Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Int J Mol Sci ; 20(23)2019 Nov 30.
Article in English | MEDLINE | ID: mdl-31801200

ABSTRACT

Paracrine factors can induce cardiac regeneration and repair post myocardial infarction by stimulating proliferation of cardiac cells and inducing the anti-fibrotic, antiapoptotic, and immunomodulatory effects of angiogenesis. Here, we screened a human secretome library, consisting of 923 growth factors, cytokines, and proteins with unknown function, in a phenotypic screen with human cardiac progenitor cells. The primary readout in the screen was proliferation measured by nuclear count. From this screen, we identified FGF1, FGF4, FGF9, FGF16, FGF18, and seven additional proteins that induce proliferation of cardiac progenitor cells. FGF9 and FGF16 belong to the same FGF subfamily, share high sequence identity, and are described to have similar receptor preferences. Interestingly, FGF16 was shown to be specific for proliferation of cardiac progenitor cells, whereas FGF9 also proliferated human cardiac fibroblasts. Biosensor analysis of receptor preferences and quantification of receptor abundances suggested that FGF16 and FGF9 bind to different FGF receptors on the cardiac progenitor cells and cardiac fibroblasts. FGF16 also proliferated naïve cardiac progenitor cells isolated from mouse heart and human cardiomyocytes derived from induced pluripotent cells. Taken together, the data suggest that FGF16 could be a suitable paracrine factor to induce cardiac regeneration and repair.


Subject(s)
Cell Proliferation/drug effects , Fibroblast Growth Factors/genetics , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Animals , CHO Cells , Cell Differentiation/drug effects , Cricetulus , Female , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , Gene Library , High-Throughput Screening Assays , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Primary Cell Culture
2.
Front Med ; 13(5): 511-530, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31495905

ABSTRACT

Members of the fibroblast growth factor (FGF) family play pleiotropic roles in cellular and metabolic homeostasis. During evolution, the ancestor FGF expands into multiple members by acquiring divergent structural elements that enable functional divergence and specification. Heparan sulfate-binding FGFs, which play critical roles in embryonic development and adult tissue remodeling homeostasis, adapt to an autocrine/paracrine mode of action to promote cell proliferation and population growth. By contrast, FGF19, 21, and 23 coevolve through losing binding affinity for extracellular matrix heparan sulfate while acquiring affinity for transmembrane α-Klotho (KL) or ß-KL as a coreceptor, thereby adapting to an endocrine mode of action to drive interorgan crosstalk that regulates a broad spectrum of metabolic homeostasis. FGF19 metabolic axis from the ileum to liver negatively controls diurnal bile acid biosynthesis. FGF21 metabolic axes play multifaceted roles in controlling the homeostasis of lipid, glucose, and energy metabolism. FGF23 axes from the bone to kidney and parathyroid regulate metabolic homeostasis of phosphate, calcium, vitamin D, and parathyroid hormone that are important for bone health and systemic mineral balance. The significant divergence in structural elements and multiple functional specifications of FGF19, 21, and 23 in cellular and organismal metabolism instead of cell proliferation and growth sufficiently necessitate a new unified and specific term for these three endocrine FGFs. Thus, the term "FGF Metabolic Axis," which distinguishes the unique pathways and functions of endocrine FGFs from other autocrine/paracrine mitogenic FGFs, is coined.


Subject(s)
Endocrine System/metabolism , Fibroblast Growth Factors/metabolism , Metabolic Diseases/metabolism , Neoplasms/metabolism , Animals , Calcium/metabolism , Cell Proliferation , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/classification , Homeostasis , Humans , Liver/metabolism , Phosphates/metabolism , Signal Transduction , Vitamin D/metabolism
3.
Int J Dev Biol ; 63(11-12): 631-639, 2019.
Article in English | MEDLINE | ID: mdl-32149373

ABSTRACT

Fibroblast growth factors (FGFs) comprise a family of signalling molecules with essential roles in early embryonic development across animal species. The role of FGFs in mesoderm formation and patterning in Xenopus has been particularly well studied. However, little is known about FGF16 in Xenopus. Using in situ hybridisation, we uncover the expression pattern of FGF16 during early Xenopus laevis development, which has not been previously described. We show that the zygotic expression of FGF16 is activated in the mesoderm of the early gastrula as a ring around the blastopore, with its first accumulation at the dorsal side of the embryo. Later, FGF16 expression is found in the otic vesicle, the branchial arches and the anterior pituitary, as well as in the chordal neural hinge region of the tailbud. In addition, we show that FGF16 can activate the MAPK pathway and expression of sp5 and sp5l. Like FGF16, sp5 is expressed in the otic vesicle and the branchial arches, with all three of these genes being expressed in the tailbud. These data provide evidence that FGF16 is present in the early mesoderm and can activate the expression of developmentally important transcription factors.


Subject(s)
Fibroblast Growth Factors/genetics , Transcription Factors/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics , Animals , Branchial Region/embryology , Branchial Region/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Fibroblast Growth Factors/classification , Gastrula/embryology , Gastrula/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization , MAP Kinase Signaling System/genetics , Mesoderm/embryology , Mesoderm/metabolism , Phylogeny , Xenopus laevis/embryology
4.
Biomed Khim ; 63(3): 211-218, 2017 May.
Article in Russian | MEDLINE | ID: mdl-28781254

ABSTRACT

Fibroblast growth factors (FGF) - growth factors that regulate many important biological processes, including proliferation and differentiation of embryonic cells during organogenesis. In this review, we will summarize current information about the involvement of FGFs in the pancreas organogenesis. Pancreas organogenesis is a complex process, which involves constant signaling from mesenchymal tissue. This orchestrates the activation of various regulator genes at specific stages, determining the specification of progenitor cells. Alterations in FGF/FGFR signaling pathway during this process lead to incorrect activation of the master genes, which leads to different pathologies during pancreas development. Understanding the full picture about role of FGF factors in pancreas development will make it possible to more accurately understand their role in other pathologies of this organ, including carcinogenesis.


Subject(s)
Enteroendocrine Cells/metabolism , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Organogenesis/genetics , Pancreas/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Animals , Cell Differentiation , Cell Lineage/genetics , Enteroendocrine Cells/cytology , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Pancreas/cytology , Pancreas/growth & development , Receptor, Fibroblast Growth Factor, Type 1/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Proc Natl Acad Sci U S A ; 112(52): 15940-5, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26644578

ABSTRACT

Extreme novelties in the shape and size of paired fins are exemplified by extinct and extant cartilaginous and bony fishes. Pectoral fins of skates and rays, such as the little skate (Batoid, Leucoraja erinacea), show a strikingly unique morphology where the pectoral fin extends anteriorly to ultimately fuse with the head. This results in a morphology that essentially surrounds the body and is associated with the evolution of novel swimming mechanisms in the group. In an approach that extends from RNA sequencing to in situ hybridization to functional assays, we show that anterior and posterior portions of the pectoral fin have different genetic underpinnings: canonical genes of appendage development control posterior fin development via an apical ectodermal ridge (AER), whereas an alternative Homeobox (Hox)-Fibroblast growth factor (Fgf)-Wingless type MMTV integration site family (Wnt) genetic module in the anterior region creates an AER-like structure that drives anterior fin expansion. Finally, we show that GLI family zinc finger 3 (Gli3), which is an anterior repressor of tetrapod digits, is expressed in the posterior half of the pectoral fin of skate, shark, and zebrafish but in the anterior side of the pelvic fin. Taken together, these data point to both highly derived and deeply ancestral patterns of gene expression in skate pectoral fins, shedding light on the molecular mechanisms behind the evolution of novel fin morphologies.


Subject(s)
Adaptation, Physiological/genetics , Animal Fins/metabolism , Fish Proteins/genetics , Skates, Fish/genetics , Animal Fins/anatomy & histology , Animal Fins/embryology , Animals , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/genetics , Fish Proteins/classification , Gene Expression Regulation, Developmental , Homeodomain Proteins/classification , Homeodomain Proteins/genetics , In Situ Hybridization , Phylogeny , Skates, Fish/embryology
6.
Cell Mol Life Sci ; 72(12): 2445-59, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25854632

ABSTRACT

Fibroblast growth factors (FGFs) deliver extracellular signals that govern many developmental and regenerative processes, but the mechanisms regulating FGF signaling remain incompletely understood. Here, we explored the relationship between intrinsic stability of FGF proteins and their biological activity for all 18 members of the FGF family. We report that FGF1, FGF3, FGF4, FGF6, FGF8, FGF9, FGF10, FGF16, FGF17, FGF18, FGF20, and FGF22 exist as unstable proteins, which are rapidly degraded in cell cultivation media. Biological activity of FGF1, FGF3, FGF4, FGF6, FGF8, FGF10, FGF16, FGF17, and FGF20 is limited by their instability, manifesting as failure to activate FGF receptor signal transduction over long periods of time, and influence specific cell behavior in vitro and in vivo. Stabilization via exogenous heparin binding, introduction of stabilizing mutations or lowering the cell cultivation temperature rescues signaling of unstable FGFs. Thus, the intrinsic ligand instability is an important elementary level of regulation in the FGF signaling system.


Subject(s)
Bone Neoplasms/metabolism , Breast Neoplasms/metabolism , Cell Proliferation , Chondrosarcoma/metabolism , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/metabolism , Signal Transduction , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Chondrosarcoma/genetics , Chondrosarcoma/pathology , Circular Dichroism , Female , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/genetics , Humans , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation/genetics , Protein Stability , Rats , Temperature , Tumor Cells, Cultured
7.
Adv Exp Med Biol ; 728: 1-24, 2012.
Article in English | MEDLINE | ID: mdl-22396159

ABSTRACT

The ability of the Fibroblast Growth Factor (FGF) 19 subfamily to signal in an endocrine fashion sets this subfamily apart from the remaining five FGF subfamilies known for their paracrine functions during embryonic development. Compared to the members of paracrine FGF subfamiles, the three members of the FGF19 subfamily, namely FGF19, FGF21 and FGF23, have poor affinity for heparan sulfate (HS) and therefore can diffuse freely in the HS-rich extracellular matrix to enter into the bloodstream. In further contrast to paracrine FGFs, FGF19 subfamily members have unusually poor affinity for their cognate FGF receptors (FGFRs) and therefore cannot bind and activate them in a solely HS-dependent fashion. As a result, the FGF19 subfamily requires α/ßklotho coreceptor proteins in order to bind, dimerize and activate their cognate FGFRs. This klotho-dependency also determines the tissue specificity of endocrine FGFs. Recent structural and biochemical studies have begun to shed light onto the molecular basis for the klotho-dependent endocrine mode of action of the FGF19 subfamily. Crystal structures of FGF19 and FGF23 show that the topology of the HS binding site (HBS) of FGF19 subfamily members deviates drastically from the common topology adopted by the paracrine FGFs. The distinct topologies of the HBS of FGF19 and FGF23 prevent HS from direct hydrogen bonding with the backbone atoms of the HBS of these ligands and accordingly decrease the HS binding affinity of this subfamily. Recent biochemical data reveal that the ?klotho ectodomain binds avidly to the ectodomain of FGFR1c, the main cognate FGFR of FGF23, creating a de novo high affinity binding site for the C-terminal tail of FGF23. The isolated FGF23 C-terminus can be used to effectively inhibit the formation of the FGF23-FGFR1c-αklotho complex and alleviate hypophosphatemia in renal phosphate disorders due to elevated levels of FGF23.


Subject(s)
Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/metabolism , Amino Acid Sequence , Animals , Endocrine System/cytology , Endocrine System/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/classification , Glucuronidase/metabolism , Humans , Klotho Proteins , Molecular Sequence Data , Signal Transduction , Structure-Activity Relationship
8.
Neuron ; 71(4): 574-88, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21867876

ABSTRACT

The generation of a functional nervous system involves a multitude of steps that are controlled by just a few families of extracellular signaling molecules. Among these, the fibroblast growth factor (FGF) family is particularly prominent for the remarkable diversity of its functions. FGFs are best known for their roles in the early steps of patterning of the neural primordium and proliferation of neural progenitors. However, other equally important functions have emerged more recently, including in the later steps of neuronal migration, axon navigation, and synaptogenesis. We review here these diverse functions and discuss the mechanisms that account for this unusual range of activities. FGFs are essential components of most protocols devised to generate therapeutically important neuronal populations in vitro or to stimulate neuronal repair in vivo. How FGFs promote the development of the nervous system and maintain its integrity will thus remain an important focus of research in the future.


Subject(s)
Fibroblast Growth Factors/metabolism , Nervous System/embryology , Nervous System/growth & development , Neurogenesis/physiology , Neurons/physiology , Animals , Cell Movement , Cell Proliferation , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/genetics , Humans , Nervous System/cytology , Nervous System/metabolism , Neural Stem Cells/physiology , Protein Isoforms/classification , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Fibroblast Growth Factor/classification , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/physiology
9.
Dev Dyn ; 238(8): 2058-72, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19544582

ABSTRACT

The cerebellum is derived from the anterior-most segment of the embryonic hindbrain, rhombomere 1 (r1). Previous studies have shown that the early development and patterning of r1 requires fibroblast growth factor (FGF) signaling. However, many of the developmental processes that shape cerebellar morphogenesis take place later in embryonic development and during the first 2 weeks of postnatal life in the mouse. Here, we present a more comprehensive analysis of the expression patterns of genes encoding FGF receptors and secreted FGF ligands during these later stages of cerebellar development. We show that these genes are expressed in multiple cell types in the developing cerebellum, in an astonishing array of distinct patterns. These data suggest that FGF signaling functions throughout cerebellar development to regulate many processes that shape the formation of a functional cerebellum.


Subject(s)
Cerebellum/embryology , Cerebellum/metabolism , Fibroblast Growth Factors/genetics , Animals , Cell Differentiation , Cell Proliferation , Cerebellum/cytology , Cerebellum/growth & development , Female , Fibroblast Growth Factors/classification , Gene Expression Regulation, Developmental , In Situ Hybridization , Ligands , Mice , Mice, Transgenic , Pregnancy , Receptors, Fibroblast Growth Factor/genetics , Signal Transduction
10.
Dev Dyn ; 237(9): 2594-603, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18729221

ABSTRACT

Signaling pathways mediated by receptor tyrosine kinases (RTKs) are under positive and negative regulation, and misregulation of RTK signaling results in developmental defects and malignancy. A major class of antagonists of Fgf and Egf signaling are the Sprouty proteins. Through an enhancer detection approach, we isolated the sprouty1 (spry1) gene, expressed in multiple developing organs during embryogenesis. We analyzed expression of spry1 between tail bud stage and 10 days postfertilization. From the tail bud stage on, transcript and reporter are detected in the craniofacial region and in the mid-hindbrain boundary, where expression persists until adulthood. Further expression domains are the telencephalon, hindbrain, dorsal diencephalon and epiphysis, branchial arches, pituitary, and the tubular gill epithelium. In the trunk spry1 is also prominently expressed in pronephros, the lateral line and tail fin. Sprouty1 acts in Fgf signaling downstream of Fgfr1, as its expression is abrogated through the small molecule inhibitor of this receptor, SU5402.


Subject(s)
Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Zebrafish Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/classification , In Situ Hybridization , Molecular Sequence Data , Nervous System/drug effects , Nervous System/embryology , Nervous System/metabolism , Phylogeny , Pyrroles/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Zebrafish/embryology , Zebrafish Proteins/chemistry , Zebrafish Proteins/classification
11.
Dev Dyn ; 237(1): 18-27, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18058912

ABSTRACT

Fibroblast Growth Factors (FGFs) are polypeptides with diverse activities in development and physiology. The mammalian Fgf family can be divided into the intracellular Fgf11/12/13/14 subfamily (iFGFs), the hormone-like Fgf15/21/23 subfamily (hFGFs), and the canonical Fgf subfamilies, including Fgf1/2/5, Fgf3/4/6, Fgf7/10/22, Fgf8/17/18, and Fgf9/16/20. However, all Fgfs are evolutionarily related. We propose that an Fgf13-like gene is the ancestor of the iFgf subfamily and the most likely evolutionary ancestor of the entire Fgf family. Potential ancestors of the canonical and hFgf subfamilies, Fgf4-, Fgf5-, Fgf8-, Fgf9-, Fgf10-, and Fgf15-like, appear to have derived from an Fgf13-like ancestral gene. Canonical FGFs function in a paracrine manner, while hFGFs function in an endocrine manner. We conclude that the ancestral Fgfs for these subfamilies acquired this functional diversity before the evolution of vertebrates. During the evolution of early vertebrates, the Fgf subfamilies further expanded to contain three or four members in each subfamily.


Subject(s)
Evolution, Molecular , Fibroblast Growth Factors/genetics , Animals , Exons/genetics , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/physiology , Gene Deletion , Introns/genetics , Mice , Phylogeny
12.
Neurosci Lett ; 430(2): 147-50, 2008 Jan 10.
Article in English | MEDLINE | ID: mdl-18061349

ABSTRACT

The fibroblast growth factor (FGF) system has previously been found to be altered in post-mortem brains of individuals with major depressive disorder (MDD). The present study tested whether the FGF system is altered following acute social defeat. Rats were exposed to four consecutive days of either a social defeat paradigm or novel cages. Animals were sacrificed after the last social defeat session and gene expression was assessed in the hippocampus by mRNA in situ hybridization. Molecular components of the FGF system were significantly downregulated following social defeat. Specifically, FGF2 and FGFR1 mRNA expression was decreased in various subfields of the hippocampus. Decreased tone of the FGF system following an acute social stressor is congruent with human post-mortem results of FGF system downregulation in depression. These findings suggest that modulating the FGF system may have therapeutic value in the treatment of MDD.


Subject(s)
Down-Regulation/physiology , Fibroblast Growth Factors/metabolism , Social Behavior , Stress, Psychological/metabolism , Animals , Behavior, Animal , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/genetics , Hippocampus/metabolism , Male , Rats , Rats, Sprague-Dawley
13.
Mol Cell Biol ; 27(9): 3417-28, 2007 May.
Article in English | MEDLINE | ID: mdl-17339340

ABSTRACT

Unique among fibroblast growth factors (FGFs), FGF19, -21, and -23 act in an endocrine fashion to regulate energy, bile acid, glucose, lipid, phosphate, and vitamin D homeostasis. These FGFs require the presence of Klotho/betaKlotho in their target tissues. Here, we present the crystal structures of FGF19 alone and FGF23 in complex with sucrose octasulfate, a disaccharide chemically related to heparin. The conformation of the heparin-binding region between beta strands 10 and 12 in FGF19 and FGF23 diverges completely from the common conformation adopted by paracrine-acting FGFs. A cleft between this region and the beta1-beta2 loop, the other heparin-binding region, precludes direct interaction between heparin/heparan sulfate and backbone atoms of FGF19/23. This reduces the heparin-binding affinity of these ligands and confers endocrine function. Klotho/betaKlotho have evolved as a compensatory mechanism for the poor ability of heparin/heparan sulfate to promote binding of FGF19, -21, and -23 to their cognate receptors.


Subject(s)
Endocrine System , Fibroblast Growth Factors/metabolism , Glucuronidase/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Crystallography, X-Ray , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/genetics , Heparin/metabolism , Humans , Klotho Proteins , Mice , Models, Molecular , Molecular Sequence Data , Paracrine Communication , Protein Binding , Protein Structure, Tertiary , Sequence Alignment
14.
Int J Mol Med ; 16(5): 959-63, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16211270

ABSTRACT

We have previously reported comparative genomics analyses on FGF3, FGF4, FGF6, FGF7, FGF8, FGF10, FGF11, FGF17, FGF18, FGF19, FGF20, FGF22 and FGF23 genes. Here, we performed comparative genomics analyses on FGF1, FGF2, FGF5, FGF9, FGF12, FGF13, FGF14, FGF16 and FGF21 genes, and further characterized the FGF16 gene. Chimpanzee FGF16, chicken fgf16, and zebrafish fgf16 genes were identified within NW_121938.1, NW_060344.1, and CR855117.3 genome sequences, respectively. Chimpanzee FGF16 (207 aa), chicken fgf16 (207 aa), and zebrafish fgf16 (203 aa) showed 100%, 89.9%, and 79.2% total amino-acid identity with human FGF16. Because FGF16, FGF9, and FGF20 constitute FGF subfamily without N-terminal signal peptide, we next searched for uncharacterized FGF9 or FGF20 orthologs. Zebrafish fgf9 gene was identified within BX927112.11 genome sequence, and chicken fgf20 gene within NW_060349.1 genome sequence. Although N-terminal part was divergent, middle and C-terminal parts were well conserved among vertebrate FGF16, FGF9 and FGF20 orthologs. Phylogenetic analyses revealed that zebrafish fgf9 and fgf20 were more related to each other than to their chicken or mammalian orthologs. TCF/LEF binding site and TATA box were well conserved among the human FGF16, rat Fgf16, and mouse Fgf16 promoters. Because nuclear complex consisting of TCF/LEF (TCF1, TCF3, TCF4 or LEF1), beta-catenin, PYGO (PYGO1 or PYGO2) and Legless (BCL9 or BCL9L) binds to the TCF/LEF-binding site to up-regulate WNT/beta-catenin target genes, FGF16 gene was characterized as the evolutionarily conserved target of the WNT/beta-catenin signaling pathway, just like FGF18 and FGF20 genes. These facts indicate that FGF16, FGF18 and FGF20 are pharmacogenomics targets in the field of oncology and regenerative medicine.


Subject(s)
Fibroblast Growth Factors/classification , Fibroblast Growth Factors/genetics , Genome , Amino Acid Sequence , Animals , Base Sequence , Fibroblast Growth Factor-23 , Genomics , Humans , Mice , Molecular Sequence Data , Phylogeny , Promoter Regions, Genetic , Proteomics , Rats , Sequence Alignment
15.
Dev Cell ; 8(1): 85-95, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15621532

ABSTRACT

The epicardium regulates growth and survival of the underlying myocardium. This activity depends on intrinsic retinoic acid (RA) and erythropoietin signals. However, these signals do not act directly on the myocardium and instead are proposed to regulate the production of an unidentified soluble epicardial derived mitogen. Here, we show that Fgf9, Fgf16, and Fgf20 are expressed in the endocardium and epicardium and that RA can induce epicardial expression of Fgf9. Using knockout mice and an embryonic heart organ culture system, we show that endocardial and epicardial derived FGF signals regulate myocardial proliferation during midgestation heart development. We further show that this FGF signal is received by both FGF receptors 1 and 2 acting redundantly in the cardiomyoblast. In the absence of this signal, premature differentiation results in cellular hypertrophy and newborn mice develop a dilated cardiomyopathy. FGFs thus constitute all or part of the epicardial signal regulating myocardial growth and differentiation.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/radiation effects , Fibroblast Growth Factors/physiology , Gene Expression Regulation, Developmental/physiology , Myocardium/metabolism , Signal Transduction/physiology , Age Factors , Animals , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Embryo, Mammalian , Fibroblast Growth Factor 9 , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/deficiency , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental/drug effects , Heart/embryology , Immunohistochemistry/methods , In Situ Hybridization/methods , Mice , Mice, Knockout , Microscopy, Electron, Transmission/methods , Myocardium/ultrastructure , Organ Culture Techniques/methods , RNA, Messenger/biosynthesis , Receptors, Fibroblast Growth Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Tretinoin/pharmacology
16.
Dev Dyn ; 228(2): 267-72, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14517998

ABSTRACT

The inner ear, which mediates hearing and equilibrium, develops from an ectodermal placode located adjacent to the developing hindbrain. Induction of the placode and its subsequent morphogenesis and differentiation into the inner ear epithelium and its sensory neurons, involves signalling interactions within and between otic and non-otic tissues. Several members of the fibroblast growth factor (FGF) family play important roles at various stages of otic development; however, there are additional family members that have not been evaluated. In this study, we surveyed the expression patterns of 18 mouse Fgf and 3 Fgf receptor (Fgfr) genes during early otic development. Two members of the Fgf family, Fgf4 and Fgf16, and all three tested members of the Fgfr family, Fgfr2c, Fgfr3c, and Fgfr4, were expressed in tissues relevant to inner ear development. Fgf4 transcripts were expressed in the preplacodal and placodal ectoderm, suggesting potential roles in placode induction and/or maintenance. Fgf16 was expressed in the posterior otic cup and vesicle, suggesting roles in otic cell fate decisions and/or axis formation.


Subject(s)
Ear, Inner/embryology , Fibroblast Growth Factors/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Animals , Ear/embryology , Ectoderm/metabolism , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Hybridization , Mice , Molecular Sequence Data , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/classification , Receptors, Fibroblast Growth Factor/genetics
17.
Nat Neurosci ; 6(7): 701-7, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12766771

ABSTRACT

Dorsoventral patterning of the telencephalon is established early in forebrain development and underlies many of the regional subdivisions that are critical to the later organization of neural circuits in the cerebral cortex and basal ganglia. Sonic hedgehog (Shh) is involved in the generation of the ventral-most telencephalic cells, but the identity of the extrinsic signal(s) that induce dorsal character in telencephalic cells is not known. Here we show in chick embryos that sequential Wnt and fibroblast growth factor (FGF) signaling specifies cells of dorsal telencephalic character.


Subject(s)
Embryonic Induction/physiology , Fibroblast Growth Factors/physiology , Gene Expression Regulation, Developmental , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , Telencephalon/physiology , Zebrafish Proteins , Aging/genetics , Aging/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors , Bone Morphogenetic Proteins/physiology , Cell Differentiation , Chick Embryo , Ectoderm/cytology , Ectoderm/physiology , Epithelium , Eye Proteins , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/genetics , Frizzled Receptors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization/methods , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , PAX6 Transcription Factor , Paired Box Transcription Factors , Proteins/physiology , Proto-Oncogene Proteins/genetics , Pyrroles/pharmacology , Receptors, Fibroblast Growth Factor/physiology , Repressor Proteins , Signal Transduction/genetics , Stem Cells/metabolism , Telencephalon/cytology , Telencephalon/embryology , Telencephalon/metabolism , Transcription Factors , Wnt Proteins
18.
Dev Genes Evol ; 212(9): 432-8, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12373588

ABSTRACT

In vertebrates, a number of fibroblast growth factors (FGFs) have been shown to play important roles in developing embryos and adult organisms. However, the molecular relationships of the vertebrate FGFs are not yet completely understood, partly due to the divergence of their amino acid sequences. To solve this problem, we have identified six FGF genes in a basal chordate, the ascidian Ciona intestinalis. A phylogenetic analysis confidently assigned two of them to vertebrate FGF8/17/18 and FGF11/12/13/14, respectively. Based on the presence of the conserved domains within or outside of the FGF domains, we speculate that three of the other genes are orthologous to vertebrate FGF3/7/10/22, FGF4/5/6 and FGF9/16/20, respectively, although we cannot assign the sixth member to any of the vertebrate FGFs. A survey of the raw whole genome shotgun sequences of C. intestinalis demonstrated the presence of no FGF genes other than the six genes in the genome. The identification of these six FGF genes in the basal chordate gave us an insight into the diversification of specific subfamilies of vertebrate FGFs.


Subject(s)
Ciona intestinalis/genetics , Fibroblast Growth Factors/genetics , Amino Acid Sequence , Animals , Ciona intestinalis/classification , Ciona intestinalis/physiology , DNA, Complementary/analysis , Fibroblast Growth Factors/classification , Genes , Molecular Sequence Data , Phylogeny , Sequence Homology, Amino Acid
20.
Expert Opin Ther Targets ; 6(4): 469-82, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12223061

ABSTRACT

The fibroblast growth factor (FGF) family of signalling molecules and its receptors (FGFRs) contribute to normal developmental and physiological processes. However, the subversion of this powerful growth stimulatory pathway has been implicated in the generation of a variety of pathological conditions. This review focuses on the role of FGF/FGFRs in cancer. The case will be made that this signalling pathway is associated with and functionally important for the growth of some human tumours. As such, FGF/FGFRs can be viewed as rational therapeutic oncology targets and strategies used to inhibit these molecules are discussed. The therapeutic exploitation of tumour-associated FGFR expression to deliver toxins or antiproliferative signals to tumour cells is also reviewed, as is the use of FGFs as protein therapeutics to alleviate the side effects of cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Fibroblast Growth Factors/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Receptors, Fibroblast Growth Factor/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Cell Division/drug effects , Drug Carriers , Drug Delivery Systems , Drug Screening Assays, Antitumor , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/analysis , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/physiology , Fibroblast Growth Factors/therapeutic use , Humans , Neoplasm Proteins/analysis , Neoplasm Proteins/physiology , Neoplasms/physiopathology , Receptors, Fibroblast Growth Factor/physiology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...