucidate their roles in embryonic external genitalia development. Fgfr2IIIb was expressed in urethral plate epithelium during GT development. Fgfr2IIIb mutant mice display urethral dysmorphogenesis. Marker gene analysis for urethral plate and bilateral mesenchymal formation suggests the existence of epithelial-mesenchymal interaction during urethral morphogenesis. Therefore, FGF10/FGFR2IIIb signals seem to constitute a developmental cascade for such morphogenesis.


Subject(s)
Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Fibroblast Growth Factor/genetics , Urethra/embryology , Animals , Female , Fibroblast Growth Factor 10 , Fibroblast Growth Factors/metabolism , Genetic Markers , Ligands , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Urethra/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL