Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 180
Filter
Add more filters

Publication year range
1.
Development ; 151(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38602479

ABSTRACT

Alveologenesis is the final stage of lung development in which the internal surface area of the lung is increased to facilitate efficient gas exchange in the mature organism. The first phase of alveologenesis involves the formation of septal ridges (secondary septae) and the second phase involves thinning of the alveolar septa. Within secondary septa, mesenchymal cells include a transient population of alveolar myofibroblasts (MyoFBs) and a stable but poorly described population of lipid-rich cells that have been referred to as lipofibroblasts or matrix fibroblasts (MatFBs). Using a unique Fgf18CreER lineage trace mouse line, cell sorting, single-cell RNA sequencing and primary cell culture, we have identified multiple subtypes of mesenchymal cells in the neonatal lung, including an immature progenitor cell that gives rise to mature MyoFB. We also show that the endogenous and targeted ROSA26 locus serves as a sensitive reporter for MyoFB maturation. These studies identify a MyoFB differentiation program that is distinct from other mesenchymal cell types and increases the known repertoire of mesenchymal cell types in the neonatal lung.


Subject(s)
Animals, Newborn , Cell Differentiation , Lung , Myofibroblasts , Animals , Myofibroblasts/metabolism , Myofibroblasts/cytology , Mice , Lung/cytology , Lung/embryology , Lung/metabolism , Cell Lineage , Organogenesis , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism
2.
Cell ; 150(6): 1121-34, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22980976

ABSTRACT

We report the results of whole-genome and transcriptome sequencing of tumor and adjacent normal tissue samples from 17 patients with non-small cell lung carcinoma (NSCLC). We identified 3,726 point mutations and more than 90 indels in the coding sequence, with an average mutation frequency more than 10-fold higher in smokers than in never-smokers. Novel alterations in genes involved in chromatin modification and DNA repair pathways were identified, along with DACH1, CFTR, RELN, ABCB5, and HGF. Deep digital sequencing revealed diverse clonality patterns in both never-smokers and smokers. All validated EFGR and KRAS mutations were present in the founder clones, suggesting possible roles in cancer initiation. Analysis revealed 14 fusions, including ROS1 and ALK, as well as novel metabolic enzymes. Cell-cycle and JAK-STAT pathways are significantly altered in lung cancer, along with perturbations in 54 genes that are potentially targetable with currently available drugs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Smoking/genetics , Smoking/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Chromosome Aberrations , Female , Gene Expression Profiling , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , INDEL Mutation , Lung Neoplasms/therapy , Male , Molecular Targeted Therapy , Point Mutation , Reelin Protein
3.
Dev Dyn ; 253(2): 233-254, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37688792

ABSTRACT

BACKGROUND: Latent TGFß binding protein-2 (LTBP2) is a fibrillin 1 binding component of the microfibril. LTBP2 is the only LTBP protein that does not bind any isoforms of TGFß, although it may interfere with the function of other LTBPs or interact with other signaling pathways. RESULTS: Here, we investigate mice lacking Ltbp2 (Ltbp2-/- ) and identify multiple phenotypes that impact bodyweight and fat mass, and affect bone and skin development. The alterations in skin and bone development are particularly noteworthy since the strength of these tissues is differentially affected by loss of Ltbp2. Interestingly, some tissues that express high levels of Ltbp2, such as the aorta and lung, do not have a developmental or homeostatic phenotype. CONCLUSIONS: Analysis of these mice show that LTBP2 has complex effects on development through direct effects on the extracellular matrix (ECM) or on signaling pathways that are known to regulate the ECM.


Subject(s)
Carrier Proteins , Extracellular Matrix , Animals , Mice , Carrier Proteins/genetics , Carrier Proteins/metabolism , Extracellular Matrix/metabolism , Phenotype , Transforming Growth Factor beta/metabolism , Protein Isoforms/metabolism , Protein Binding
4.
PLoS Pathog ; 18(6): e1010228, 2022 06.
Article in English | MEDLINE | ID: mdl-35675358

ABSTRACT

Influenza A virus (IAV) preferentially infects conducting airway and alveolar epithelial cells in the lung. The outcome of these infections is impacted by the host response, including the production of various cytokines, chemokines, and growth factors. Fibroblast growth factor-9 (FGF9) is required for lung development, can display antiviral activity in vitro, and is upregulated in asymptomatic patients during early IAV infection. We therefore hypothesized that FGF9 would protect the lungs from respiratory virus infection and evaluated IAV pathogenesis in mice that overexpress FGF9 in club cells in the conducting airway epithelium (FGF9-OE mice). However, we found that FGF9-OE mice were highly susceptible to IAV and Sendai virus infection compared to control mice. FGF9-OE mice displayed elevated and persistent viral loads, increased expression of cytokines and chemokines, and increased numbers of infiltrating immune cells as early as 1 day post-infection (dpi). Gene expression analysis showed an elevated type I interferon (IFN) signature in the conducting airway epithelium and analysis of IAV tropism uncovered a dramatic shift in infection from the conducting airway epithelium to the alveolar epithelium in FGF9-OE lungs. These results demonstrate that FGF9 signaling primes the conducting airway epithelium to rapidly induce a localized IFN and proinflammatory cytokine response during viral infection. Although this response protects the airway epithelial cells from IAV infection, it allows for early and enhanced infection of the alveolar epithelium, ultimately leading to increased morbidity and mortality. Our study illuminates a novel role for FGF9 in regulating respiratory virus infection and pathogenesis.


Subject(s)
Fibroblast Growth Factor 9 , Influenza A virus , Influenza, Human , Interferon Type I , Orthomyxoviridae Infections , Animals , Cytokines/metabolism , Epithelial Cells/metabolism , Fibroblast Growth Factor 9/biosynthesis , Humans , Influenza A virus/metabolism , Influenza, Human/metabolism , Influenza, Human/virology , Interferon Type I/metabolism , Mice , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology
5.
FASEB J ; 37(3): e22777, 2023 03.
Article in English | MEDLINE | ID: mdl-36734881

ABSTRACT

The enthesis is a transitional tissue between tendon and bone that matures postnatally. The development and maturation of the enthesis involve cellular processes likened to an arrested growth plate. In this study, we explored the role of fibroblast growth factor 9 (Fgf9), a known regulator of chondrogenesis and vascularization during bone development, on the structure and function of the postnatal enthesis. First, we confirmed spatial expression of Fgf9 in the tendon and enthesis using in situ hybridization. We then used Cre-lox recombinase to conditionally knockout Fgf9 in mouse tendon and enthesis (Scx-Cre) and characterized enthesis morphology as well as mechanical properties in Fgf9ScxCre and wild-type (WT) entheses. Fgf9ScxCre mice had smaller calcaneal and humeral apophyses, thinner cortical bone at the attachment, increased cellularity, and reduced failure load in mature entheses compared to WT littermates. During postnatal development, we found reduced chondrocyte hypertrophy and disrupted type X collagen (Col X) in Fgf9ScxCre entheses. These findings support that tendon-derived Fgf9 is important for functional development of the enthesis, including its postnatal mineralization. Our findings suggest the potential role of FGF signaling during enthesis development.


Subject(s)
Fibroblast Growth Factor 9 , Tendons , Mice , Animals , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factor 9/metabolism , Tendons/metabolism , Bone and Bones , Bone Development/genetics , Chondrogenesis
6.
Dev Dyn ; 252(9): 1180-1188, 2023 09.
Article in English | MEDLINE | ID: mdl-37212424

ABSTRACT

BACKGROUND: Tendons and ligaments attach to bone are essential for joint mobility and stability in vertebrates. Tendon and ligament attachments (ie, entheses) are found at bony protrusions (ie, eminences), and the shape and size of these protrusions depend on both mechanical forces and cellular cues during growth. Tendon eminences also contribute to mechanical leverage for skeletal muscle. Fibroblast growth factor receptor (FGFR) signaling plays a critical role in bone development, and Fgfr1 and Fgfr2 are highly expressed in the perichondrium and periosteum of bone where entheses can be found. RESULTS AND CONCLUSIONS: We used transgenic mice for combinatorial knockout of Fgfr1 and/or Fgfr2 in tendon/attachment progenitors (ScxCre) and measured eminence size and shape. Conditional deletion of both, but not individual, Fgfr1 and Fgfr2 in Scx progenitors led to enlarged eminences in the postnatal skeleton and shortening of long bones. In addition, Fgfr1/Fgfr2 double conditional knockout mice had more variation collagen fibril size in tendon, decreased tibial slope, and increased cell death at ligament attachments. These findings identify a role for FGFR signaling in regulating growth and maintenance of tendon/ligament attachments and the size and shape of bony eminences.


Subject(s)
Bone and Bones , Tendons , Animals , Mice , Cell Death/genetics , Mice, Knockout , Mice, Transgenic , Stem Cells , Tendons/metabolism
7.
Am J Physiol Heart Circ Physiol ; 325(2): H203-H231, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37204871

ABSTRACT

Heart failure (HF) is a leading cause of morbidity and mortality particularly in older adults and patients with multiple metabolic comorbidities. Heart failure with preserved ejection fraction (HFpEF) is a clinical syndrome with multisystem organ dysfunction in which patients develop symptoms of HF as a result of high left ventricular (LV) diastolic pressure in the context of normal or near normal LV ejection fraction (LVEF; ≥50%). Challenges to create and reproduce a robust rodent phenotype that recapitulates the multiple comorbidities that exist in this syndrome explain the presence of various animal models that fail to satisfy all the criteria of HFpEF. Using a continuous infusion of angiotensin II and phenylephrine (ANG II/PE), we demonstrate a strong HFpEF phenotype satisfying major clinically relevant manifestations and criteria of this pathology, including exercise intolerance, pulmonary edema, concentric myocardial hypertrophy, diastolic dysfunction, histological signs of microvascular impairment, and fibrosis. Conventional echocardiographic analysis of diastolic dysfunction identified early stages of HFpEF development and speckle tracking echocardiography analysis including the left atrium (LA) identified strain abnormalities indicative of contraction-relaxation cycle impairment. Diastolic dysfunction was validated by retrograde cardiac catheterization and analysis of LV end-diastolic pressure (LVEDP). Among mice that developed HFpEF, two major subgroups were identified with predominantly perivascular fibrosis and interstitial myocardial fibrosis. In addition to major phenotypic criteria of HFpEF that were evident at early stages of this model (3 and 10 days), accompanying RNAseq data demonstrate activation of pathways associated with myocardial metabolic changes, inflammation, activation of extracellular matrix (ECM) deposition, microvascular rarefaction, and pressure- and volume-related myocardial stress.NEW & NOTEWORTHY Heart failure with preserved ejection fraction (HFpEF) is an emerging epidemic affecting up to half of patients with heart failure. Here we used a chronic angiotensin II/phenylephrine (ANG II/PE) infusion model and instituted an updated algorithm for HFpEF assessment. Given the simplicity in generating this model, it may become a useful tool for investigating pathogenic mechanisms, identification of diagnostic markers, and for drug discovery aimed at both prevention and treatment of HFpEF.


Subject(s)
Cardiomyopathies , Heart Failure , Animals , Mice , Heart Failure/drug therapy , Stroke Volume/physiology , Angiotensin II , Ventricular Function, Left/physiology , Disease Models, Animal , Fibrosis , Phenylephrine
8.
Development ; 147(2)2020 01 17.
Article in English | MEDLINE | ID: mdl-31862844

ABSTRACT

Alveologenesis is an essential developmental process that increases the surface area of the lung through the formation of septal ridges. In the mouse, septation occurs postnatally and is thought to require the alveolar myofibroblast (AMF). Though abundant during alveologenesis, markers for AMFs are minimally detected in the adult. After septation, the alveolar walls thin to allow efficient gas exchange. Both loss of AMFs or retention and differentiation into another cell type during septal thinning have been proposed. Using a novel Fgf18:CreERT2 allele to lineage trace AMFs, we demonstrate that most AMFs are developmentally cleared during alveologenesis. Lung mesenchyme also contains other poorly described cell types, including alveolar lipofibroblasts (ALF). We show that Gli1:CreERT2 marks both AMFs as well as ALFs, and lineage tracing shows that ALFs are retained in adult alveoli while AMFs are lost. We further show that multiple immune cell populations contain lineage-labeled particles, suggesting a phagocytic role in the clearance of AMFs. The demonstration that the AMF lineage is depleted during septal thinning through a phagocytic process provides a mechanism for the clearance of a transient developmental cell population.


Subject(s)
Fibroblast Growth Factors/metabolism , Myofibroblasts/metabolism , Organogenesis , Pulmonary Alveoli/growth & development , Animals , Animals, Newborn , Cell Lineage , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Mesoderm/cytology , Mice, Inbred C57BL , Models, Biological , Myofibroblasts/cytology , Phagocytosis , Time Factors
9.
Development ; 147(21)2020 10 19.
Article in English | MEDLINE | ID: mdl-32907848

ABSTRACT

The closure of the embryonic ventral body wall in amniotes is an important morphogenetic event and is essential for life. Defects in human ventral wall closure are a major class of birth defect and a significant health burden. Despite this, very little is understood about how the ventral body wall is formed. Here, we show that fibroblast growth factor (FGF) ligands FGF8, FGF17 and FGF18 are essential for this process. Conditional mouse mutants for these genes display subtle migratory defects in the abdominal muscles of the ventral body wall and an enlarged umbilical ring, through which the internal organs are extruded. By refining where and when these genes are required using different Cre lines, we show that Fgf8 and Fgf17 are required in the presomitic mesoderm, whereas Fgf18 is required in the somites. This study identifies complex and multifactorial origins of ventral wall defects and has important implications for understanding their origins during embryonic development.


Subject(s)
Body Patterning , Embryo, Mammalian/metabolism , Fibroblast Growth Factor 8/metabolism , Fibroblast Growth Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning/genetics , Female , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Hernia, Umbilical , Male , Mice , Models, Biological , Morphogenesis , Mutation/genetics , Organ Specificity , Protein Domains , Somites/metabolism , Stem Cells/metabolism
10.
Cell Mol Life Sci ; 79(12): 609, 2022 Nov 29.
Article in English | MEDLINE | ID: mdl-36445537

ABSTRACT

The specification, characterization, and fate of alveolar type 1 and type 2 (AT1 and AT2) progenitors during embryonic lung development are poorly defined. Current models of distal epithelial lineage formation fail to capture the heterogeneity and dynamic contribution of progenitor pools present during early development. Furthermore, few studies explore the pathways involved in alveolar progenitor specification and fate. In this paper, we build upon our previously published work on the regulation of airway epithelial progenitors by fibroblast growth factor receptor 2b (FGFR2b) signalling during early (E12.5) and mid (E14.5) pseudoglandular stage lung development. Our results suggest that a significant proportion of AT2 and AT1 progenitors are lineage-flexible during late pseudoglandular stage development, and that lineage commitment is regulated in part by FGFR2b signalling. We have characterized a set of direct FGFR2b targets at E16.5 which are likely involved in alveolar lineage formation. These signature genes converge on a subpopulation of AT2 cells later in development and are downregulated in AT2 cells transitioning to the AT1 lineage during repair after injury in adults. Our findings highlight the extensive heterogeneity of pneumocytes by elucidating the role of FGFR2b signalling in these cells during early airway epithelial lineage formation, as well as during repair after injury.


Subject(s)
Alveolar Epithelial Cells , Lung , Receptor, Fibroblast Growth Factor, Type 2 , Stem Cells , Animals , Mice , Embryonic Development , Receptor, Fibroblast Growth Factor, Type 2/genetics , Signal Transduction , Lung/embryology , Cell Lineage
11.
Genes Dev ; 29(14): 1463-86, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26220993

ABSTRACT

Fibroblast growth factor (FGF) signaling pathways are essential regulators of vertebrate skeletal development. FGF signaling regulates development of the limb bud and formation of the mesenchymal condensation and has key roles in regulating chondrogenesis, osteogenesis, and bone and mineral homeostasis. This review updates our review on FGFs in skeletal development published in Genes & Development in 2002, examines progress made on understanding the functions of the FGF signaling pathway during critical stages of skeletogenesis, and explores the mechanisms by which mutations in FGF signaling molecules cause skeletal malformations in humans. Links between FGF signaling pathways and other interacting pathways that are critical for skeletal development and could be exploited to treat genetic diseases and repair bone are also explored.


Subject(s)
Bone Diseases/genetics , Bone and Bones/embryology , Bone and Bones/physiopathology , Fibroblast Growth Factors/physiology , Signal Transduction , Animals , Bone Diseases/therapy , Bone Regeneration/genetics , Chondrogenesis , Fibroblast Growth Factors/genetics , Humans , Minerals/metabolism , Mutation , Osteogenesis
12.
PLoS Genet ; 15(7): e1008254, 2019 07.
Article in English | MEDLINE | ID: mdl-31276493

ABSTRACT

The mouse organ of Corti, housed inside the cochlea, contains hair cells and supporting cells that transduce sound into electrical signals. These cells develop in two main steps: progenitor specification followed by differentiation. Fibroblast Growth Factor (FGF) signaling is important in this developmental pathway, as deletion of FGF receptor 1 (Fgfr1) or its ligand, Fgf20, leads to the loss of hair cells and supporting cells from the organ of Corti. However, whether FGF20-FGFR1 signaling is required during specification or differentiation, and how it interacts with the transcription factor Sox2, also important for hair cell and supporting cell development, has been a topic of debate. Here, we show that while FGF20-FGFR1 signaling functions during progenitor differentiation, FGFR1 has an FGF20-independent, Sox2-dependent role in specification. We also show that a combination of reduction in Sox2 expression and Fgf20 deletion recapitulates the Fgfr1-deletion phenotype. Furthermore, we uncovered a strong genetic interaction between Sox2 and Fgf20, especially in regulating the development of hair cells and supporting cells towards the basal end and the outer compartment of the cochlea. To explain this genetic interaction and its effects on the basal end of the cochlea, we provide evidence that decreased Sox2 expression delays specification, which begins at the apex of the cochlea and progresses towards the base, while Fgf20-deletion results in premature onset of differentiation, which begins near the base of the cochlea and progresses towards the apex. Thereby, Sox2 and Fgf20 interact to ensure that specification occurs before differentiation towards the cochlear base. These findings reveal an intricate developmental program regulating organ of Corti development along the basal-apical axis of the cochlea.


Subject(s)
Fibroblast Growth Factors/genetics , Organ of Corti/cytology , Receptor, Fibroblast Growth Factor, Type 1/genetics , SOXB1 Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Fibroblast Growth Factors/metabolism , Gene Knockout Techniques , Male , Mice , Organ of Corti/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction
13.
Proc Natl Acad Sci U S A ; 116(12): 5570-5575, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30819894

ABSTRACT

Testes and ovaries undergo sex-specific morphogenetic changes and adopt strikingly different morphologies, despite the fact that both arise from a common precursor, the bipotential gonad. Previous studies showed that recruitment of vasculature is critical for testis patterning. However, vasculature is not recruited into the early ovary. Peripheral innervation is involved in patterning development of many organs but has been given little attention in gonad development. In this study, we show that while innervation in the male reproductive complex is restricted to the epididymis and vas deferens and never invades the interior of the testis, neural crest-derived innervation invades the interior of the ovary around E16.5. Individual neural crest cells colonize the ovary, differentiate into neurons and glia, and form a dense neural network within the ovarian medulla. Using a sex-reversing mutant mouse line, we show that innervation is specific to ovary development, is not dependent on the genetic sex of gonadal or neural crest cells, and may be blocked by repressive guidance signals elevated in the male pathway. This study reveals another aspect of sexually dimorphic gonad development, establishes a precise timeline and structure of ovarian innervation, and raises many questions for future research.


Subject(s)
Gonads/innervation , Ovary/innervation , Testis/innervation , Animals , Female , Gene Expression Regulation, Developmental/genetics , Gonads/cytology , Male , Mice , Mice, Inbred C57BL , Morphogenesis , Neural Crest/innervation , Neurons/physiology , Ovary/cytology , Sex Characteristics , Sex Determination Processes/genetics , Sex Determination Processes/physiology , Sex Differentiation/genetics , Testis/cytology
14.
Proc Natl Acad Sci U S A ; 116(42): 21054-21060, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31570588

ABSTRACT

Development of multicellular organs requires the coordination of cell differentiation and patterning. Critical for sound detection, the mammalian organ of Corti contains functional units arranged tonotopically along the cochlear turns. Each unit consists of sensory hair cells intercalated by nonsensory supporting cells, both specified and radially patterned with exquisite precision during embryonic development. However, how cell identity and radial patterning are jointly controlled is poorly understood. Here we show that ß-catenin is required for specification of hair cell and supporting cell subtypes and radial patterning of the cochlea in vivo. In 2 mouse models of conditional ß-catenin deletion, early specification of Myosin7-expressing hair cells and Prox1-positive supporting cells was preserved. While ß-catenin-deficient cochleae expressed FGF8 and FGFR3, both of which are essential for pillar cell specification, the radial patterning of organ of Corti was disrupted, revealed by aberrant expression of cadherins and the pillar cell markers P75 and Lgr6. Moreover, ß-catenin ablation caused duplication of FGF8-positive inner hair cells and reduction of outer hair cells without affecting the overall hair cell density. In contrast, in another transgenic model with suppressed transcriptional activity of ß-catenin but preserved cell adhesion function, both specification and radial patterning of the organ of Corti were intact. Our study reveals specific functions of ß-catenin in governing cell identity and patterning mediated through cell adhesion in the developing cochlea.


Subject(s)
Cochlea/metabolism , Cochlea/physiology , beta Catenin/metabolism , Animals , Biomarkers/metabolism , Cell Adhesion/physiology , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/physiology , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/physiology , Mice , Organ of Corti/metabolism , Organogenesis/physiology
15.
Dev Dyn ; 250(2): 134-144, 2021 02.
Article in English | MEDLINE | ID: mdl-32735383

ABSTRACT

BACKGROUND: Fibroblast Growth Factor 20 (FGF20)-FGF receptor 1 (FGFR1) signaling is essential for cochlear hair cell (HC) and supporting cell (SC) differentiation. In other organ systems, FGFR1 signals through several intracellular pathways including MAPK (ERK), PI3K, phospholipase C ɣ (PLCɣ), and p38. Previous studies implicated MAPK and PI3K pathways in HC and SC development. We hypothesized that one or both would be important downstream mediators of FGF20-FGFR1 signaling for HC differentiation. RESULTS: By inhibiting pathways downstream of FGFR1 in cochlea explant cultures, we established that both MAPK and PI3K pathways are required for HC differentiation while PLCɣ and p38 pathways are not. Examining the canonical PI3K pathway, we found that while AKT is necessary for HC differentiation, it is not sufficient to rescue the Fgf20-/- phenotype. To determine whether PI3K functions downstream of FGF20, we inhibited Phosphatase and Tensin Homolog (PTEN) in Fgf20-/- explants. Overactivation of PI3K resulted in a partial rescue of the Fgf20-/- phenotype, demonstrating a requirement for PI3K downstream of FGF20. Consistent with a requirement for the MAPK pathway for FGF20-regulated HC differentiation, we show that treating Fgf20-/- explants with FGF9 increased levels of dpERK. CONCLUSIONS: Together, these data provide evidence that both MAPK and PI3K are important downstream mediators of FGF20-FGFR1 signaling during HC and SC differentiation.


Subject(s)
Cell Differentiation , Fibroblast Growth Factors/metabolism , MAP Kinase Signaling System , Organ of Corti/growth & development , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Female , Fibroblast Growth Factor 9 , Male , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Organ of Corti/cytology , Organ of Corti/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Dev Dyn ; 250(12): 1778-1795, 2021 12.
Article in English | MEDLINE | ID: mdl-34091985

ABSTRACT

BACKGROUND: The growth of most bony tuberosities, like the deltoid tuberosity (DT), rely on the transmission of muscle forces at the tendon-bone attachment during skeletal growth. Tuberosities distribute muscle forces and provide mechanical leverage at attachment sites for joint stability and mobility. The genetic factors that regulate tuberosity growth remain largely unknown. In mouse embryos with global deletion of fibroblast growth factor 9 (Fgf9), the DT size is notably enlarged. In this study, we explored the tissue-specific regulation of DT size using both global and targeted deletion of Fgf9. RESULTS: We showed that cell hypertrophy and mineralization dynamics of the DT, as well as transcriptional signatures from skeletal muscle but not bone, were influenced by the global loss of Fgf9. Loss of Fgf9 during embryonic growth led to increased chondrocyte hypertrophy and reduced cell proliferation at the DT attachment site. This endured hypertrophy and limited proliferation may explain the abnormal mineralization patterns and locally dysregulated expression of markers of endochondral development in Fgf9null attachments. We then showed that targeted deletion of Fgf9 in skeletal muscle leads to postnatal enlargement of the DT. CONCLUSION: Taken together, we discovered that Fgf9 may play an influential role in muscle-bone cross-talk during embryonic and postnatal development.


Subject(s)
Bone Diseases/genetics , Fibroblast Growth Factor 9/genetics , Muscle, Skeletal/metabolism , Tendons/pathology , Animals , Animals, Newborn , Bone Development/genetics , Bone Diseases/pathology , Chondrogenesis/genetics , Embryo, Mammalian , Female , Fibroblast Growth Factor 9/metabolism , Gene Deletion , Hypertrophy/genetics , Hypertrophy/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity/genetics , Osteogenesis/genetics , Pregnancy , Tendons/embryology , Tendons/growth & development , Tendons/metabolism
17.
Dev Biol ; 464(1): 11-23, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32450229

ABSTRACT

Development of the complex structure of the vertebrate limb requires carefully orchestrated interactions between multiple regulatory pathways and proteins. Among these, precise regulation of 5' Hox transcription factor expression is essential for proper limb bud patterning and elaboration of distinct limb skeletal elements. Here, we identified Geminin (Gmnn) as a novel regulator of this process. A conditional model of Gmnn deficiency resulted in loss or severe reduction of forelimb skeletal elements, while both the forelimb autopod and hindlimb were unaffected. 5' Hox gene expression expanded into more proximal and anterior regions of the embryonic forelimb buds in this Gmnn-deficient model. A second conditional model of Gmnn deficiency instead caused a similar but less severe reduction of hindlimb skeletal elements and hindlimb polydactyly, while not affecting the forelimb. An ectopic posterior SHH signaling center was evident in the anterior hindlimb bud of Gmnn-deficient embryos in this model. This center ectopically expressed Hoxd13, the HOXD13 target Shh, and the SHH target Ptch1, while these mutant hindlimb buds also had reduced levels of the cleaved, repressor form of GLI3, a SHH pathway antagonist. Together, this work delineates a new role for Gmnn in modulating Hox expression to pattern the vertebrate limb.


Subject(s)
Embryo, Mammalian/embryology , Geminin/metabolism , Gene Expression Regulation, Developmental , Hindlimb/embryology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Embryo, Mammalian/cytology , Geminin/genetics , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hindlimb/cytology , Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Patched-1 Receptor/genetics , Patched-1 Receptor/metabolism , Transcription Factors/genetics
18.
Growth Factors ; 39(1-6): 14-27, 2021.
Article in English | MEDLINE | ID: mdl-34879776

ABSTRACT

Utilising rabbit corneal endothelial cells (CEC) in three different paradigms, two human FGF1 derivatives (TTHX1001 and TTHX1114), engineered to exhibit greater stability, were tested as proliferative agents. Primary CECs and mouse NIH 3T3 cells treated with the two FGF1 derivatives showed equivalent EC50 ranges (3.3-24 vs.1.9-16. ng/mL) and, in organ culture, chemically lesioned corneas regained half of the lost endothelial layer in three days after treatment with the FGF1 derivatives as compared to controls. In vivo, following cryolesioning, the CEC monolayer, as judged by specular microscopy, regenerated 10-11 days faster when treated with TTHX1001. Over two weeks, all treated eyes showed clearing of opacity about twice that of untreated controls. In all three rabbit models, both FGF1 derivatives were effective in inducing CEC proliferation over control conditions, supporting the prediction that these stabilised FGF1 derivatives can potentially regenerate corneal endothelial deficits in humans.


Subject(s)
Endothelial Cells , Fibroblast Growth Factor 1 , Animals , Cells, Cultured , Cornea , Endothelium, Corneal/metabolism , Fibroblast Growth Factor 1/pharmacology , Mice , Rabbits
19.
BMC Biol ; 18(1): 87, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32664967

ABSTRACT

BACKGROUND: The formation of supernumerary teeth is an excellent model for studying the molecular mechanisms that control stem/progenitor cell homeostasis needed to generate a renewable source of replacement cells and tissues. Although multiple growth factors and transcriptional factors have been associated with supernumerary tooth formation, the regulatory inputs of extracellular matrix in this regenerative process remains poorly understood. RESULTS: In this study, we present evidence that disrupting glycosaminoglycans (GAGs) in the dental epithelium of mice by inactivating FAM20B, a xylose kinase essential for GAG assembly, leads to supernumerary tooth formation in a pattern reminiscent of replacement teeth. The dental epithelial GAGs confine murine tooth number by restricting the homeostasis of Sox2(+) dental epithelial stem/progenitor cells in a non-autonomous manner. FAM20B-catalyzed GAGs regulate the cell fate of dental lamina by restricting FGFR2b signaling at the initial stage of tooth development to maintain a subtle balance between the renewal and differentiation of Sox2(+) cells. At the later cap stage, WNT signaling functions as a relay cue to facilitate the supernumerary tooth formation. CONCLUSIONS: The novel mechanism we have characterized through which GAGs control the tooth number in mice may also be more broadly relevant for potentiating signaling interactions in other tissues during development and tissue homeostasis.


Subject(s)
Glycosaminoglycans/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Signal Transduction , Tooth, Supernumerary/genetics , Animals , Cell Differentiation , Mice , Odontogenesis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Stem Cells/metabolism
20.
Genes Dev ; 27(4): 450-8, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23431057

ABSTRACT

In hair follicle development, a placode-derived signal is believed to induce formation of the dermal condensation, an essential component of ectodermal organs. However, the identity of this signal is unknown. Furthermore, although induction and patterning of hair follicles are intimately linked, it is not known whether the mesenchymal condensation is necessary for inducing the initial epithelial pattern. Here, we show that fibroblast growth factor 20 (Fgf20) is expressed in hair placodes and is induced by and functions downstream from epithelial ectodysplasin (Eda)/Edar and Wnt/ß-Catenin signaling to initiate formation of the underlying dermal condensation. Fgf20 governs formation of primary and secondary dermal condensations in developing hair follicles and subsequent formation of guard, awl, and auchene hairs. Although primary dermal condensations are absent in Fgf20 mutant mice, a regular array of hair placodes is formed, demonstrating that the epithelial patterning process is independent of known histological and molecular markers of underlying mesenchymal patterns during the initial stages of hair follicle development.


Subject(s)
Fibroblast Growth Factors/metabolism , Hair Follicle/embryology , Animals , Ectodysplasins/metabolism , Edar Receptor/metabolism , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Mice , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL