Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
J Clin Invest ; 131(17)2021 09 01.
Article in English | MEDLINE | ID: mdl-34623323

ABSTRACT

Hypoxia-induced pulmonary hypertension (PH) is one of the most common and deadliest forms of PH. Fibroblast growth factor receptors 1 and 2 (FGFR1/2) are elevated in patients with PH and in mice exposed to chronic hypoxia. Endothelial FGFR1/2 signaling is important for the adaptive response to several injury types and we hypothesized that endothelial FGFR1/2 signaling would protect against hypoxia-induced PH. Mice lacking endothelial FGFR1/2, mice with activated endothelial FGFR signaling, and human pulmonary artery endothelial cells (HPAECs) were challenged with hypoxia. We assessed the effect of FGFR activation and inhibition on right ventricular pressure, vascular remodeling, and endothelial-mesenchymal transition (EndMT), a known pathologic change seen in patients with PH. Hypoxia-exposed mice lacking endothelial FGFRs developed increased PH, while mice overexpressing a constitutively active FGFR in endothelial cells did not develop PH. Mechanistically, lack of endothelial FGFRs or inhibition of FGFRs in HPAECs led to increased TGF-ß signaling and increased EndMT in response to hypoxia. These phenotypes were reversed in mice with activated endothelial FGFR signaling, suggesting that FGFR signaling inhibits TGF-ß pathway-mediated EndMT during chronic hypoxia. Consistent with these observations, lung tissue from patients with PH showed activation of FGFR and TGF-ß signaling. Collectively, these data suggest that activation of endothelial FGFR signaling could be therapeutic for hypoxia-induced PH.


Subject(s)
Fibroblast Growth Factors/metabolism , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Animals , Endothelium/metabolism , Endothelium/pathology , Female , Humans , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/prevention & control , Hypoxia/complications , Male , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Knockout , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Vascular Remodeling
2.
J Clin Invest ; 131(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-33983905

ABSTRACT

Synovial sarcoma is an aggressive malignancy with no effective treatments for patients with metastasis. The synovial sarcoma fusion SS18-SSX, which recruits the SWI/SNF-BAF chromatin remodeling and polycomb repressive complexes, results in epigenetic activation of FGF receptor (FGFR) signaling. In genetic FGFR-knockout models, culture, and xenograft synovial sarcoma models treated with the FGFR inhibitor BGJ398, we show that FGFR1, FGFR2, and FGFR3 were crucial for tumor growth. Transcriptome analyses of BGJ398-treated cells and histological and expression analyses of mouse and human synovial sarcoma tumors revealed prevalent expression of two ETS factors and FGFR targets, ETV4 and ETV5. We further demonstrate that ETV4 and ETV5 acted as drivers of synovial sarcoma growth, most likely through control of the cell cycle. Upon ETV4 and ETV5 knockdown, we observed a striking upregulation of DUX4 and its transcriptional targets that activate the zygotic genome and drive the atrophy program in facioscapulohumeral dystrophy patients. In addition to demonstrating the importance of inhibiting all three FGFRs, the current findings reveal potential nodes of attack for the cancer with the discovery of ETV4 and ETV5 as appropriate biomarkers and molecular targets, and activation of the embryonic DUX4 pathway as a promising approach to block synovial sarcoma tumors.


Subject(s)
Proto-Oncogene Proteins c-ets/metabolism , Sarcoma, Synovial/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Heterografts , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenylurea Compounds/pharmacology , Proto-Oncogene Proteins c-ets/genetics , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Sarcoma, Synovial/genetics , Sarcoma, Synovial/pathology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
3.
J Neurosci ; 37(50): 12094-12105, 2017 12 13.
Article in English | MEDLINE | ID: mdl-29097598

ABSTRACT

Fibroblast growth factors (FGFs) and FGF receptors (FGFRs) are known for their potent effects on cell proliferation/differentiation and cortical patterning in the developing brain. However, little is known regarding the roles of FGFs/FGFRs in cortical circuit formation. Here we show that Fgfr1/2/3 and Fgf7/9/10/22 mRNAs are expressed in the developing primary somatosensory (S1) barrel cortex. Barrel cortex layer IV spiny stellate cells (bSCs) are the primary recipients of ascending sensory information via thalamocortical axons (TCAs). Detail quantification revealed distinctive phases for bSC dendritogenesis: orienting dendrites toward TCAs, adding de novo dendritic segments, and elongating dendritic length, while maintaining dendritic patterns. Deleting Fgfr1/2/3 in bSCs had minimal impact on dendritic polarity but transiently increased the number of dendritic segments. However, 6 d later, FGFR1/2/3 loss of function reduced dendritic branch numbers. These data suggest that FGFs/FGFRs have a role in stabilizing dendritic patterning. Depolarization of cultured mouse cortical neurons upregulated the levels of several Fgf/Fgfr mRNAs within 2 h. In vivo, within 6 h of systemic kainic acid administration at postnatal day 6, mRNA levels of Fgf9, Fgf10, Fgfr2c, and Fgfr3b in S1 cortices were enhanced, and this was accompanied by exuberant dendritogenesis of bSCs by 24 h. Deleting Fgfr1/2/3 abolished kainic acid-induced bSC dendritic overgrowth. Finally, FGF9/10 gain of function also resulted in extensive dendritogenesis. Together, our data suggest that FGFs/FGFRs can be regulated by glutamate transmission to modulate/stabilize bSC dendritic complexity. Both male and female mice were used for our study.SIGNIFICANCE STATEMENT Glutamatergic transmission plays critical roles in cortical circuit formation. Its dysregulation has been proposed as a core factor in the etiology of many neurological diseases. We found that excessive glutamate transmission upregulated mRNA expression of Fgfrs and their ligands Fgfs Deleting Fgfr1/2/3 not only impaired bSC dendritogenesis but also abolished glutamate transmission-induced dendritic overgrowth. Overexpressing FGF9 or FGF10 in cortical glutamatergic neurons results in excessive dendritic outgrowth within 24 h, resembling the changes induced by excessive glutamate transmission. Our findings provide strong evidence for the physiological role of fibroblast growth factors (FGFs) and FGF receptors (FGFRs) in establishing and maintaining cortical circuits. Perturbing the expression levels of FGFs/FGFRs by excessive glutamatergic neurotransmission could lead to abnormal neuronal circuits, which may contribute to neurological and psychiatric disease.


Subject(s)
Dendrites/physiology , Fibroblast Growth Factors/physiology , Nerve Tissue Proteins/physiology , Neurogenesis , Receptors, Fibroblast Growth Factor/physiology , Somatosensory Cortex/embryology , Vibrissae/innervation , Animals , Cells, Cultured , Convulsants/toxicity , Electroporation , Female , Fibroblast Growth Factors/genetics , Gain of Function Mutation , Kainic Acid/toxicity , Loss of Function Mutation , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Mice, Transgenic , Neurogenesis/drug effects , Neurons/cytology , Pregnancy , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics , Recombinant Fusion Proteins/metabolism , Somatosensory Cortex/cytology , Somatosensory Cortex/drug effects
4.
Dev Biol ; 415(1): 87-97, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27131625

ABSTRACT

Cell death can have both cell autonomous and non-autonomous roles in normal development. Previous studies have shown that the central cell death regulators grim and reaper are required for the developmentally important elimination of stem cells and neurons in the developing central nervous system (CNS). Here we show that cell death in the nervous system is also required for normal muscle development. In the absence of grim and reaper, there is an increase in the number of fibers in the ventral abdominal muscles in the Drosophila adult. This phenotype can be partially recapitulated by inhibition of cell death specifically in the CNS, indicating a non-autonomous role for neuronal death in limiting muscle fiber number. We also show that FGFs produced in the cell death defective nervous system are required for the increase in muscle fiber number. Cell death in the muscle lineage during pupal stages also plays a role in specifying fiber number. Our work suggests that FGFs from the CNS act as a survival signal for muscle founder cells. Thus, proper muscle fiber specification requires cell death in both the nervous system and in the developing muscle itself.


Subject(s)
Apoptosis/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Muscle Cells/ultrastructure , Muscle Development , Neuropeptides/physiology , Animals , Cell Count , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Fibroblast Growth Factors/physiology , Gene Expression Regulation, Developmental , Glutamates/physiology , Larva , Luminescent Proteins/analysis , Motor Neurons/cytology , Muscles/innervation , Myoblasts/cytology , Neuropeptides/deficiency , Neuropeptides/genetics , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/physiology , Pupa , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/physiology , Sequence Deletion
5.
Blood ; 122(24): 3993-4001, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24106206

ABSTRACT

Beyond its well-established roles in mediating leukocyte rolling, E-selectin is emerging as a multifunctional receptor capable of inducing integrin activation in neutrophils, and of regulating various biological processes in hematopoietic precursors. Although these effects suggest important homeostatic contributions of this selectin in the immune and hematologic systems, the ligands responsible for transducing these effects in different leukocyte lineages are not well defined. We have characterized mice deficient in E-selectin ligand-1 (ESL-1), or in both P-selectin glycoprotein-1 (PSGL-1) and ESL-1, to explore and compare the contributions of these glycoproteins in immune and hematopoietic cell trafficking. In the steady state, ESL-1 deficiency resulted in a moderate myeloid expansion that became more prominent when both glycoproteins were eliminated. During inflammation, PSGL-1 dominated E-selectin binding, rolling, integrin activation, and extravasation of mature neutrophils, but only the combined deficiency in PSGL-1 and ESL-1 completely abrogated leukocyte recruitment. Surprisingly, we find that the levels of ESL-1 were strongly elevated in hematopoietic progenitor cells. These elevations correlated with a prominent function of ESL-1 for E-selectin binding and for migration of hematopoietic progenitor cells into the bone marrow. Our results uncover dominant roles for ESL-1 in the immature compartment, and a functional shift toward PSGL-1 dependence in mature neutrophils.


Subject(s)
Hematopoietic Stem Cells/immunology , Inflammation/immunology , Receptors, Fibroblast Growth Factor/immunology , Sialoglycoproteins/immunology , Animals , Blotting, Western , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Movement/immunology , E-Selectin/metabolism , Female , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Inflammation/genetics , Inflammation/metabolism , Leukocyte Rolling/genetics , Leukocyte Rolling/immunology , Leukocytes/immunology , Leukocytes/metabolism , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism , Peritonitis/genetics , Peritonitis/immunology , Peritonitis/metabolism , Protein Binding/immunology , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics , Sialoglycoproteins/deficiency , Sialoglycoproteins/genetics
6.
Proc Natl Acad Sci U S A ; 110(18): 7336-41, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23589896

ABSTRACT

TGF-ß is abundantly produced in the skeletal system and plays a crucial role in skeletal homeostasis. E-selectin ligand-1 (ESL-1), a Golgi apparatus-localized protein, acts as a negative regulator of TGF-ß bioavailability by attenuating maturation of pro-TGF-ß during cartilage homeostasis. However, whether regulation of intracellular TGF-ß maturation by ESL-1 is also crucial during bone homeostasis has not been well defined. Here, we show that Esl-1(-/-) mice exhibit a severe osteopenia with elevated bone resorption and decreased bone mineralization. In primary culture, Esl-1(-/-) osteoclast progenitors show no difference in osteoclastogenesis. However, Esl-1(-/-) osteoblasts show delayed differentiation and mineralization and stimulate osteoclastogenesis more potently in the osteoblast-osteoclast coculture, suggesting that ESL-1 primarily acts in osteoblasts to regulate bone homeostasis. In addition, Esl-1(-/-) calvaria exhibit an elevated mature TGF-ß/pro-TGF-ß ratio, with increased expression of TGF-ß downstream targets (plasminogen activator inhibitor-1, parathyroid hormone-related peptide, connective tissue growth factor, and matrix metallopeptidase 13, etc.) and a key regulator of osteoclastogenesis (receptor activator of nuclear factor κB ligand). Moreover, in vivo treatment with 1D11, a pan-TGF-ß antibody, significantly improved the low bone mass of Esl-1(-/-) mice, suggesting that elevated TGF-ß signaling is the major cause of osteopenia in Esl-1(-/-) mice. In summary, our study identifies ESL-1 as an important regulator of bone remodeling and demonstrates that the modulation of TGF-ß maturation is pivotal in the maintenance of a homeostatic bone microenvironment and for proper osteoblast-osteoclast coupling.


Subject(s)
Bone Remodeling , Receptors, Fibroblast Growth Factor/metabolism , Sialoglycoproteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antibodies/pharmacology , Bone Diseases, Metabolic/complications , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Bone Diseases, Metabolic/physiopathology , Bone Remodeling/drug effects , Bone Remodeling/genetics , Bone Resorption/complications , Bone Resorption/genetics , Bone Resorption/pathology , Bone Resorption/physiopathology , Calcification, Physiologic/drug effects , Calcification, Physiologic/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage/drug effects , Cell Lineage/genetics , Cells, Cultured , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Femur/physiopathology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Mice , Organ Size/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis/drug effects , Osteogenesis/genetics , Phenotype , Radiography , Receptors, Fibroblast Growth Factor/deficiency , Sialoglycoproteins/deficiency , Signal Transduction/genetics
7.
Nature ; 494(7438): 476-9, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23426263

ABSTRACT

Schistosomiasis is among the most prevalent human parasitic diseases, affecting more than 200 million people worldwide. The aetiological agents of this disease are trematode flatworms (Schistosoma) that live and lay eggs within the vasculature of the host. These eggs lodge in host tissues, causing inflammatory responses that are the primary cause of morbidity. Because these parasites can live and reproduce within human hosts for decades, elucidating the mechanisms that promote their longevity is of fundamental importance. Although adult pluripotent stem cells, called neoblasts, drive long-term homeostatic tissue maintenance in long-lived free-living flatworms (for example, planarians), and neoblast-like cells have been described in some parasitic tapeworms, little is known about whether similar cell types exist in any trematode species. Here we describe a population of neoblast-like cells in the trematode Schistosoma mansoni. These cells resemble planarian neoblasts morphologically and share their ability to proliferate and differentiate into derivatives of multiple germ layers. Capitalizing on available genomic resources and RNA-seq-based gene expression profiling, we find that these schistosome neoblast-like cells express a fibroblast growth factor receptor orthologue. Using RNA interference we demonstrate that this gene is required for the maintenance of these neoblast-like cells. Our observations indicate that adaptation of developmental strategies shared by free-living ancestors to modern-day schistosomes probably contributed to the success of these animals as long-lived obligate parasites. We expect that future studies deciphering the function of these neoblast-like cells will have important implications for understanding the biology of these devastating parasites.


Subject(s)
Adult Stem Cells/cytology , Parasites/cytology , Pluripotent Stem Cells/cytology , Schistosoma mansoni/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Female , Gene Expression Profiling , Genes, Helminth/genetics , Helminth Proteins/genetics , Helminth Proteins/metabolism , Humans , Male , Mice , Pluripotent Stem Cells/metabolism , RNA Interference , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Schistosomiasis mansoni/parasitology
8.
Atherosclerosis ; 224(2): 363-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22939356

ABSTRACT

E-selectin-1 (ESL-1), also known as golgi complex-localized glycoprotein-1 (GLG1), homocysteine-rich fibroblast growth factor receptor (CGR-1), and latent transforming growth factor-ß complex protein 1 (LTCP-1), is a multifunctional protein with widespread tissue distribution. To determine the functional consequences of ESL-1 deficiency, mice were generated carrying an ESL-1 gene trap. After backcrossing to C57BL6/J for 6 generations, mice heterozygous for the gene trap (ESL-1(+/-)) were intercrossed to produce ESL-1(-/-) mice, however ESL-1(-/-) mice were not viable, even at embryonic day E10.5. To determine the effect of heterozygous ESL-1 deficiency on atherosclerosis, apolipoprotein E deficient (ApoE(-/-)), ESL-1(+/-) mice were generated and fed western diet. Compared to ApoE(-/-), ESL-1(+)(/)(+) mice, atherosclerotic lesions from ApoE(-/-), ESL-1(+/-) contained more collagen and fewer macrophages, suggesting increased plaque stability. In conclusion, heterozygous deficiency of ESL-1 is associated with features of increased atherosclerotic plaque stability while complete deficiency of ESL-1 leads to embryonic lethality.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Embryo Loss/metabolism , Haploinsufficiency , Macrophages/metabolism , Plaque, Atherosclerotic , Receptors, Fibroblast Growth Factor/deficiency , Sialoglycoproteins/deficiency , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Collagen/metabolism , Disease Models, Animal , Embryo Loss/genetics , Genotype , Gestational Age , Heterozygote , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Fibroblast Growth Factor/genetics , Sialoglycoproteins/genetics
9.
J Investig Dermatol Symp Proc ; 15(1): 48-52, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22076327

ABSTRACT

Fibrosis is associated with a variety of skin diseases and causes severe aesthetic and functional impairments. Functional studies in rodents, together with clinical observations, strongly suggest a crucial role of chronic injury and inflammation in the pathogenesis of fibrotic diseases. The phenotype of mice lacking fibroblast growth factor (FGF) receptors 1 and 2 in keratinocytes supports this concept. In these mice, a defect in keratinocytes alone initiated an inflammatory response, which in turn caused keratinocyte hyperproliferation and dermal fibrosis. As the mechanism underlying this phenotype, we identified a loss of FGF-induced expression of claudins and occludin, which caused abnormalities in tight junctions with concomitant deficits in epidermal barrier function. This resulted in severe transepidermal water loss and skin dryness. In turn, activation of keratinocytes and epidermal γδ T cells occurred, which produced IL-1 family member 8 and S100A8 and S100A9. These cytokines attracted immune cells and activated fibroblasts, resulting in a double paracrine loop through production of keratinocyte mitogens by dermal cells. In addition, a profibrotic response was induced in fibroblasts. Our results highlight the importance of an intact epidermal barrier for the prevention of inflammation and fibrosis and the role of chronic inflammation in the pathogenesis of fibrotic diseases.


Subject(s)
Dermatitis/metabolism , Dermatitis/pathology , Keratinocytes/metabolism , Receptors, Fibroblast Growth Factor/deficiency , Skin/pathology , Animals , Cell Proliferation , Chronic Disease , Claudins/biosynthesis , Cytokines/immunology , Dermatitis/immunology , Fibrosis , Keratinocytes/immunology , Membrane Proteins/biosynthesis , Mice , Occludin , Receptors, Fibroblast Growth Factor/immunology , Skin/immunology , Skin/metabolism , T-Lymphocytes/immunology , Tight Junctions/metabolism , Water Loss, Insensible/physiology
10.
J Clin Invest ; 121(6): 2210-20, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21537081

ABSTRACT

Patients with atopic dermatitis (AD) often suffer from food allergy and develop flares upon skin contact with food allergens. However, it is unclear whether T cells sensitized to allergens in the gut promote this skin inflammation. To address this question, we orally immunized WT mice and mice lacking the skin-homing chemokine receptor Ccr4 (Ccr4-/- mice) with OVA and then challenged them epicutaneously with antigen. Allergic skin inflammation developed in the WT mice but not in the mutants and was characterized by epidermal thickening, dermal infiltration by eosinophils and CD4+ T cells, and upregulation of Th2 cytokines. T cells purified from mesenteric lymph nodes (MLNs) of orally immunized WT mice transferred allergic skin inflammation to naive recipients cutaneously challenged with antigen, but this effect was lost in T cells purified from Ccr4-/- mice. In addition, the ability of adoptively transferred OVA-activated T cells to home to the skin following cutaneous OVA challenge was ablated in mice that lacked lymph nodes. These results indicate that cutaneous exposure to food antigens can reprogram gut-homing effector T cells in LNs to express skin-homing receptors, eliciting skin lesions upon food allergen contact in orally sensitized AD patients.


Subject(s)
Allergens/administration & dosage , Chemotaxis, Leukocyte , Dermatitis, Allergic Contact/immunology , Immunization , Receptors, CCR4/physiology , Skin/immunology , T-Lymphocyte Subsets/immunology , Administration, Cutaneous , Administration, Oral , Adoptive Transfer , Allergens/toxicity , Animals , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Cholera Toxin/toxicity , Dermatitis, Allergic Contact/pathology , Food Hypersensitivity/complications , Food Hypersensitivity/immunology , Integrins/deficiency , Integrins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/administration & dosage , Ovalbumin/immunology , Ovalbumin/toxicity , Receptors, CCR4/deficiency , Receptors, CCR4/genetics , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/physiology , Receptors, Lymphocyte Homing/immunology , Sialoglycoproteins/deficiency , Sialoglycoproteins/physiology , Skin/pathology , Specific Pathogen-Free Organisms , T-Lymphocyte Subsets/transplantation
11.
Crit Rev Eukaryot Gene Expr ; 20(4): 295-311, 2010.
Article in English | MEDLINE | ID: mdl-21395503

ABSTRACT

Fibroblast growth factor receptors comprise a family of four evolutionarily conserved transmembrane proteins (FGFR1, FGFR2, FGFR3 and FGFR4) known to be critical for the normal development of multiple organ systems. In this review we will primarily focus upon the role of FGF/FGFR signaling as it influences the development of the craniofacial skeleton. Signaling by FGF receptors is regulated by the tissue-specific expression of FGFR isoforms, receptor subtype specific fibroblast growth factors and heparin sulfate proteoglycans. Signaling can also be limited by the expression of endogenous inhibitors. Gain-of-function mutations in FGFRs are associated with a series of congenital abnormality syndromes referred to as the craniosynostosis syndromes. Craniosynostosis is the clinical condition of premature cranial bone fusion and patients who carry craniosynostosis syndrome-associated mutations in FGFRs commonly have abnormalities of the skull vault in the form of craniosynostosis. Patients may also have abnormalities in the facial skeleton, vertebrae and digits. In this review we will discuss recent in vitro and in vivo studies investigating biologic mechanisms by which signaling through FGFRs influences skeletal development and can lead to craniosynostosis.


Subject(s)
Craniosynostoses/etiology , Facial Bones/growth & development , Fibroblast Growth Factors/physiology , Skull/growth & development , Animals , Craniosynostoses/genetics , Fibroblast Growth Factors/deficiency , Fibroblast Growth Factors/genetics , Humans , Mice , Mice, Knockout , Models, Biological , Mutation , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/physiology , Signal Transduction
12.
Development ; 136(14): 2457-65, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19542358

ABSTRACT

The FGF family of extracellular signaling factors has been proposed to play multiple roles in patterning the telencephalon, the precursor to the cerebrum. In this study, unlike previous ones, we effectively abolish FGF signaling in the anterior neural plate via deletion of three FGF receptor (FGFR) genes. Triple FGFR mutant mice exhibit a complete loss of the telencephalon, except the dorsal midline. Disruption of FGF signaling prior to and coincident with telencephalic induction reveals that FGFs promote telencephalic character and are strictly required to keep telencephalic cells alive. Moreover, progressively more severe truncations of the telencephalon are observed in FGFR single, double and triple mutants. Together with previous gain-of-function studies showing induction of Foxg1 expression and mirror-image duplications of the cortex by exogenous FGF8, our loss-of-function results suggest that, rather than independently patterning different areas, FGF ligands and receptors act in concert to mediate organizer activity for the whole telencephalon.


Subject(s)
Fibroblast Growth Factors/metabolism , Telencephalon/embryology , Telencephalon/metabolism , Animals , Body Patterning , Cell Survival/genetics , Cell Survival/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization , Mice , Mice, Knockout , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Plate/cytology , Neural Plate/embryology , Neural Plate/metabolism , Neurogenesis/genetics , Neurogenesis/physiology , Pregnancy , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Telencephalon/cytology
13.
J Neurosci ; 29(6): 1608-14, 2009 Feb 11.
Article in English | MEDLINE | ID: mdl-19211868

ABSTRACT

Axon-glial interactions are critical for normal functioning of peripheral nerves, and their disruption leads to peripheral neuropathies. Fibroblast growth factors (FGFs) are key players in peripheral nerve regeneration after injury. We investigated the role of FGF receptor (Fgfr) signaling in Schwann cells and the consequent regulation of normal Schwann cell-axon interactions. Fgfr1 and Fgfr2 were conditionally inactivated, either singly or in combination, in myelinating and nonmyelinating Schwann cells (NMSCs) of transgenic mice. The double mutant mice displayed significant loss of thermal sensitivity accompanied by marked neuropathy of unmyelinated nociceptive sensory axons terminating in the dorsal horn of spinal cords, the primary site for integrating pain and temperature inputs. Neuropathy, although to a lesser extent, was also observed in the nociceptive C-fibers in the Remak bundles of sciatic nerves; however, there was no loss of NMSCs that ensheathe these axons. Furthermore, axons wrapped by myelinating Schwann cells and associated myelin sheaths appeared to be unaffected. Relative to the double mutants, axonal neuropathy developed much later in the Fgfr1 but not Fgfr2 single mutant, indicating a difference in signaling potential of the two receptors, with Fgfr1 being more robust than Fgfr2. These findings emphasize the importance of Fgfr1 and Fgfr2 signaling as potential mediators of axon-glial interaction in the peripheral sensory pain pathway primarily via influencing NMSC function, which in turn modulates the structure and function of unmyelinated sensory axons. This study provides a novel molecular mechanism for nociception with possible implications for pain sensitivity in peripheral sensory neuropathies.


Subject(s)
Axons/physiology , Hot Temperature , Neuralgia/metabolism , Pain/metabolism , Receptors, Fibroblast Growth Factor/deficiency , Schwann Cells/physiology , Sensory Receptor Cells/pathology , Signal Transduction , Animals , Axons/pathology , Hot Temperature/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelin Sheath/genetics , Myelin Sheath/metabolism , Myelin Sheath/pathology , Neuralgia/genetics , Neuralgia/pathology , Pain/genetics , Pain/pathology , Pain Measurement/methods , Receptors, Fibroblast Growth Factor/genetics , Schwann Cells/pathology , Sensory Receptor Cells/physiology , Signal Transduction/genetics
14.
Mech Dev ; 126(1-2): 1-17, 2009.
Article in English | MEDLINE | ID: mdl-19015027

ABSTRACT

The roles of the FGF family growth factors and their receptors (FGFRs) in zebrafish embryos were examined using variously modified versions of the four FGFR genes (fgfr1-4). Constitutively active forms of all of the examined FGFRs (ca-FGFRs) caused dorsalization, brain caudalization, and secondary axis formation, indicating that the main FGF signal transduction downstream of the receptor is highly similar among FGFRs. All of the membrane-bound type of dominant-negative FGFRs (mdn-FGFRs) derived from the four fgfr genes, which interfere with endogenous FGFRs, produced posterior truncation, as previously reported in both Xenopus and zebrafish. mdn-FGFR3c had the strongest effects on embryos, progressively disrupting the posterior structure as the dose increased. At the highest dose, only the forebrain was formed. At lower doses, mdn-FGFR3c mainly suppressed the paraxial mesoderm. The co-injection of mRNA for different mdn-FGFRs and FGFs resulted in diverse suppression spectra of the respective FGFRs against FGFs. Only mdn-FGFR3c severely suppressed all of the FGFs examined. We also examined the effects of the secretory type of dominant-negative FGFRs (sdn-FGFRs), which are released from cells and trap FGF ligands. Only sdn-FGFR3c resulted in the characteristic effect of selectively disrupting the isthmic development, as well as the tailbud. The co-injection of the mRNA for sdn-FGFRs and FGFs suggested that sdn-FGFR3c inhibits FGFs of the FGF8 subfamily, which is consistent with its specific effects on development. We discuss the implications of our findings obtained in the present study.


Subject(s)
Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Fibroblast Growth Factors/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Zebrafish/embryology , Zebrafish/metabolism , Animals , Body Patterning , Gene Expression Regulation, Developmental , Ligands , RNA, Messenger/genetics , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics , Solubility , Zebrafish/genetics
15.
Development ; 133(15): 2937-46, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16818446

ABSTRACT

Sonic hedgehog (SHH) is required to generate ventral cell types throughout the central nervous system. Its role in directly specifying ventral cells, however, has recently been questioned because loss of the Shh gene has little effect on ventral development if the Gli3 gene is also mutant. Consequently, another ventral determinant must exist. Here, genetic evidence establishes that FGFs are required for ventral telencephalon development. First, simultaneous deletion of Fgfr1 and Fgfr3 specifically in the telencephalon results in the loss of differentiated ventromedial cells; and second, in the Fgfr1;Fgfr2 double mutant, ventral precursor cells are lost, mimicking the phenotype obtained previously with a loss of SHH signalling. Yet, in the Fgfr1;Fgfr2 mutant, Shh remains expressed, as does Gli1, the transcription of which depends on SHH activity, suggesting that FGF signalling acts independently of SHH to generate ventral precursors. Moreover, the Fgfr1;Fgfr2 phenotype, unlike the Shh phenotype, is not rescued by loss of Gli3, further indicating that FGFs act downstream of Shh and Gli3 to generate ventral telencephalic cell types.


Subject(s)
Fibroblast Growth Factors/physiology , Telencephalon/embryology , Trans-Activators/physiology , Animals , Body Patterning , Female , Hedgehog Proteins , Mice , Mice, Knockout , Pregnancy , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics , Signal Transduction
16.
J Biol Chem ; 280(18): 17707-14, 2005 May 06.
Article in English | MEDLINE | ID: mdl-15750181

ABSTRACT

The fibroblast growth factor (FGF) receptor complex is a regulator of adult organ homeostasis in addition to its central role in embryonic development and wound healing. FGF receptor 4 (FGFR4) is the sole FGFR receptor kinase that is significantly expressed in mature hepatocytes. Previously, we showed that mice lacking mouse FGFR4 (mR4(-/-)) exhibited elevated fecal bile acids, bile acid pool size, and expression of liver cholesterol 7alpha-hydroxylase (CYP7A1), the rate-limiting enzyme for canonical neutral bile acid synthesis. To prove that hepatocyte FGFR4 was a negative regulator of cholesterol metabolism and bile acid synthesis independent of background, we generated transgenic mice overexpressing a constitutively active human FGFR4 (CahR4) in hepatocytes and crossed them with the FGFR4-deficient mice to generate CahR4/mR4(-/-) mice. In mice expressing active FGFR4 in liver, fecal bile acid excretion was 64%, bile acid pool size was 47%, and Cyp7a1 expression was 10-30% of wild-type mice. The repressed level of Cyp7a1 expression was resistant to induction by a high cholesterol diet relative to wild-type mice. Expression of CahR4 in mR4(-/-) mouse livers depressed bile acid synthesis below wild-type levels from the elevated levels observed in mR4(-/-). Levels of phosphorylated c-Jun N-terminal kinase (JNK), which is part of a pathway implicated in bile acid-mediated repression of synthesis, was 30% of wild-type levels in mR4(-/-) livers, whereas CahR4 livers exhibited an average 2-fold increase. However, cholate still strongly induced phospho-JNK in mR4(-/-) livers. These results confirm that hepatocyte FGFR4 regulates bile acid synthesis by repression of Cyp7a1 expression. Hepatocyte FGFR4 may contribute to the repression of bile acid synthesis through JNK signaling but is not required for activation of JNK signaling by bile acids.


Subject(s)
Bile Acids and Salts/antagonists & inhibitors , Bile Acids and Salts/biosynthesis , Hepatocytes/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Receptors, Fibroblast Growth Factor/physiology , Animals , Down-Regulation/genetics , Down-Regulation/physiology , Enzyme Activation/genetics , Feedback, Physiological/genetics , Humans , Mice , Mice, Knockout , Mice, Transgenic , Receptor, Fibroblast Growth Factor, Type 4 , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics
17.
Dev Dyn ; 233(2): 516-27, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15778993

ABSTRACT

Fibroblast growth factors (FGFs) play important roles in many aspects of development, including lens development. The lens is derived from the surface ectoderm and consists of an anterior layer of epithelial cells and elongated, terminally differentiated fiber cells that form the bulk of the tissue. FGF signaling has been implicated in lens induction, proliferation, and differentiation. To address the role of FGFs in lens development, we inactivated FGF receptor-2 (Fgfr2) using a Cre transgene that is expressed in all prospective lens cells from embryonic day 9.0. Inactivation of Fgfr2 shows that signaling through this receptor is not required for lens induction or for the proliferation of lens epithelial cells. However, Fgfr2 signaling is needed to drive lens fiber cells out of the cell cycle during their terminal differentiation. It also contributes to the normal elongation of primary lens fiber cells and to the survival of lens epithelial cells.


Subject(s)
Cell Cycle , Cell Differentiation , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Animals , Cell Survival , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Lens, Crystalline/embryology , Mice , Mice, Knockout , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics
18.
J Biol Chem ; 280(21): 20509-15, 2005 May 27.
Article in English | MEDLINE | ID: mdl-15781473

ABSTRACT

Signaling by fibroblast growth factor (FGF) 18 and FGF receptor 3 (FGFR3) have been shown to regulate proliferation, differentiation, and matrix production of articular and growth plate chondrocytes in vivo and in vitro. Notably, the congenital absence of either FGF18 or FGFR3 resulted in similar expansion of the growth plates of fetal mice and the addition of FGF18 to human articular chondrocytes in culture enhanced proliferation and matrix production. Based on these and other experiments it has been proposed that FGF18 signals through FGFR3 to promote cartilage production by chondrocytes. Its role in chondrogenesis remains to be defined. In the current work we used the limb buds of FGFR3(+/+) and FGFR3(-/-) embryonic mice as a source of mesenchymal cells to determine how FGF18 signaling affects chondrogenesis. Confocal laser-scanning microscopy demonstrated impaired cartilage nodule formation in the FGFR3(-/-) cultures. Potential contributing factors to the phenotype were identified as impaired mitogenic response to FGF18, decreased production of type II collagen and proteoglycan in response to FGF18 stimulation, impaired interactions with the extracellular matrix resulting from altered integrin receptor expression, and altered expression of FGFR1 and FGFR2. The data identified FGF18 as a selective ligand for FGFR3 in limb bud mesenchymal cells, which suppressed proliferation and promoted their differentiation and production of cartilage matrix. This work, thus, identifies FGF18 and FGFR3 as potential molecular targets for intervention in tissue engineering aimed at cartilage repair and regeneration of damaged cartilage.


Subject(s)
Chondrogenesis/physiology , Fibroblast Growth Factors/physiology , Protein-Tyrosine Kinases/physiology , Receptors, Fibroblast Growth Factor/physiology , Signal Transduction , Animals , Cartilage/cytology , Cartilage/embryology , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Chondrocytes/cytology , Collagen Type II/analysis , Collagen Type X/genetics , Extremities/embryology , Female , Fibroblast Growth Factors/pharmacology , Fluorescent Antibody Technique , Gene Expression/drug effects , Ligands , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Knockout , Microscopy, Confocal , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , RNA, Messenger/analysis , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
19.
J Surg Res ; 120(2): 201-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15234214

ABSTRACT

BACKGROUND: Intestinal atresia represents a significant surgically correctable cause of intestinal obstruction in neonates. Intestinal development proceeds as a tube-like structure with differentiation along its axis. As the intestine differentiates, the cecum develops at the transition from small to large intestine. Fgf10 is known to serve a key role in budding morphogenesis; however, little is known about its role in the development of this transitional structure. Here we evaluate the effect of Fgf10/Fgfr2b invalidation on the developing cecum. MATERIALS AND METHODS: Wild-type C57Bl/6, Fgf10(-/-), and Fgfr2b(-/-) embryos harvested from timed pregnant mothers were analyzed for cecal phenotype, Fgf10 expression, and differentiation of smooth muscle actin. RESULTS: Wt cecal development is first evident at E11.5. FGF10 is discreetly expressed in the area of the developing cecum at early stages of development. One hundred percent of Fgf10(-/-) and Fgfr2b(-/-) mutant embryos demonstrate cecal atresia with absence of epithelial and muscular layers. The development of neighboring anatomical structures such as the ileocecal valve is not affected by Fgf10/Fgfr2b invalidation. CONCLUSIONS: FGF10 expression is localized to the cecum early in the normal development of the cecum. Fgf10(-/-) and Fgfr2b(-/-) mutant embryos demonstrate cecal atresia with complete penetrance. Epithelial and muscular layers of the cecum are not present in the atretic cecum. The Fgf10(-/-) and Fgfr2b(-/-) mutants represent a genetically reproducible animal model of autosomal recessive intestinal atresia.


Subject(s)
Cecum , Fibroblast Growth Factors/metabolism , Intestinal Atresia/physiopathology , Signal Transduction , Animals , Cecum/metabolism , Embryonic and Fetal Development , Fibroblast Growth Factor 10 , Fibroblast Growth Factors/deficiency , Fibroblast Growth Factors/genetics , Intestinal Atresia/etiology , Intestinal Atresia/metabolism , Intestinal Atresia/pathology , Intestinal Mucosa , Mice , Mice, Knockout , Muscle, Smooth/embryology , Muscle, Smooth/pathology , Mutation , Penetrance , Peristalsis , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism
20.
J Pediatr Surg ; 39(6): 872-4, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15185216

ABSTRACT

BACKGROUND/PURPOSE: Duodenal atresia (DA) occurs in 1 in every 6,000 live births and represents a significant surgically correctable cause of intestinal obstruction in the neonate. Familial or congenital DA has been reported, implying that at least some cases of DA are the result of genetic, heritable abnormalities. The genes controlling duodenal development are incompletely understood. Fibroblast growth factor receptor 2IIIb (Fgfr2b) is known to play a critical role in the development of multiple organ systems including other gastrointestinal tract (GIT) structures. This study shows the key role of Fgfr2b in normal duodenal development and the pathogenesis of DA. METHODS: Wild type (Wt) and Fgfr2b-/- embryos were harvested from timed pregnant mothers at stage E18.5 and were analyzed for duodenal phenotype. RESULTS: Inactivation of Fgfr2b results in DA. DA is present in the Fgf2b-/- mutants with a 35% penetrance. The duodenal phenotype of the Fgf2b-/- mutants ranges from normal to a mucosal web, type I, and type III atresia. CONCLUSIONS: Fgfr2b is a critical regulatory gene in the development of the duodenum. Fgfr2b invalidation (Fgfr2b-/- mutant) results in a reproducible, autosomal recessive duodenal atresia phenotype with incomplete penetrance and a variable phenotype.


Subject(s)
Duodenal Obstruction/congenital , Intestinal Atresia/genetics , Receptors, Fibroblast Growth Factor/physiology , Animals , Disease Models, Animal , Duodenal Obstruction/embryology , Duodenal Obstruction/genetics , Duodenum/embryology , Gestational Age , Intestinal Atresia/embryology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...