Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Asian J Androl ; 21(3): 309-318, 2019.
Article in English | MEDLINE | ID: mdl-30719983

ABSTRACT

While hallmarks of rodent spermatogonia stem cell biomarkers' heterogeneity have recently been identified, their stage and subset distributions remain unclear. Furthermore, it is currently difficult to accurately identify subset-specific SSC marker distributions due to the poor nuclear morphological characteristics associated with fixation in 4% paraformaldehyde. In the present study, testicular cross-sections and whole-mount samples were Bouin fixed to optimize nuclear resolution and visualized by immunohistochemistry (IHC) and immunofluorescence (IF). The results identified an expression pattern of PLZFhighc-KITpos in A1 spermatogonia, while A2-A4-differentiating spermatogonia were PLZFlowc-KITpos. Additionally, this procedure was used to examine asymmetrically expressing GFRA1 and PLZF clones, asymmetric Apr and false clones were distinguished based on the presence or absence of TEX14, a molecular maker of intercellular bridges, despite having identical nuclear morphology and intercellular distances that were <25 µm. In conclusion, this optimized Bouin fixation procedure facilitates the accurate identification of spermatogonium subsets based on their molecular profiles and is capable of distinguishing asymmetric and false clones. Therefore, the findings presented herein will facilitate further morphological and functional analysis studies and provide further insight into spermatogonium subtypes.


Subject(s)
Promyelocytic Leukemia Zinc Finger Protein/genetics , Proto-Oncogene Proteins c-kit/genetics , Spermatogonia/metabolism , Animals , Cell Differentiation , Fluorescent Antibody Technique , Gene Expression Regulation/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Promyelocytic Leukemia Zinc Finger Protein/biosynthesis , Proto-Oncogene Proteins c-kit/biosynthesis , Seminiferous Tubules/cytology , Spermatogenesis , Testis/cytology , Tissue Fixation , Transcription Factors/genetics
2.
Minerva Urol Nefrol ; 70(1): 79-86, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28651425

ABSTRACT

BACKGROUND: Urothelial carcinoma (UC) is a major health problem in the general population. We aimed to evaluate the function of GFRα3 and unravel its underlying molecular mechanism to develop novel treatment options equivalent to UC. METHODS: To evaluate the function of GFRα3, a group of 60 pairs of UC patients were recruited in for this study. UC tissues and their adjacent normal control tissues (NCTs) were collected between 2012 and 2015. We used immunohistochemistry to analyze the correlation between GFRα3 expression and clinicopathologic variables and patient survival. The role of regulation of GFRα3 in UC was applied in vitro. In addition, we further investigated the signaling pathway of GFRα3 in UC progression. RESULTS: The expression level of GFRα3 was remarkably upregulated in 49.3% (19/60) patients and downregulated in 25.0% (15/60) patients. The GFRα3 protein expression was upregulated in UC tissues. GFRα3 promotes UC cell migration and invasion in vitro. GFRα3 also promotes UC cell metastasis in vitro. High level of GFRα3 promotes UC cell migration via upregulation of MMP9 expression. CONCLUSIONS: Our results demonstrate that increased GFRα3 expression is significantly correlated with poor prognosis of patients with UC. Thus, GFRα3 might be an important marker and a therapeutic target for UC.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Urologic Neoplasms/genetics , Urologic Neoplasms/pathology , Adult , Aged , Animals , Biomarkers, Tumor , Cell Line, Tumor , Female , Humans , Male , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred BALB C , Middle Aged , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Transplantation , Prognosis , Real-Time Polymerase Chain Reaction
3.
Cell Death Dis ; 7: e2140, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26962690

ABSTRACT

Spermatogonial stem cells (SSCs) are unique male germline stem cells that support spermatogenesis and male fertility. Long non-coding RNAs (lncRNA) have been identified as key regulators of stem cell fate; however, their role in SSCs has not been explored. Here, we report that a novel spermatogonia-specific lncRNA (lncRNA033862) is essential for the survival of murine SSCs. LncRNA033862 is expressed in early spermatogonia including SSC and was among 805 lncRNAs identified by global expression profiling as responsive to glial cell-derived neurotrophic factor (GDNF), a growth factor required for SSC self-renewal and survival. LncRNA033862 is an antisense transcript of the GDNF receptor alpha1 (Gfra1) that lacks protein coding potential and regulates Gfra1 expression levels by interacting with Gfra1 chromatin. Importantly, lncRNA033862 knockdown severely impairs SSC survival and their capacity to repopulate recipient testes in a transplantation assay. Collectively, our data provide the first evidence that long non-coding RNAs (lncRNAs) regulate SSC fate.


Subject(s)
Adult Stem Cells/metabolism , Chromatin/metabolism , Gene Expression Regulation/physiology , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , RNA, Long Noncoding/metabolism , Adult Stem Cells/cytology , Animals , Cell Survival/physiology , Chromatin/genetics , Gene Knockdown Techniques , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Male , Mice , RNA, Long Noncoding/genetics
4.
Neoplasia ; 18(3): 162-71, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26992917

ABSTRACT

Crizotinib is the first anaplastic lymphoma kinase (ALK) inhibitor to have been approved for the treatment of non-small cell lung cancer (NSCLC) harboring an ALK fusion gene, but it has been found that, in the clinic, patients develop resistance to it. Alectinib and ceritinib are second-generation ALK inhibitors which show remarkable clinical responses in both crizotinib-naive and crizotinib-resistant NSCLC patients harboring an ALK fusion gene. Despite their impressive activity, clinical resistance to alectinib and ceritinib has also emerged. In the current study, we elucidated the resistance mechanisms to these second-generation ALK inhibitors in the H3122 NSCLC cell line harboring the EML4-ALK variant 1 fusion in vitro. Prolonged treatment of the parental H3122 cells with alectinib and ceritinib led to two cell lines which are 10 times less sensitive to alectinib and ceritinib than the parental H3122 cell line. Although mutations of ALK in its kinase domain are a common resistance mechanism for crizotinib, we did not detect any ALK mutation in these resistant cell lines. Rather, overexpression of phospho-ALK and alternative receptor tyrosine kinases such as phospho-EGFR, phospho-HER3, and phospho-IGFR-1R was observed in both resistant cell lines. Additionally, NRG1, a ligand for HER3, is upregulated and responsible for resistance by activating the EGFR family pathways through the NRG1-HER3-EGFR axis. Combination treatment with EGFR inhibitors, in particular afatinib, was shown to be effective at overcoming resistance. Our study provides new mechanistic insights into adaptive resistance to second-generation ALK inhibitors and suggests a potential clinical strategy to combat resistance to these second-generation ALK inhibitors in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/genetics , ErbB Receptors/biosynthesis , Neuregulin-1/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Anaplastic Lymphoma Kinase , Carbazoles/administration & dosage , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Humans , Mutation , Neuregulin-1/genetics , Oncogene Proteins, Fusion/genetics , Piperidines/administration & dosage , Pyrimidines/administration & dosage , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, ErbB-3/biosynthesis , Signal Transduction/drug effects , Sulfones/administration & dosage
5.
BMC Cancer ; 16: 225, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26984265

ABSTRACT

BACKGROUND: A large number of epigenetic alterations has been found to be implicated in the etiology of gastric cancer. We have studied the DNA methylation status of 27 500 gene promoter regions in 24 gastric adenocarcinomas from a Norwegian cohort, and aimed at identifying the hypermethylated regions. We have compared our findings to the gene expression in the same tissue, and linked our results to prognosis and survival. METHODS: Biopsies from gastric adenocarcinomas and adjacent normal gastric mucosa were obtained from 24 patients following surgical resection of the tumor. Genome-wide DNA methylation profiling of the tumor and matched non-cancerous mucosa was performed. The results were compared to whole transcriptome cDNA microarray analysis of the same material. RESULTS: Most of the gene promoter regions in both types of tissue showed a low degree of methylation, however there was a small, but significant hypermethylation of the tumors. Hierarchical clustering showed separate grouping of the tumor and normal tissue. Hypermethylation of the promoter region of the GFRA3 gene showed a strong correlation to post-operative survival and several of the clinicopathological parameters, however no difference was found between the two main histological types of gastric cancer. There was only a modest correlation between the DNA methylation status and gene expression. CONCLUSIONS: The different DNA methylation clusters of the tumors and normal tissue indicate that aberrant DNA methylation is a distinct feature of gastric cancer, although there is little difference in the overall, and low, methylation levels between the two tissue types. The GFRA3 promoter region showed marked hypermethylation in almost all tumors, and its correlation with survival and other clinicopathological parameters may have important prognostic significance.


Subject(s)
Biomarkers, Tumor/biosynthesis , DNA Methylation/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Stomach Neoplasms/surgery , Aged , Biomarkers, Tumor/genetics , CpG Islands , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Humans , Lymphatic Metastasis , Male , Middle Aged , Postoperative Period , Prognosis , Promoter Regions, Genetic , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
6.
J Cutan Med Surg ; 20(2): 130-4, 2016.
Article in English | MEDLINE | ID: mdl-26337382

ABSTRACT

BACKGROUND: Vitiligo is an idiopathic skin disease, characterized by circumscribed white macules or patches on the skin due to loss of the functional melanocytes. Glial cell line-derived neurotrophic factor (GDNF) and its cognate receptor (GFRα-1) are distal members of the transforming growth factor-ß superfamily. GDNF, produced by the basal cell keratinocytes, is involved in the migration and differentiation of the melanocytes from the neural crest to the epidermis. This study examines the hypothesis that expression of GDNF protein and its cognate receptor GFRα-1 protein is altered in vitiliginous skin. PATIENTS AND METHODS: To test our hypothesis, we examined the expression patterns of these proteins in vitiliginous and corresponding healthy (control) skin biopsies (20 specimens each) using immunoperoxidase staining techniques. RESULTS: We found variations between the vitiliginous skin and healthy skin. In healthy skin, the expression of GDNF and GFRα-1 proteins was strong (basal cell keratinocytes and melanocytes), moderate (spinous layer), and weak (granular cell layer). In contrast, weak expression of GDNF protein was observed in all epidermal layers of vitiliginous skin. GFRα-1 protein expression was strong (basal cell keratinocytes and melanocytes), moderate (spinous layer), and weak (granular cell layer). In both healthy skin and vitiliginous skin, the expression of GDNF and GFRα-1 proteins was strong in the adnexal structures. CONCLUSIONS: We report, for the first time, decreased expression of GDNF proteins in the epidermal keratinocytes of vitiliginous skin. Our findings suggest possible pathogenetic roles for these proteins in the development of vitiligo. The clinical ramifications of these observations mandate further investigations.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Immunohistochemistry/methods , Skin/pathology , Vitiligo/metabolism , Adult , Cell Differentiation , Cells, Cultured , Female , Humans , Male , Middle Aged , Reproducibility of Results , Skin/metabolism , Vitiligo/pathology
7.
Andrology ; 2(4): 615-22, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24919455

ABSTRACT

Protease activated receptor-2 (PAR-2) is the receptor for the prototype mast cell product tryptase. PAR-2 expression by cells of the human germinal epithelium was reported, but the exact cellular sites of testicular expression remained unknown. That became of interest, because mast cells, expressing tryptase, were found in the walls of seminiferous tubules of patients suffering from sub- and infertility. This location suggested that mast cells via tryptase might be able to influence PAR-2-expressing cells in the germinal epithelium. To explore these points, we used testicular paraffin-embedded sections for immunohistochemistry. PAR-2-positive cells were mostly basally located cells of the seminiferous epithelium, namely spermatogonia. Some stained for the receptor for GDNF (GFRalpha-1), and possibly represent spermatogonial stem cells (SSCs). As true human SSCs could not be examined, we turned to TCam-2 seminoma cells, expressing PAR-2 and stem cell markers, including GFRalpha-1. TCam-2 cells robustly responded to stimulation with a specific PAR-2 agonist (SLIGKV) by increased intracellular Ca(2+) levels. Recombinant tryptase and trypsin, but not a control peptide (VKGILS) evoked this response, implying functional PAR-2. Video imaging and caspase 3/7 assays showed that SLIGKV and tryptase prevented spontaneous apoptosis and increased proliferation of TCam-2 cells. The expression of the marker of pluripotency OCT3/4 was unchanged upon activation of PAR-2, suggesting that the stem cell-like character is not changed. Furthermore, human germ cell cancers were examined. A subset of seminoma and carcinoma in situ samples expressed PAR-2, indicating that yet unknown subgroups exist. Collectively, the descriptive data obtained in human testicular sections, in germ cell cancers and the functional results in TCam-2 cells imply a trophic role of mast cell-derived tryptase for human germ cells. This may be relevant for subtypes of human germ cell cancers, and possibly SSCs. It also raises the possibility that PAR-2 agonists might be useful for the in vitro propagation of human SSCs.


Subject(s)
Germ Cells/metabolism , Infertility, Male/physiopathology , Mast Cells/physiology , Receptor, PAR-2/biosynthesis , Seminiferous Epithelium/metabolism , Biopsy , Cells, Cultured , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Humans , Infertility, Male/pathology , Male , Neoplasms, Germ Cell and Embryonal/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Seminoma/metabolism , Testis/pathology , Tryptases/metabolism
8.
Neuroreport ; 24(13): 704-11, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-23921591

ABSTRACT

The glial cell-line derived neurotrophic factor (GDNF) is crucial for ureteric bud morphogenesis, spermatogenesis, and development of the enteric nervous system and is a potent survival factor for various neuronal populations. However, the impact of GDNF, at least on cell survival, was found to depend strongly on the presence of transforming growth factor ß (TGF-ß). In this study, we investigate the role of TGF-ß in GDNF-induced neuronal differentiation. In a cell culture paradigm of N2aGT cells (neuroblastoma cell line), we show that TGF-ß signaling localizes the GDNF ligand-binding receptor GFRa1 to the cell surface, which is a known mechanism by which TGF-ß is able to facilitate GDNF signaling. TGF-ß-mediated GDNF signaling slightly elevated the phosphorylation state of Ret, the canonical coreceptor for the GPI-linked (glycosyl-phosphatidylinositol) GFRa1. On the basis of morphological as well as immunocytological data, we finally show that GDNF-mediated neuronal differentiation is intensified when GDNF and TGF-ß act in concert.


Subject(s)
Cell Differentiation/physiology , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Neurons/physiology , Transforming Growth Factor beta/pharmacology , Animals , Biotinylation , Blotting, Western , Cell Line , Cell Membrane/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Immunohistochemistry , Immunoprecipitation , Mice , Neurites/drug effects , Phosphorylation , Proto-Oncogene Proteins c-ret/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Transforming Growth Factor beta/drug effects , Smad2 Protein/metabolism
9.
Proc Natl Acad Sci U S A ; 110(31): 12685-90, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23858447

ABSTRACT

The retinoblastoma tumor suppressor gene Rb is essential for maintaining the quiescence and for regulating the differentiation of somatic stem cells. Inactivation of Rb in somatic stem cells typically leads to their overexpansion, often followed by increased apoptosis, defective terminal differentiation, and tumor formation. However, Rb's roles in germ-line stem cells have not been explored. We conditionally disrupted the Rb gene in mouse germ cells in vivo and discovered unanticipated consequences for GFRa1-protein-expressing A(single) (GFRa1(+) A(s)) spermatogonia, the major source of male germ-line stem cells. Rb-deficient GFRa1(+) A(s) spermatogonia were present at normal density in testes 5 d after birth, but they lacked the capacity for self-renewal, resulting in germ cell depletion by 2 mo of age. Rb deficiency did not affect the proliferative activity of GFRa1(+) A(s) spermatogonia, but their progeny were exclusively transit-amplifying progenitor spermatogonia and did not include GFRa1(+) A(s) spermatogonia. In addition, Rb deficiency caused prolonged proliferation of progenitor spermatogonia, transiently enlarging this population. Despite these defects, Rb deficiency did not block terminal differentiation into functional sperm; offspring were readily obtained from young males whose germ cell pool was not yet depleted. We conclude that Rb is required for self-renewal of germ-line stem cells, but contrary to its critical roles in somatic stem cells, it is dispensable for their proliferative activity and terminal differentiation. Thus, this study identifies an unexpected function for Rb in maintaining the stem cell pool in the male germ line.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , Retinoblastoma Protein/metabolism , Spermatogonia/metabolism , Stem Cells/metabolism , Animals , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Male , Mice , Mice, Transgenic , Retinoblastoma Protein/genetics , Spermatogonia/cytology , Stem Cells/cytology
10.
Pharm Biol ; 51(9): 1188-95, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23767459

ABSTRACT

CONTEXT: Docosahexaenoic acid (DHA) is one of the critical fatty acids for optimal health, which affect the expression of nerve growth factor and brain-derived neurotrophic factor in brain. OBJECTIVE: This study investigates whether DHA supplementation affects lipid peroxidation and activates the glial-derived neurotrophic factor (GDNF)-mitogen-activated protein kinase pathway (MAPK pathway) in hippocampus of natural aged rat. MATERIALS AND METHODS: Rats were randomly divided into four groups; DHA was orally administered at 80 and 160 mg/kg/day to 24-month female rats for 50 days. The antioxidant parameters and GDNF-GDNF family receptor α-1 (GFRα1)-tyrosine-protein kinase receptor (RET)-MAPK-cyclic AMP response element-binding protein (CERB) pathway were assayed in natural aged rat's hippocampus. RESULTS AND DISCUSSION: The results demonstrated that DHA supplementation significantly increased the activities of superoxide dismutase (SOD) by 37.39 and 57.69%, glutathione peroxidase (GSH-Px) by 27.62 and 32.57% decreased TBARS level by 28.49 and 49.05%, respectively, but did not significantly affect catalase (CAT), in hippocampus, when compared with the aged group. DHA supplementation in diet resulted in an increase of DHA level in hippocampus. Furthermore, we found that DHA supplementation markedly increased the levels of GDNF and GFRα1 and the phosphorylation of RET, and led to the activation of the MAPK pathway in hippocampus tissue. CONCLUSION: DHA supplementation can change fatty acids composition, improve antioxidant parameters and activate the GDNF-MAPK pathway in natural aged rat's hippocampus.


Subject(s)
Aging/metabolism , Dietary Supplements , Docosahexaenoic Acids/therapeutic use , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor/agonists , Hippocampus/metabolism , Neuroprotective Agents/therapeutic use , Animals , Cyclic AMP Response Element-Binding Protein/agonists , Cyclic AMP Response Element-Binding Protein/metabolism , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/metabolism , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Hippocampus/enzymology , Hippocampus/growth & development , Lipid Peroxidation , MAP Kinase Signaling System , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/metabolism , Neurons/enzymology , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Oxidative Stress , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Phosphorylation , Protein Processing, Post-Translational , Random Allocation , Rats , Rats, Sprague-Dawley , Up-Regulation
11.
Int J Androl ; 35(5): 758-68, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22519471

ABSTRACT

The glial cell line-derived neurotrophic factor (GDNF) has multiple functions that promote cell survival, proliferation and migration in different cell types. The experimental over-expression of GDNF in mouse testis leads to infertility and promotes seminomatous germ cell tumours in older animals, which suggests that deregulation of the GDNF pathway may be implicated in germ cell carcinogenesis. GDNF activates downstream pathways upon binding to its specific co-receptor GDNF family receptor-a 1 (GFRA1). This complex then interacts with Ret and other co-receptors to activate several intracellular signalling cascades. To explore the involvement of the GDNF pathway in the onset and progression of testicular germ cell tumours, we analysed GFRA1 and Ret expression patterns in seminoma samples. We demonstrated, via immunohistochemistry, that GFRA1, but not Ret, is over-expressed in in situ carcinoma (CIS) and in intratubular and invasive seminoma cells compared with normal human germ cells. Functional analysis of the GDNF biological activity was performed on TCam-2 seminoma cell line. Reverse transcription-PCR (RT-PCR) and immunohistochemical analyses demonstrate that TCam-2 cells express both GFRA1 and Ret mRNA, but only GFRA1 was detected at the protein level. In TCam-2 cells, although GDNF is not mitogenic, it is able to induce migration, as demonstrated by a Boyden chamber assay, possibly through the Src and MEK pathways. Moreover, GDNF promotes invasive behaviour, an effect dependent on pericellular protease activity, possibly through the activity of matrix metalloproteinases. GFRA1 over-expression in CIS and seminoma cells, along with the functional analyses in TCam-2 cells, suggests an involvement of the GDNF pathway in the progression of testicular germ cell cancer.


Subject(s)
Seminoma/pathology , Adult , Carcinoma in Situ/metabolism , Cell Line, Tumor , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Humans , Male , Middle Aged , Neoplasm Invasiveness/physiopathology , Proto-Oncogene Proteins c-ret/biosynthesis , RNA, Messenger/metabolism , Seminoma/metabolism , Testicular Neoplasms/pathology
12.
Anat Rec (Hoboken) ; 295(3): 532-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22266674

ABSTRACT

This study aimed to explore the effect of high glucose concentration on the expression of glial cell line-derived neurotrophic factor (GDNF) and its family ligand receptors (GFRs) GFRα1 and GFRα2 in Müller cells and the protective role of GDNF in cultured Müller cells under high glucose circumstance. Cultured Müller cells (untreated or treated with 200 ng/mL of GDNF) were exposed to high glucose conditions (20 mmol/L glucose). We found that the expression levels of GDNF and GFRα1 mRNA and protein increased gradually over time under high glucose and exogenous GDNF-treated conditions, whereas the upregulation in GFRα2 expression was observed only in the early stage of high glucose conditions. Exogenous GDNF not only decreased apoptosis in cultured Müller cells under high glucose circumstance, but also accelerated the levels and speed of synthesis of GDNF and GFRα1 proteins in Müller cells. These results suggest that Müller cells can synthesize GDNF and GFRs under high glucose conditions, and GDNF may play important role in protecting Müller cells during the early stage of diabetic retinopathy. The difference in GFRs expression indicated that GDNF and neurturin may exert different effects on Müller cells under high glucose circumstance.


Subject(s)
Diabetic Retinopathy/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Hyperglycemia/metabolism , Hyperglycemia/pathology , Neuroglia/metabolism , Retina/metabolism , Animals , Animals, Newborn , Diabetic Retinopathy/pathology , Diabetic Retinopathy/physiopathology , Disease Models, Animal , Down-Regulation/genetics , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/antagonists & inhibitors , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Hyperglycemia/physiopathology , Neuroglia/pathology , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Retina/pathology , Retina/physiopathology , Up-Regulation/genetics
13.
J Dent Res ; 90(10): 1240-5, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21828353

ABSTRACT

This study investigated the effects of glial cell line-derived neurotrophic factor (GDNF) on dental pulp cells (DPCs). Cultures of DPCs expressed GDNF as well as its receptors, GFRα1 and RET. Addition of recombinant GDNF to cultures in serum-containing medium did not significantly affect DPC growth; however, GDNF dose-dependently increased viable cell number under serum-free culture conditions. Live/dead, lactate dehydrogenase (LDH), and caspases-3/-7 assays demonstrated that cell death occurred under serum-free conditions, and that GDNF significantly reduced the number of dead cells by inhibiting apoptotic cell death. GDNF also stimulated cell proliferation in serum-free conditions, as assessed by the BrdU incorporation assay. The effect of GDNF was abolished in the presence of inhibitors to GFRα1 and RET suggesting receptor-mediated events. This study also demonstrated that GDNF counteracted TNFα-induced DPC cytotoxicity, suggesting that GDNF may be cytoprotective under disease conditions. In conclusion, our findings indicate that GDNF promotes cell survival and proliferation of DPCs and suggest that GDNF may play a multifunctional role in the regulation of dental pulp homeostasis.


Subject(s)
Apoptosis , Dental Pulp/cytology , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Proto-Oncogene Proteins c-ret/physiology , Animals , Cell Proliferation , Cell Survival , Cells, Cultured , Culture Media, Serum-Free , Dental Pulp/metabolism , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Humans , Male , Proto-Oncogene Proteins c-ret/biosynthesis , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/antagonists & inhibitors
14.
Am J Physiol Lung Cell Mol Physiol ; 300(5): L790-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21335521

ABSTRACT

We combined retrograde tracing techniques with single-neuron RT-PCR to compare the expression of neurotrophic factor receptors in nodose vs. jugular vagal sensory neurons. The neurons were further categorized based on location of their terminals (tracheal or lungs) and based on expression of the ionotropic capsaicin receptor TRPV1. Consistent with functional studies, nearly all jugular neurons innervating the trachea and lungs expressed TRPV1. With respect to the neurotrophin receptors, the TRPV1-expressing jugular C-fiber neurons innervating both the trachea and lung compartments preferentially expressed tropomyosin-receptor kinase A (TrkA), with only a minority of neurons expressing TrkB or TrkC. The nodose neurons that express TRPV1 (presumed nodose C-fibers) innervate mainly intrapulmonary structures. These neurons preferentially expressed TrkB, with only a minority expressing TrkA or TrkC. The expression pattern in tracheal TRPV1-negative neurons, nodose tracheal presumed Aδ-fiber neurons as well as the intrapulmonary TRPV1-negative presumed Aß-fiber neurons, was similar to that observed in the nodose C-fiber neurons. We also evaluated the expression of GFRα receptors and RET (receptors for the GDNF family ligands). Virtually all vagal sensory neurons innervating the respiratory tract expressed RET and GFRα1. The jugular neurons also categorically expressed GFRα3, as well as ∼50% of the nodose neurons. GFRα2 was expressed in ∼50% of the neurons irrespective of subtype. The results reveal that Trk receptor expression in vagal afferent neurons innervating the adult respiratory tract depends more on the location of the cell bodies (jugular vs. nodose ganglion) than either the location of the terminals or the functional phenotype of the nerve. The data also reveal that in addition to neurotrophins, the GDNF family ligands may be important neuromodulators of vagal afferent nerves innervating the adult respiratory tract.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Lung/innervation , Nodose Ganglion/physiology , Receptors, Nerve Growth Factor/biosynthesis , Trachea/innervation , Animals , Guinea Pigs , Male , Receptor, trkA/biosynthesis , Receptor, trkB/biosynthesis , Receptor, trkC/biosynthesis , Sensory Receptor Cells , TRPV Cation Channels/biosynthesis
15.
J Neurochem ; 116(4): 486-98, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21133924

ABSTRACT

Many of the cellular effects of glial cell line-derived neurotrophic factor are initiated by binding to GNDF family receptor alpha-1 (GFRα1), and mediated by diverse intracellular signaling pathways, most notably through the Ret tyrosine kinase. Ret may be activated by the cell autonomous expression of GFRα1 ('in cis'), or by its non-cell autonomous presence ('in trans'), in either a soluble or immobilized state. GFRα1 is expressed in the striatum, a target of the dopaminergic projection of the substantia nigra. To determine whether post-synaptic expression of GFRα1 in striatum in trans has effects on the development or adult responses to injury of dopamine neurons, we have created transgenic mice in which GFRα1 expression is selectively increased in striatum and other forebrain targets of the dopaminergic projection. Post-synaptic GFRα1 has profound effects on the development of dopamine neurons, resulting in a 40% increase in their adult number. This morphologic effect was associated with an augmented motor response to amphetamine. In adult mice, post-synaptic GFRα1 expression did not affect neuron survival following neurotoxic lesion, but it did increase the preservation of striatal dopaminergic innervation. We conclude that post-synaptic striatal GFRα1 expression has important effects on the biology of dopamine neurons in vivo.


Subject(s)
Corpus Striatum/growth & development , Corpus Striatum/injuries , Dopamine/physiology , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Substantia Nigra/growth & development , Substantia Nigra/injuries , Animals , Cell Differentiation/genetics , Corpus Striatum/pathology , Gene Expression Regulation, Developmental , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Neurons/pathology , Neurons/physiology , Rats , Substantia Nigra/pathology
16.
J Neurosci ; 30(48): 16272-83, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21123573

ABSTRACT

We have shown recently that following saphenous nerve transection and successful regeneration, cutaneous polymodal nociceptors (CPMs) lacking transient receptor potential vanilloid 1 (TRPV1) are sensitized to heat stimuli and that mechanically insensitive, heat-sensitive C-fibers (CHs) that contain TRPV1 increase in prevalence. Target-derived neurotrophic factor levels were also enhanced after axotomy and regeneration. In particular, the glial-cell line-derived neurotrophic factor (GDNF) family member artemin was found to be significantly enhanced in the hairy hindpaw skin and its receptor GDNF family receptor α3 (GFRα3) was increased in the L2/L3 dorsal root ganglia (DRGs) following nerve injury. In this study, we assessed the role of enhanced artemin/GFRα3 levels on the changes in mouse cutaneous CH neurons following saphenous nerve regeneration. We used a newly developed siRNA-mediated in vivo knockdown strategy to specifically inhibit the injury-induced expression of GFRα3 and coupled this with an ex vivo recording preparation to examine response characteristics and neurochemical phenotype of different types of functionally defined neurons after injury. We found that inhibition of GFRα3 did not affect the axotomy-induced decrease in CPM threshold, but transiently prevented the recruitment of CH neurons. Western blot and real-time PCR analysis of hairy hindpaw skin and L2/L3 DRGs after saphenous nerve regeneration suggested that inhibition of the potential initial injury-induced increase in enhanced target-derived artemin signaling resulted in dynamic changes in TRPV1 expression after regeneration. These changes in TRPV1 expression may underlie the functional alterations observed in CH neurons after nerve regeneration.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Hot Temperature , Nerve Fibers, Unmyelinated/physiology , Nerve Regeneration/physiology , Nerve Tissue Proteins/metabolism , Recruitment, Neurophysiological/physiology , Animals , Axotomy , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Male , Mice , Nerve Tissue Proteins/biosynthesis , Physical Stimulation/methods , TRPV Cation Channels/biosynthesis , Up-Regulation/physiology
17.
J Neurochem ; 115(3): 707-15, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20731758

ABSTRACT

We recently reported that age-related bradykinesia was associated with reduced dopamine (DA) tissue content, ser31 tyrosine hydroxylase (TH) phosphorylation, and total TH levels in substantia nigra (SN) only. In this study, we propose that these decreases result from reduced glial cell line-derived neurotrophic factor family receptor α-1 (GFR α-1) levels in the aged (30-month-old) cohort of rats. Analysis of GFR α-1 receptor protein in SN, striatum, ventral tegmental area, and nucleus accumbens from 12- and 30-month-old Brown-Norway/Fischer 344 F(1) hybrid rats revealed immunoreactivity at ∼48 and 52 kDa, bands previously characterized to correspond to soluble and glycosyl-phosphatidylinositol-linked forms of GFR α-1, respectively. The nigrostriatal pathway had significantly greater levels of the soluble GFR α-1 than the mesoaccumbens pathway. Aging significantly reduced soluble and total GFR α-1 in the SN. The levels of GFR α-1 significantly correlated with TH protein in SN, striatum, and nucleus accumbens, but only in the SN did GFR α-1 significantly correlate with DA levels. Based on these observations and findings from the literature, we speculate that (i) GFR α-1 receptor expression may regulate nigral DA bioavailability in vivo, (ii) age-related decreases in soluble GFR α-1 in SN may contribute to bradykinesia in aging, and (iii) differences in expression of the GFR α-1 forms between the nigrostriatal and mesoaccumbens pathways and allied tissue may indicate that glial cell line-derived neurotrophic factor-signaling differs between these DA pathways.


Subject(s)
Aging/metabolism , Corpus Striatum/growth & development , Corpus Striatum/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Neural Pathways/growth & development , Neural Pathways/metabolism , Nucleus Accumbens/growth & development , Nucleus Accumbens/metabolism , Substantia Nigra/growth & development , Substantia Nigra/metabolism , Animals , Blotting, Western , Dopamine/physiology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Kinetics , Male , Motor Activity/physiology , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Rats , Rats, Inbred BN , Rats, Inbred F344 , Signal Transduction/physiology , Tyrosine 3-Monooxygenase/metabolism
18.
Exp Neurol ; 226(1): 136-47, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20713051

ABSTRACT

The objective was to analyze functional effects of the combination of GDNF and TGF-ß1 in the retrograde model of Parkinsonism in rats, based on the intrastriatal infusion of 6-hydroxydopamine, which leads to protracted and progressive cell death in the substantia nigra. Hemiparkinsonian rats were implanted with osmotic minipumps 2 months after striatal lesion, pumps delivering GDNF alone (10 ng/day), TGF-ß1 alone (2 ng/day), or a GDNF and TGF-ß1 combination. The findings confirmed that GDNF alone has potent dopaminotrophic effects but they also revealed, for the first time, that GDNF and TGF-ß1 co-infusion led to stronger trophic effects relative to the infusion of GDNF alone. TGF-ß1 allowed further reducing dopamine receptor hypersensitivity, and potentiated GDNF-mediated effects. This cooperation could be accounted for by the recruitment of GFRα1 on striatal membranes, and by enhanced expression and activation of TH through augmented pSer31TH and pSer40TH. Co-infusion induced striatal sprouting, as revealed by augmentation of p21-Arc, stathmin, and synaptophysin, and led to a reliable recovery of phenotypic expression of TH in surviving nigral neurons. Functional recovery and improvement of TH signal in the nigrostriatal system were long-lasting and sustained, remaining after cessation of trophic infusion.


Subject(s)
Antiparkinson Agents , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Parkinson Disease, Secondary/drug therapy , Transforming Growth Factor beta1/pharmacology , Algorithms , Amphetamine , Animals , Apomorphine/pharmacology , Behavior, Animal/drug effects , Blotting, Western , Central Nervous System Stimulants , Dopamine Agonists/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Immunohistochemistry , Infusion Pumps, Implantable , Male , Microinjections , Neostriatum , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/pathology , Rats , Rats, Wistar , Stereotyped Behavior , Sympatholytics , Transforming Growth Factor beta1/administration & dosage
19.
J Comp Neurol ; 518(15): 3024-45, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20533358

ABSTRACT

Most small unmyelinated neurons in adult rat dorsal root ganglia (DRG) express one or more of the coreceptors targeted by glial cell line-derived neurotrophic factor (GDNF), neurturin, and artemin (GFRalpha1, GFRalpha2, and GFRalpha3, respectively). The function of these GDNF family ligands (GFLs) is not fully elucidated but recent evidence suggests GFLs could function in sensory neuron regeneration after nerve injury and peripheral nociceptor sensitization. In this study we used immunohistochemistry to determine if the DRG neurons targeted by each GFL change after sciatic nerve injury. We compared complete sciatic nerve transection and the chronic constriction model and found that the pattern of changes incurred by each injury was broadly similar. In lumbar spinal cord there was a widespread increase in neuronal GFRalpha1 immunoreactivity (IR) in the L1-6 dorsal horn. GFRalpha3-IR also increased but in a more restricted area. In contrast, GFRalpha2-IR decreased in patches of superficial dorsal horn and this loss was more extensive after transection injury. No change in calcitonin gene-related peptide-IR was detected after either injury. Analysis of double-immunolabeled L5 DRG sections suggested the main effect of injury on GFRalpha1- and GFRalpha3-IR was to increase expression in both myelinated and unmyelinated neurons. In contrast, no change in basal expression of GFRalpha2-IR was detected in DRG by analysis of fluorescence intensity and there was a small but significant reduction in GFRalpha2-IR neurons. Our results suggest that the DRG neuronal populations targeted by GDNF, neurturin, or artemin and the effect of exogenous GFLs could change significantly after a peripheral nerve injury.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology , Animals , Cell Count , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Immunohistochemistry , Male , Nerve Fibers/pathology , Nerve Tissue Proteins/metabolism , Neurturin/metabolism , Pain Measurement , Phenotype , Posterior Horn Cells/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Up-Regulation
20.
Cells Tissues Organs ; 191(5): 372-81, 2010.
Article in English | MEDLINE | ID: mdl-20090300

ABSTRACT

Development of a culture system that supports self-renewal and proliferation of spermatogonial stem cells (SSCs) is enormously valuable for experimental research and potential treatment for male infertility. Although several research groups had reported their successes in SSC isolation and culture, the two current accepted culture systems are different in cell enrichment methods, serum and growth factors. Previous researches also indicated SSCs from different mouse strains required different culture conditions. Here we report for the first time that SSCs from BALB/c mice could be cultured in an improved culture system for 3 months. The modified culture system consisted of an improved enzymatic procedure, the enrichment of undifferentiated spermatogonia by differential adherence selection of isolated SSCs, mouse embryonic fibroblast feeder cells, StemPro-34 SFM medium supplemented with glial cell line-derived neurotrophic factor (GDNF), basic fibroblast growth factor and GDNF-family receptor alpha1 (GFRalpha1). The improved digestion method increased the viability and enrichment efficiency of isolated testis cells. Furthermore, basal culture medium with 10% fetal bovine serum as selected medium could increase the number of germ cell colonies in the initiation stage of culture. Cultured SSCs were characterized morphologically and formed typical colonies. Immunocytochemical staining and RT-PCR showed that cultured SSCs expressed Oct-4, GFRalpha1, Sox2 and several other special genes resembling undifferentiated spermatogonia. Spermatogonia transplantation further confirmed that cultured SSCs were functionally normal and could restore complete spermatogenesis. The culture methods described here could serve as a paradigm to establish conditions for the culture of SSCs from other species, allowing identification of universal factors necessary for proliferation of SSCs.


Subject(s)
Mice, Inbred BALB C/surgery , Spermatogonia/transplantation , Stem Cells/cytology , Animals , Culture Media , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Male , Mice , Octamer Transcription Factor-3/biosynthesis , Spermatogonia/cytology , Spermatogonia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...