Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cell Cycle ; 18(24): 3562-3580, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31735119

ABSTRACT

Osteoporosis and sarcopenia (osteosarcopenia (OS)) are twin-aging diseases. The biochemical crosstalk between muscle and bone seems to play a role in OS. We have previously shown that osteocytes produce soluble factors with beneficial effects on muscle and vice versa. Recently, enhanced FGF9 production was observed in the OmGFP66 osteogenic cell line. To test its role in myogenic differentiation, C2C12 myoblasts were treated with recombinant FGF9. FGF9 as low as 10 ng/mL inhibited myogenic differentiation, suggesting that FGF9 might be a potential inhibitory factor produced from bone cells with effects on muscle cells. FGF9 (10-50 ng/mL) significantly decreased mRNA expression of MyoG and Mhc while increasing the expression of Myostatin. Consistent with the phenotype, RT-qPCR array revealed that FGF9 (10 ng/mL) increased the expression of Icam1 while decreased the expression of Wnt1 and Wnt6 decreased, respectively. FGF9 decreased caffeine-induced Ca2+ release from the sarcoplasmic reticulum (SR) of C2C12 myotubes and reduced the expression of genes (i.e. Cacna1s, RyR2, Naftc3) directly associated with intracellular Ca2+ homeostasis. Myogenic differentiation in human skeletal muscle cells was similarly inhibited by FGF9 but required higher doses of 200 ng/mL FGF9. FGF9 was also shown to stimulate C2C12 myoblast proliferation. FGF2 and the FGF9 subfamily members FGF16 and FGF20 also inhibited C2C12 myoblast differentiation and enhanced proliferation. Intriguingly, the differentiation inhibition was independent of proliferation enhancement. These findings suggest that FGF9 may modulate myogenesis via a complex signaling mechanism.


Subject(s)
Fibroblast Growth Factor 9/physiology , Muscle Development/physiology , Muscle Fibers, Skeletal/cytology , Animals , Caffeine/pharmacology , Calcium/metabolism , Cell Line , Cell Proliferation , Cells, Cultured , Fibroblast Growth Factors/physiology , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Muscle Development/genetics , Organelle Biogenesis , Regeneration , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Signal Transduction
2.
Anim Reprod Sci ; 197: 177-184, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30154034

ABSTRACT

Fibroblast growth factor 9 (FGF9) is an important signaling molecule in early gonadal development. Hu sheep are noted for reproductive precociousness and fertility. The present study was conducted to investigate the gene expression and functions of FGF9 in ovine testis steroidogenesis during sexual maturity. A 874 bp cDNA fragment of FGF9 was detected that included a 627 bp coding sequence, encoding 208 amino acids. The FGF9 amino acid sequence of sheep had high homology with this molecule of other mammalian species. Additionally, the abundance of FGF9 in ovine testis was greater (P < 0.05) at 9 months (M) and 24 M of age compared with those at 3 M. Immunohistochemistry further revealed that FGF9 mainly localized in the Leydig cells and that there were small amounts in elongating spermatids. The functions of FGF9 in sheep Leydig cells was investigated using a siRNA-FGF9. Secretion of T and abundance of testosterone synthesis-related enzymes in Leydig cells were inhibited (P < 0.05) by siRNA-FGF9. Thus, these results demonstrated FGF9 is an important regulator of testosterone biosynthesis in rams. Results of the present research provide a new perspective for genetic and molecular research on modulation of physiological mechanisms during sexual maturity in male sheep.


Subject(s)
Fibroblast Growth Factor 9/metabolism , Sexual Maturation/physiology , Sheep , Testis/physiology , Animals , Fertility , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factor 9/physiology , Leydig Cells , Male , Spermatids , Testis/embryology , Testis/metabolism , Testosterone
3.
Appl Microbiol Biotechnol ; 102(2): 605-613, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29198068

ABSTRACT

The fibroblast growth factor (FGF) 9 subfamily is a member of the FGF family, including FGF9, 16, and 20, potentially sharing similar biochemical functions due to their high degree of sequence homology. Unlike other secreted proteins which have a cleavable N-terminal secreted signal peptide, FGF9/16/20 have non-cleaved N-terminal signal peptides. As an intercellular signaling molecule, they are involved in a variety of complex responses in animal development. Cardiogenesis is controlled by many members of the transcription factor family. Evidence suggests that FGF signaling, including the FGF9 subfamily, has a pretty close association with these cardiac-specific genes. In addition, recent studies have shown that the FGF9 subfamily maintains functional adaptation and survival after myocardial infarction in adult myocardium. Since FGF9/16/20 are secreted proteins, their function characterization in cardiac regeneration can promote their potential to be developed for the treatment of cardioprotection and revascularization. Here, we conclude that the FGF9 subfamily roles in cardiac development and maintenance of postnatal cardiac homeostasis, especially cardiac function maturation and functional maintenance of the heart after injury.


Subject(s)
Fibroblast Growth Factor 9/classification , Fibroblast Growth Factor 9/physiology , Heart/physiology , Animals , Embryonic Development , Fibroblast Growth Factor 9/genetics , Gene Expression Regulation , Heart/physiopathology , Heart Diseases/physiopathology , Heart Diseases/therapy , Humans , Myocardial Infarction , Signal Transduction , Transcription Factors
4.
Biol Reprod ; 96(1): 122-133, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28395336

ABSTRACT

Male differentiation of primordial germ cells (PGCs) is initiated by the inhibition of entry into meiosis and exposure to male-inducing factor(s), which are regulated by somatic elements of the developing gonad. Fibroblast growth factor 9 (FGF9) produced by pre-Sertoli cells is essential for male gonadal differentiation and also contributes to survival and male differentiation of XY PGCs. However, it is not clear how FGF9 regulates PGC fate. Using a PGC culture system, we identified dose-dependent, fate-determining functions of FGF9 in XY PGCs. Treatment with low levels of FGF9 (0.2 ng/ml) increased expression of male-specific Dnmt3L and Nanos2 in XY PGCs. Conversely, treatment with high levels of FGF9 (25 ng/ml) suppressed male-specific gene expression and stimulated proliferation of XY PGCs. Western blotting showed that low FGF9 treatment enhanced p38 MAPK (mitogen-activated protein kinase) phosphorylation in the same cells. In contrast, high FGF9 treatment significantly stimulated the ERK (extracellular signal-regulated kinase)1/2 signaling pathway in XY PGCs. We investigated the relationship between the ERK1/2 signaling pathway stimulated by high FGF9 and regulation of PGC proliferation. An ERK1/2 inhibitor (U0126) suppressed the PGC proliferation that would otherwise be stimulated by high FGF9 treatment, and increased Nanos2 expression in XY PGCs. Conversely, a p38 MAPK inhibitor (SB202190) significantly suppressed Nanos2 expression that would otherwise be stimulated by low FGF9 in XY PGCs. Taken together, our results suggest that stage-specific expression of FGF9 in XY gonads regulates the balance between proliferation and differentiation of XY PGCs in a dose-dependent manner.


Subject(s)
Cell Differentiation , Fibroblast Growth Factor 9/physiology , Germ Cells/physiology , MAP Kinase Signaling System , Animals , Male , Mice , Primary Cell Culture
5.
Dev Biol ; 411(1): 72-84, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26794256

ABSTRACT

Fibroblast growth factor (FGF) signaling is a critical regulator of skeletal development. Fgf9 and Fgf18 are the only FGF ligands with identified functions in embryonic bone growth. Mice lacking Fgf9 or Fgf18 have distinct skeletal phenotypes; however, the extent of overlapping or redundant functions for these ligands and the stage-specific contributions of FGF signaling to chondrogenesis and osteogenesis are not known. To identify separate versus shared roles for FGF9 and FGF18, we generated a combined series of Fgf9 and Fgf18 null alleles. Analysis of embryos lacking alleles of Fgf9 and Fgf18 shows that both encoded ligands function redundantly to control all stages of skeletogenesis; however, they have variable potencies along the proximodistal limb axis, suggesting gradients of activity during formation of the appendicular skeleton. Congenital absence of both Fgf9 and Fgf18 results in a striking osteochondrodysplasia and revealed functions for FGF signaling in early proximal limb chondrogenesis. Additional defects were also noted in craniofacial bones, vertebrae, and ribs. Loss of alleles of Fgf9 and Fgf18 also affect the expression of genes encoding other key intrinsic skeletal regulators, including IHH, PTHLH (PTHrP), and RUNX2, revealing potential direct, indirect, and compensatory mechanisms to coordinate chondrogenesis and osteogenesis.


Subject(s)
Bone Development/genetics , Bone and Bones/embryology , Chondrogenesis/genetics , Fibroblast Growth Factor 9/physiology , Fibroblast Growth Factors/physiology , Osteochondrodysplasias/genetics , Osteogenesis/genetics , Animals , Bone and Bones/abnormalities , Cell Differentiation , Chondrocytes/metabolism , Core Binding Factor Alpha 1 Subunit/biosynthesis , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factors/genetics , Growth Plate/embryology , Hedgehog Proteins/biosynthesis , Mice , Mice, Knockout , Parathyroid Hormone-Related Protein/biosynthesis , Signal Transduction/genetics
6.
Am J Physiol Lung Cell Mol Physiol ; 310(7): L615-29, 2016 04 01.
Article in English | MEDLINE | ID: mdl-26773067

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by an accumulation of extracellular matrix proteins and fibroblasts in the distal airways. Key developmental lung signaling pathways are reactivated in IPF. For instance, fibroblast growth factor 9 (FGF9) and FGF18, involved in epithelial-mesenchymal interactions, are critical for lung development. We evaluated the expression of FGF9, FGF18, and FGF receptors (FGFRs) in lung tissue from controls and IPF patients and assessed their effect on proliferation, survival, migration, and differentiation of control and IPF human lung fibroblasts (HLFs). FGF9, FGF18, and all FGFRs were present in the remodeled alveolar epithelium close to the fibroblast foci in IPF lungs. FGFR3 was generally detected in fibroblast foci by immunohistochemistry. In vitro, HLFs mainly expressed mesenchyme-associated FGFR isoforms (FGFR1c and FGFR3c) and FGFR4. FGF9 did not affect fibroblast proliferation, whereas FGF18 inhibited cell growth in control fibroblasts. FGF9 and FGF18 decreased Fas-ligand-induced apoptosis in control but not in IPF fibroblasts. FGF9 prevented transforming growth factor ß1-induced myofibroblast differentiation. FGF9 and FGF18 increased the migratory capacities of HLF, and FGF9 actively modulated matrix metalloproteinase activity. In addition, FGFR3 inhibition by small interfering RNA impacted p-ERK activation by FGF9 and FGF18 and their effects on differentiation and migration. These results identify FGF9 as an antiapoptotic and promigratory growth factor on HLF, maintaining fibroblasts in an undifferentiated state. The biological effects of FGF9 and FGF18 were partially driven by FGFR3. FGF18 was a less potent molecule. Both growth factors likely contribute to the fibrotic process in vivo.


Subject(s)
Fibroblast Growth Factor 9/physiology , Fibroblast Growth Factors/physiology , Myofibroblasts/physiology , Receptors, Fibroblast Growth Factor/metabolism , Aged , Case-Control Studies , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Gene Expression , Humans , Idiopathic Pulmonary Fibrosis , Lung/metabolism , Lung/pathology , Middle Aged
7.
Int J Oncol ; 46(1): 423-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25353145

ABSTRACT

Cancer cells are surrounded by the extracellular fluid, matrix, and stroma cells. Little is known about how extracellular components such as growth factor ligands affect the biology of lung cancer cells. The objective of this study was to determine whether extracellular fibroblast growth factors (FGFs) can affect the biology of lung cancer cells and to understand how extracellular FGFs affect the biology of lung cancer cells, including non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) cells. Out of the 23 reported FGFs, we focused on FGF2, FGF9 and FGF10. We examined the effect of FGFs on proliferation, treatment sensitivity, and apoptosis of NSCLC (PC9) and SCLC (H69, H82 and H146) cells in vitro. To determine which FGF was the most clinically relevant, we also examined FGF2 and FGF9 concentrations in the serum of patients with lung cancer. We found that extracellular FGFs can affect proliferation, treatment sensitivity, and apoptosis of lung cancer cells in a cell-specific manner. Our results indicate that extracellular FGFs affect the biology of lung cancer cells through multiple functions.


Subject(s)
Fibroblast Growth Factors/physiology , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Extracellular Space , Fibroblast Growth Factor 2/blood , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 2/physiology , Fibroblast Growth Factor 9/blood , Fibroblast Growth Factor 9/pharmacology , Fibroblast Growth Factor 9/physiology , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/pharmacology , Humans , Lung Neoplasms/blood , MAP Kinase Signaling System/drug effects , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Small Cell Lung Carcinoma/blood , Small Cell Lung Carcinoma/pathology , Tumor Cells, Cultured
9.
Dev Cell ; 22(6): 1191-207, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22698282

ABSTRACT

The identity of niche signals necessary to maintain embryonic nephron progenitors is unclear. Here we provide evidence that Fgf20 and Fgf9, expressed in the niche, and Fgf9, secreted from the adjacent ureteric bud, are necessary and sufficient to maintain progenitor stemness. Reduction in the level of these redundant ligands in the mouse led to premature progenitor differentiation within the niche. Loss of FGF20 in humans, or of both ligands in mice, resulted in kidney agenesis. Sufficiency was shown in vitro where Fgf20 or Fgf9 (alone or together with Bmp7) maintained isolated metanephric mesenchyme or sorted nephron progenitors that remained competent to differentiate in response to Wnt signals after 5 or 2 days in culture, respectively. These findings identify a long-sought-after critical component of the nephron stem cell niche and hold promise for long-term culture and utilization of these progenitors in vitro.


Subject(s)
Cell Differentiation , Fibroblast Growth Factor 9/physiology , Fibroblast Growth Factors/physiology , Animals , Bone Morphogenetic Protein 7/physiology , Congenital Abnormalities/genetics , Female , Fibroblast Growth Factors/genetics , Humans , Kidney/abnormalities , Kidney/growth & development , Kidney Diseases/congenital , Kidney Diseases/genetics , Male , Mesenchymal Stem Cells/physiology , Mice , Mutation , Nephrons/growth & development , Organ Culture Techniques , Stem Cell Niche/physiology , Wnt Signaling Pathway
10.
Prostate Cancer Prostatic Dis ; 15(1): 8-14, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22006051

ABSTRACT

BACKGROUND: Fibroblast growth factor 9 (FGF9) enhances cell proliferation and invasiveness in several malignant diseases. The aim of the present study is to investigate the role of FGF9 in postoperative recurrence after radical prostatectomy. METHODS: Cell viability and invasion of LNCaP cells were assessed using MTT assay and Matrigel invasion assay, respectively, in the presence or absence of treatment with recombinant FGF9. Tissues obtained during a radical prostatectomy in 133 male patients were immunohistochemically stained using anti-FGF9 antibody. RESULTS: Cell viability and invasion of LNCaP was significantly enhanced by treatment with recombinant FGF9. Immunohistochemical staining detected FGF9-positive cells in 20 samples. The prevalence of FGF9-positive cells in cases with a Gleason score of 8 or higher was 34.2%, which was significantly higher than that in those with Gleason scores of 7 or lower (7.3%, P=0.0003), respectively. The 3-year biochemical relapse-free survival rate was 17.5% in cases with FGF9-positive cells, which was significantly lower than that in cases in which FGF9-positive cells were not detectable (75.5%, P < 0.0001). CONCLUSIONS: These results indicate that FGF9 can stimulate proliferation and invasion in prostate cancer cells, thus FGF9 could be a candidate of a predictive factor for recurrence after radical prostatectomy.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/physiology , Fibroblast Growth Factor 9/physiology , Neoplasm Recurrence, Local , Prostatic Neoplasms/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/surgery , Aged , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Cytoplasm/metabolism , Disease-Free Survival , Fibroblast Growth Factor 9/metabolism , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Grading , Proportional Hazards Models , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/surgery , Statistics, Nonparametric
12.
Dev Cell ; 19(3): 440-9, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20833365

ABSTRACT

Sex determination of mammalian germ cells occurs during fetal development and depends on signals from gonadal somatic cells. Previous studies have established that retinoic acid (RA) triggers ovarian germ cells to enter meiosis and thereby commit to oogenesis, whereas in the developing testis, the enzyme CYP26B1 degrades RA and germ cells are not induced to enter meiosis. Using in vitro and in vivo models, we demonstrate that fibroblast growth factor 9 (FGF9) produced in the fetal testis acts directly on germ cells to inhibit meiosis; in addition, FGF9 maintains expression of pluripotency-related genes and upregulates markers associated with male germ cell fate. We conclude that two independent and mutually antagonistic pathways involving RA and FGF9 act in concert to determine mammalian germ cell sexual fate commitment and support a model in which the mitosis/meiosis switch is robustly controlled by both positive and negative regulatory factors.


Subject(s)
Fibroblast Growth Factor 9/physiology , Germ Cells/physiology , Meiosis/physiology , Pluripotent Stem Cells/metabolism , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Differentiation/drug effects , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Female , Fetus/cytology , Fetus/drug effects , Fetus/metabolism , Fluorescent Antibody Technique , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oogenesis/drug effects , Oogenesis/physiology , Ovary/embryology , Ovary/metabolism , RNA, Messenger/genetics , Retinoic Acid 4-Hydroxylase , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Testis/embryology , Testis/metabolism , Tretinoin/pharmacology
13.
Proc Natl Acad Sci U S A ; 107(26): 11853-8, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20547837

ABSTRACT

Bone healing requires a complex interaction of growth factors that establishes an environment for efficient bone regeneration. Among these, FGFs have been considered important for intrinsic bone-healing capacity. In this study, we analyzed the role of Fgf-9 in long bone repair. One-millimeter unicortical defects were created in tibias of Fgf-9(+/-) and wild-type mice. Histomorphometry revealed that half-dose gene of Fgf-9 markedly reduced bone regeneration as compared with wild-type. Both immunohistochemistry and RT-PCR analysis revealed markedly decreased levels of proliferating cell nuclear antigen (PCNA), Runt-related transcription factor 2 (Runx2), osteocalcin, Vega-a, and platelet endothelial cell adhesion molecule 1 (PECAM-1) in Fgf-9(+/-) defects. muCT angiography indicated dramatic impairment of neovascularization in Fgf-9(+/-) mice as compared with controls. Treatment with FGF-9 protein promoted angiogenesis and successfully rescued the healing capacity of Fgf-9(+/-) mice. Importantly, although other pro-osteogenic factors [Fgf-2, Fgf-18, and bone morphogenic protein 2 (Bmp-2)] still were present in Fgf-9(+/-) mice, they could not compensate for the haploinsufficiency of the Fgf-9 gene. Therefore, endogenous Fgf-9 seems to play an important role in long bone repair. Taken together our data suggest a unique role for Fgf-9 in bone healing, presumably by initiating angiogenesis through Vegf-a. Moreover, this study further supports the embryonic phenotype previously observed in the developing limb, thus promoting the concept that healing processes in adult organisms may recapitulate embryonic skeletal development.


Subject(s)
Fibroblast Growth Factor 9/physiology , Neovascularization, Physiologic/physiology , Osteogenesis/physiology , Animals , Biomarkers/metabolism , Bone Regeneration/genetics , Bone Regeneration/physiology , Female , Fibroblast Growth Factor 9/deficiency , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factor 9/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Osteogenesis/drug effects , Osteogenesis/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Recombinant Proteins/pharmacology , Tibia/blood supply , Tibia/drug effects , Tibia/injuries , Tibia/physiology , Vascular Endothelial Growth Factor A/pharmacology
14.
J Appl Genet ; 50(4): 347-60, 2009.
Article in English | MEDLINE | ID: mdl-19875885

ABSTRACT

Genetic control of gonadal development proceeds through either the male or female molecular pathways, driving bipotential gonadal anlage differentiation into a testis or ovary. Antagonistic interactions between the 2 pathways determine the gonadal sex. Essentially sex determination is the enhancement of one of the 2 pathways according to genetic sex. Initially, Sry with other factors upregulates Sox9 expression in XY individuals. Afterwards the expression of Sox9 is maintained by a positive feedback loop with Fgf9 and prostaglandin D2 as well as by autoregulative ability of Sox9. If these factors reach high concentrations, then Sox9 and/or Fgf9 may inhibit the female pathway. Surprisingly, splicing, nuclear transport, and extramatrix proteins may be involved in sex determination. The male sex determination pathway switches on the expression of genes driving Sertoli cell differentiation. Sertoli cells orchestrate testicular differentiation. In the absence of Sry, the predomination of the female pathway results in the realization of a robust genetic program that drives ovarian differentiation.


Subject(s)
Mammals/genetics , Sex Differentiation/genetics , Animals , Disorders of Sex Development , Female , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factor 9/physiology , Genes, sry , Male , Mammals/physiology , Models, Genetic , Ovary/embryology , Paracrine Communication , Prostaglandin D2/physiology , SOX9 Transcription Factor/genetics , Sex Determination Processes , Sex Differentiation/physiology , Testis/embryology , Transcription Factors/genetics
15.
Development ; 136(11): 1813-21, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19429785

ABSTRACT

Activation by the Y-encoded testis determining factor SRY and maintenance of expression of the Sox9 gene encoding the central transcription factor of Sertoli cell differentiation are key events in the mammalian sexual differentiation program. In the mouse XY gonad, SOX9 upregulates Fgf9, which initiates a Sox9/Fgf9 feedforward loop, and Sox9 expression is stimulated by the prostaglandin D2 (PGD2) producing lipocalin prostaglandin D synthase (L-PGDS, or PTDGS) enzyme, which accelerates commitment to the male pathway. In an attempt to decipher the genetic relationships between Sox9 and the L-Pgds/PGD2 pathway during mouse testicular organogenesis, we found that ablation of Sox9 at the onset or during the time window of expression in embryonic Sertoli cells abolished L-Pgds transcription. By contrast, L-Pgds(-/-) XY embryonic gonads displayed a reduced level of Sox9 transcript and aberrant SOX9 protein subcellular localization. In this study, we demonstrated genetically that the L-Pgds/PGD2 pathway acts as a second amplification loop of Sox9 expression. Moreover, examination of Fgf9(-/-) and L-Pgds(-/-) XY embryonic gonads demonstrated that the two Sox9 gene activity amplifying pathways work independently. These data suggest that, once activated and maintained by SOX9, production of testicular L-PGDS leads to the accumulation of PGD2, which in turn activates Sox9 transcription and nuclear translocation of SOX9. This mechanism participates together with FGF9 as an amplification system of Sox9 gene expression and activity during mammalian testicular organogenesis.


Subject(s)
Fibroblast Growth Factor 9/physiology , Prostaglandin D2/metabolism , SOX9 Transcription Factor/metabolism , Sertoli Cells/physiology , Sex Differentiation/physiology , Testis/embryology , Active Transport, Cell Nucleus/physiology , Animals , Cell Nucleus/metabolism , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Male , Mice , Mutation , Prostaglandin D2/genetics , SOX9 Transcription Factor/genetics , Sertoli Cells/cytology , Sex-Determining Region Y Protein/metabolism , Testis/cytology , Testis/growth & development
17.
Dev Biol ; 314(1): 71-83, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18155190

ABSTRACT

In mammals, sex is determined in the bipotential embryonic gonad by a balanced network of gene actions which when altered causes disorders of sexual development (DSD, formerly known as intersex). In the XY gonad, presumptive Sertoli cells begin to differentiate when SRY up-regulates SOX9, which in turn activates FGF9 and PGDS to maintain its own expression. This study identifies a new and essential component of FGF signaling in sex determination. Fgfr2 mutant XY mice on a mixed 129/C57BL6 genetic background had either normal testes, or developed ovotestes, with predominantly testicular tissue. However, backcrossing to C57BL6 mice resulted in a wide range of gonadal phenotypes, from hypoplastic testes to ovotestes with predominantly ovarian tissue, similar to Fgf9 knockout mice. Since typical male-specific FGF9-binding to the coelomic epithelium was abolished in Fgfr2 mutant XY gonads, these results suggest that FGFR2 acts as the receptor for FGF9. Pgds and SOX9 remained expressed within the testicular portions of Fgfr2 mutant ovotestes, suggesting that the Prostaglandin pathway acts independently of FGFR2 to maintain SOX9 expression. We could further demonstrate that double-heterozygous Fgfr2/Sox9 knockout mice developed ovotestes, demonstrating that both Fgfr2 and Sox9 can act as modifier intersex genes in the heterozygous state. In summary, we provide evidence that FGFR2 is important for male sex determination in mice, thereby rendering human FGFR2 a candidate gene for unsolved DSD cases such as 10q26 deletions.


Subject(s)
Disorders of Sex Development , Receptor, Fibroblast Growth Factor, Type 2/physiology , Sex Determination Processes , Animals , Female , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factor 9/physiology , Gonads/cytology , Gonads/embryology , High Mobility Group Proteins/genetics , High Mobility Group Proteins/physiology , Male , Mice , Mice, Knockout , Receptor, Fibroblast Growth Factor, Type 2/genetics , SOX9 Transcription Factor , Sertoli Cells/cytology , Sertoli Cells/physiology , Transcription Factors/genetics , Transcription Factors/physiology
18.
Biochem J ; 410(1): 205-11, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18039182

ABSTRACT

Two independent gain-of-function point mutations (S252W and P253R) in the extracellular region of the FGFR2 (fibroblast growth factor receptor 2) increase the binding affinity for the growth factor. The effect of this enhanced growth factor binding by these mutants is expected to be an increase in activation of regular signalling pathways from FGFR2 as a result of more receptors being engaged by ligand at any given time. Using PC12 (pheochromocytoma) cells as a model cell system we investigated the effect of these mutations on protein phosphorylation including the receptor, the activation of downstream signalling pathways and cell differentiation. Our results show that the effects of both of these extracellular mutations have unexpected intracellular phenotypes and cellular responses. Receptor phosphorylation was altered in both the ligand-stimulated and unstimulated states. The mutants also resulted in differential phosphorylation of a number of intracellular proteins. Both mutations resulted in enhanced ERK1/2 (extracellular-signalregulated kinase1/2) activation. Although ERK1/2 activation is believed to transduce signals resulting in cell differentiation, this response was abrogated in the cells expressing the mutant receptors. The results of the present study demonstrate that single extracellular point mutations in the FGFR2 have a profound effect on intracellular signalling and ultimately on cell fate.


Subject(s)
Cell Differentiation/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/cytology , Point Mutation , Receptor, Fibroblast Growth Factor, Type 2/genetics , Animals , Cell Lineage , Enzyme Activation , Fibroblast Growth Factor 9/physiology , PC12 Cells , Phosphorylation , Rats , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction
19.
Dev Biol ; 307(2): 300-13, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17544391

ABSTRACT

Gain-of-function mutations in fibroblast growth factor (FGF) receptors result in chondrodysplasia and craniosynostosis syndromes, highlighting the critical role for FGF signaling in skeletal development. Although the FGFRs involved in skeletal development have been well characterized, only a single FGF ligand, FGF18, has been identified that regulates skeletal development during embryogenesis. Here we identify Fgf9 as a second FGF ligand that is critical for skeletal development. We show that Fgf9 is expressed in the proximity of developing skeletal elements and that Fgf9-deficient mice exhibit rhizomelia (a disproportionate shortening of proximal skeletal elements), which is a prominent feature of patients with FGFR3-induced chondrodysplasia syndromes. Although Fgf9 is expressed in the apical ectodermal ridge in the limb bud, we demonstrate that the Fgf9-/- limb phenotype results from loss of FGF9 functions after formation of the mesenchymal condensation. In developing stylopod elements, FGF9 promotes chondrocyte hypertrophy at early stages and regulates vascularization of the growth plate and osteogenesis at later stages of skeletal development.


Subject(s)
Bone Development/physiology , Chondrocytes/cytology , Chondrogenesis/physiology , Fibroblast Growth Factor 9/physiology , Animals , Body Patterning , Bone Development/genetics , Bone and Bones/blood supply , Bone and Bones/embryology , Cell Differentiation , Chondrogenesis/genetics , Embryo Culture Techniques , Fibroblast Growth Factor 9/deficiency , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/physiology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Mice , Mice, Knockout , Models, Biological , Osteogenesis/genetics , Osteogenesis/physiology , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/metabolism , Phenotype , Receptor, Fibroblast Growth Factor, Type 3/deficiency , Receptor, Fibroblast Growth Factor, Type 3/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
20.
PLoS Biol ; 4(6): e187, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16700629

ABSTRACT

The genes encoding members of the wingless-related MMTV integration site (WNT) and fibroblast growth factor (FGF) families coordinate growth, morphogenesis, and differentiation in many fields of cells during development. In the mouse, Fgf9 and Wnt4 are expressed in gonads of both sexes prior to sex determination. Loss of Fgf9 leads to XY sex reversal, whereas loss of Wnt4 results in partial testis development in XX gonads. However, the relationship between these signals and the male sex-determining gene, Sry, was unknown. We show through gain- and loss-of-function experiments that fibroblast growth factor 9 (FGF9) and WNT4 act as opposing signals to regulate sex determination. In the mouse XY gonad, Sry normally initiates a feed-forward loop between Sox9 and Fgf9, which up-regulates Fgf9 and represses Wnt4 to establish the testis pathway. Surprisingly, loss of Wnt4 in XX gonads is sufficient to up-regulate Fgf9 and Sox9 in the absence of Sry. These data suggest that the fate of the gonad is controlled by antagonism between Fgf9 and Wnt4. The role of the male sex-determining switch--Sry in the case of mammals--is to tip the balance between these underlying patterning signals. In principle, sex determination in other vertebrates may operate through any switch that introduces an imbalance between these two signaling pathways.


Subject(s)
Fibroblast Growth Factor 9/physiology , Proto-Oncogene Proteins/physiology , Sex Determination Processes , Signal Transduction , Wnt Proteins/physiology , Animals , Female , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factor 9/metabolism , Gene Expression Regulation, Developmental , Gonads/cytology , Gonads/embryology , Gonads/metabolism , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Male , Mice , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , SOX9 Transcription Factor , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt4 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...