Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 106
1.
Proc Jpn Acad Ser B Phys Biol Sci ; 98(3): 112-125, 2022.
Article En | MEDLINE | ID: mdl-35283407

The RET proto-oncogene encodes a receptor tyrosine kinase whose alterations are responsible for various human cancers and developmental disorders, including thyroid cancer, non-small cell lung cancer, multiple endocrine neoplasia type 2, and Hirschsprung's disease. RET receptors are physiologically activated by glial cell line-derived neurotrophic factor (GDNF) family ligands that bind to the coreceptor GDNF family receptor α (GFRα). Signaling via the GDNF/GFRα1/RET ternary complex plays crucial roles in the development of the enteric nervous system, kidneys, and urinary tract, as well as in the self-renewal of spermatogonial stem cells. In addition, another ligand, growth differentiation factor-15 (GDF15), has been shown to bind to GFRα-like and activate RET, regulating body weight. GDF15 is a stress response cytokine, and its elevated serum levels affect metabolism and anorexia-cachexia syndrome. Moreover, recent development of RET-specific kinase inhibitors contributed significantly to progress in the treatment of patients with RET-altered cancer. This review focuses on the broad roles of RET in development, metabolic diseases, and cancer.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Metabolic Diseases , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , Proto-Oncogene Proteins c-ret/physiology
2.
Biomolecules ; 11(6)2021 06 09.
Article En | MEDLINE | ID: mdl-34207842

Medullary thyroid cancer (MTC) is a neuroendocrine tumor that arises from the parafollicular C-cells, which produces the hormone calcitonin. RET is a transmembrane receptor protein-tyrosine kinase, which is highly expressed in MTC. Our previous studies reported that cyclin-dependent kinase 5 (CDK5) plays a crucial role in cancer progression, including MTC. However, the role of CDK5 in GDNF-induced RET signaling in medullary thyroid cancer proliferation remains unknown. Here, we investigated RET activation and its biochemically interaction with CDK5 in GDNF-induced medullary thyroid cancer proliferation. Our results demonstrated that GDNF stimulated RET phosphorylation and thus subsequently resulted in CDK5 activation by its phosphorylation. Activated CDK5 further caused STAT3 activation by its specific phosphorylation at Ser727. Moreover, we also found that GDNF treatment enhanced ERK1/2 and EGR1 activity, which is involved in p35 activation. Interestingly, we identified for the first time that CDK5 physically interacted with RET protein in MTC. Overall, our results provide a new mechanism for medullary thyroid cancer cell proliferation, suggesting that targeting CDK5 may be a promising therapeutic candidate for human medullary thyroid cancer in the near future.


Cyclin-Dependent Kinase 5/metabolism , Proto-Oncogene Proteins c-ret/metabolism , STAT3 Transcription Factor/metabolism , Carcinoma, Neuroendocrine/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Cyclin-Dependent Kinase 5/genetics , Early Growth Response Protein 1/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , MAP Kinase Signaling System/physiology , Phosphorylation , Proto-Oncogene Proteins c-ret/physiology , Receptor Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor/genetics , Signal Transduction , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
5.
J Clin Oncol ; 38(11): 1209-1221, 2020 04 10.
Article En | MEDLINE | ID: mdl-32083997

Activating receptor tyrosine kinase RET (rarranged during transfection) gene alterations have been identified as oncogenic in multiple malignancies. RET gene rearrangements retaining the kinase domain are oncogenic drivers in papillary thyroid cancer, non-small-cell lung cancer, and multiple other cancers. Activating RET mutations are associated with different phenotypes of multiple endocrine neoplasia type 2 as well as sporadic medullary thyroid cancer. RET is thus an attractive therapeutic target in patients with oncogenic RET alterations. Multikinase inhibitors with RET inhibitor activity, such as cabozantinib and vandetanib, have been explored in the clinic for tumors with activating RET gene alterations with modest clinical efficacy. As a result of the nonselective nature of these multikinase inhibitors, patients had off-target adverse effects, such as hypertension, rash, and diarrhea. This resulted in a narrow therapeutic index of these drugs, limiting ability to dose for clinically effective RET inhibition. In contrast, the recent discovery and clinical validation of highly potent selective RET inhibitors (pralsetinib, selpercatinib) demonstrating improved efficacy and a more favorable toxicity profile are poised to alter the landscape of RET-dependent cancers. These drugs appear to have broad activity across tumors with activating RET alterations. The mechanisms of resistance to these next-generation highly selective RET inhibitors is an area of active research. This review summarizes the current understanding of RET alterations and the state-of-the-art treatment strategies in RET-dependent cancers.


Neoplasms/drug therapy , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Drug Resistance, Neoplasm , Gene Rearrangement , Germ-Line Mutation , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/physiology
6.
Biosystems ; 189: 104099, 2020 Mar.
Article En | MEDLINE | ID: mdl-31935434

Many biological processes show switching behaviors in response to parameter changes. Although numerous surveys have been conducted on bifurcations in biological systems, they commonly focus on over-represented parts of signaling cascades, known as motifs, ignoring the multi-motif structure of biological systems and the communication links between these building blocks. In this paper, a method is proposed which partitions molecular interactions to modules based on a control theory point of view. The modules are defined so that downstream effect of one module is a regulator for its neighboring modules. Communication links between these modules are then considered as bifurcation parameters to reveal change in steady state status of each module. As a case-study, we generated a molecular interaction map of signaling molecules during the development of mammalian embryonic kidneys. The whole system was divided to modules, where each module is defined as a group of interacting molecules that result in expression of a vital downstream regulator. Bifurcation analysis was then performed on these modules by considering the communication signals as bifurcation parameters. Two-parameter bifurcation analysis was then performed to assess the effects of simultaneous input signals on each module behavior. In the case where a module had more than two inputs, a series of two parameter bifurcation diagrams were calculated each corresponding to different values of the third parameter. We detected multi-stability for RET protein as a key regulator for fate determination. This finding is in agreement with experimental data indicating that ureteric bud cells are bi-potential, able to form tip or trunk of the bud based on their RET activity level. Our findings also indicate that Glial cell-derived neurotrophic factor (GDNF), a known potent regulator of kidney development, exerts its fate-determination function on cell placement through destruction of saddle node bifurcation points in RET steady states and confining RET activity level to high activity in ureteric bud tip. In conclusion, embryonic cells usually show a huge decision making potential; the proposed modular modeling of the system in association with bifurcation analysis provides a quantitative holistic view of organ development.


Embryonic Development/physiology , Kidney/embryology , Kidney/physiology , Systems Biology/methods , Glial Cell Line-Derived Neurotrophic Factor/physiology , Humans , Morphogenesis/physiology , Proto-Oncogene Proteins c-ret/physiology
7.
J Dent Res ; 98(6): 705-712, 2019 06.
Article En | MEDLINE | ID: mdl-30958726

During organogenesis, the timing and patterning of dental pulp innervation require both chemoattractive and chemorepellent cues for precise spatiotemporal regulation. Our understanding of the signaling mechanisms that regulate tooth innervation during development, as well as the basic biology of these sensory neurons, remains rudimentary. In this study, we analyzed the expression and function of glial cell line-derived neurotrophic factor (GDNF) and its receptor tyrosine kinase, Ret, in the regulation of innervation of the mouse tooth pulp by dental pulpal afferent (DPA) neurons of the trigeminal ganglion (TG). Using reporter mouse models, we demonstrate that Ret is highly expressed by a subpopulation of DPA neurons projecting to the tooth pulp at both postnatal day 7 (P7) and in the adult. In the adult tooth, GDNF is highly expressed by many cell types throughout the dental pulp. Using a ubiquitous tamoxifen (TMX)-inducible Cre ( UBC-Cre/ERT2) line crossed to Ret conditional knockout mice ( Retfx/fx), Ret was deleted immediately prior to tooth innervation, and the neural projections into P7 molars were analyzed. TMX treatment was efficient in ablating >95% of Ret protein. We observed that UBC-Cre/ERT2; Retfx/fx mice had a significant reduction in the total number of neurites present within the pulp at P7, with a significant accumulation of aberrant fibers in the dental follicle and periodontium. In agreement with these findings, inhibition of Ret signaling through in vivo administration of a highly specific pharmacologic inhibitor (1NM-PP1) of Ret also caused a substantial reduction in pulpal innervation. Taken together, these findings indicate that Ret signaling regulates the timing and patterning of tooth innervation by dental primary afferent neurons of the TG during organogenesis and provide a rationale to explore whether alterations in the GDNF-Ret pathway contribute to pathophysiological conditions in the adult dentition.


Dental Pulp/innervation , Organogenesis , Proto-Oncogene Proteins c-ret/physiology , Tooth , Animals , Mice , Signal Transduction , Trigeminal Ganglion
8.
Nat Metab ; 1(10): 985-995, 2019 10.
Article En | MEDLINE | ID: mdl-32021964

Muscle undergoes progressive weakening and regenerative dysfunction with age due in part to the functional decline of skeletal muscle stem cells (MuSCs). MuSCs are heterogeneous but whether their gene expression changes with age and the implication of such changes are unclear. Here we show that in mice, Growth arrest-specific gene 1 (Gas1) is expressed in a small subset of young MuSCs with its expression progressively increasing in larger fractions of MuSCs later in life. Over-expression of Gas1 in young MuSCs and inactivation of Gas1 in aged MuSCs support that Gas1 reduces the quiescence and self-renewal capacity of MuSCs. Gas1 reduces Ret signaling, which is required for MuSC quiescence and self-renewal. Indeed, we show that the Ret ligand, Glial Cell-Derived Neurotrophic Factor (GDNF), can counteract Gas1 by stimulating Ret signaling and enhancing MuSC self-renewal and regeneration, thus improving muscle function. We propose that strategies aimed to target this pathway can be exploited to improve the regenerative decline of muscle stem cells.


Cell Cycle Proteins/genetics , Cell Self Renewal/genetics , Glial Cell Line-Derived Neurotrophic Factor/genetics , Muscle, Skeletal/cytology , Stem Cells/metabolism , Aging/drug effects , Animals , Cell Division , Female , GPI-Linked Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/growth & development , Proto-Oncogene Proteins c-ret/physiology , Regeneration/genetics , Regeneration/physiology , Signal Transduction , Transcriptome
9.
Oncogene ; 37(11): 1417-1429, 2018 03.
Article En | MEDLINE | ID: mdl-29321660

Activating mutations of the ALK receptor occur in a subset of neuroblastoma tumors. We previously demonstrated that Alk mutations cooperate with MYCN overexpression to induce neuroblastoma in mice and identified Ret as being strongly upregulated in MYCN/Alkmut tumors. By a genetic approach in vivo, we now document an oncogenic cooperation between activated Ret and MYCN overexpression in neuroblastoma formation. We show that MYCN/RetM919T tumors exhibit histological features and expression profiles close to MYCN/Alkmut tumors. We show that RET transcript levels decrease precedes RET protein levels decrease upon ALK inhibition in neuroblastoma cell lines. Etv5 was identified as a candidate transcription factor regulating Ret expression from murine MYCN/Alkmut tumor transcriptomic data. We demonstrate that ETV5 is regulated both at the protein and mRNA levels upon ALK activation or inhibition in neuroblastoma cell lines and that this regulation precedes RET modulation. We document that ALK activation induces ETV5 protein upregulation through stabilization in a MEK/ERK-dependent manner. We show that RNAi-mediated inhibition of ETV5 decreases RET expression. Reporter assays indicate that ETV5 is able to drive RET gene transcription. ChIP-seq analysis confirmed ETV5 binding on the RET promoter and identified an enhancer upstream of the promoter. Finally, we demonstrate that combining RET and ALK inhibitors reduces tumor growth more efficiently than each single agent in MYCN and AlkF1178L-driven murine neuroblastoma. Altogether, these results define the ERK-ETV5-RET pathway as a critical axis driving neuroblastoma oncogenesis downstream of activated ALK.


Anaplastic Lymphoma Kinase/genetics , Carcinogenesis/genetics , Gain of Function Mutation , Neuroblastoma/genetics , Anaplastic Lymphoma Kinase/metabolism , Animals , Carcinogenesis/pathology , Cells, Cultured , DNA-Binding Proteins/physiology , Female , Gain of Function Mutation/physiology , HEK293 Cells , Humans , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Neuroblastoma/pathology , Proto-Oncogene Proteins c-ret/physiology , Signal Transduction/genetics , Transcription Factors/physiology , Xenograft Model Antitumor Assays
10.
Radiother Oncol ; 124(3): 448-454, 2017 09.
Article En | MEDLINE | ID: mdl-28784438

BACKGROUND AND PURPOSE: Recently, stem cell therapy has been proposed to allow regeneration of radiation damaged salivary glands. It has been suggested that glial-cell-derived neurotrophic factor (GDNF) promotes survival of mice salivary gland stem cells (mSGSCs). The purpose of this study was to investigate the role of GDNF in the modulation of mSGSC response to irradiation and subsequent salivary gland regeneration. METHODS: Salivary gland sphere derived cells of Gdnf hypermorphic (Gdnfwt/hyper) and wild type mice (Gdnfwt/wt) were irradiated (IR) with γ-rays at 0, 1, 2, 4 and 8Gy. mSGSC survival and stemness were assessed by calculating surviving fraction measured as post-IR sphere forming potential and population doublings. Flow cytometry was used to determine the CD24hi/CD29hi stem cell (SC) population. QPCR and immunofluorescence was used to detect GDNF expression. RESULTS: The IR survival responses of mSGSCs were similar albeit resulted in larger spheres and an increased cell number in the Gdnfwt/hyper compared to Gdnfwt/wt group. Indeed, mSGSC of Gdnfwt/hyper mice showed high sphere forming efficiency upon replating. Interestingly, GDNF expression co-localized with receptor tyrosine kinase (RET) and was upregulated after IR in vitro and in vivo, but normalized in vivo after mSGSC transplantation. CONCLUSION: GDNF does not protect mSGSCs against irradiation but seems to promote mSGSCs proliferation through the GDNF-RET signaling pathway. Post-transplantation stimulation of GDNF/RET pathway may enhance the regenerative potential of mSGSCs.


Glial Cell Line-Derived Neurotrophic Factor/physiology , Salivary Glands/radiation effects , Stem Cells/radiation effects , Animals , Female , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-ret/physiology , Regeneration , Salivary Glands/cytology , Signal Transduction , Stem Cells/physiology
11.
PLoS One ; 12(2): e0171416, 2017.
Article En | MEDLINE | ID: mdl-28152033

BACKGROUND: Glial cell line-derived neurotrophic factor (GDNF) is a potent neurotrophic factor known to promote the survival and maintenance of neurons not only in the developing but also in the adult enteric nervous system. As diverticular disease (DD) is associated with reduced myenteric neurons, alterations of the GDNF system were studied in asymptomatic diverticulosis (diverticulosis) and DD. METHODS: Morphometric analysis for quantifying myenteric ganglia and neurons were assessed in colonic full-thickness sections of patients with diverticulosis and controls. Samples of tunica muscularis (TM) and laser-microdissected myenteric ganglia from patients with diverticulosis, DD and controls were analyzed for mRNA expression levels of GDNF, GFRA1, and RET by RT-qPCR. Myenteric protein expression of both receptors was quantified by fluorescence-immunohistochemistry of patients with diverticulosis, DD, and controls. RESULTS: Although no myenteric morphometric alterations were found in patients with diverticulosis, GDNF, GFRA1 and RET mRNA expression was down-regulated in the TM of patients with diverticulosis as well as DD. Furthermore GFRA1 and RET myenteric plexus mRNA expression of patients with diverticulosis and DD was down-regulated, whereas GDNF remained unaltered. Myenteric immunoreactivity of the receptors GFRα1 and RET was decreased in both asymptomatic diverticulosis and DD patients. CONCLUSION: Our data provide evidence for an impaired GDNF system at gene and protein level not only in DD but also during early stages of diverticula formation. Thus, the results strengthen the idea of a disturbed GDNF-responsiveness as contributive factor for a primary enteric neuropathy involved in the pathogenesis and disturbed intestinal motility observed in DD.


Diverticulum/physiopathology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Aged , Case-Control Studies , Colon/innervation , Colon/pathology , Diverticulum/pathology , Fluorescent Antibody Technique , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Humans , Laser Capture Microdissection , Male , Myenteric Plexus/pathology , Proto-Oncogene Proteins c-ret/physiology , Real-Time Polymerase Chain Reaction
12.
Leukemia ; 31(5): 1166-1176, 2017 05.
Article En | MEDLINE | ID: mdl-27780967

Acute myeloid leukemias (AMLs) result from a series of genetic events occurring in a stem or progenitor hematopoietic cell that gives rise to their clonal expansion and an impaired capacity to differentiate. To circumvent the genetic heterogeneity of AML patient cohorts, we have developed a model system, driven by the MLL-AF9 (MA9) oncogene, to generate multiple human leukemias using progenitor cells from a single healthy donor. Through stepwise RNA-sequencing data generated using this model and AML patients, we have identified consistent changes associated with MA9-driven leukemogenesis and demonstrate that no recurrent secondary mutations are required. We identify 39 biomarkers whose high expression level is specific to this genetic subtype of AML and validate that many of these have diagnostic utility. We further examined one biomarker, the receptor tyrosine kinase (RTK) RET, and show through shRNA knockdowns that its expression is essential for in vivo and in vitro growth of MA9-AML. These results highlight the value of novel human models of AML derived from single donors using specific oncogenic fusions to understand their biology and to uncover potential therapeutic targets.


Leukemia, Myeloid, Acute/pathology , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins c-ret/physiology , Animals , Biomarkers , Cell Line , Cell Line, Tumor , Cell Proliferation , Clone Cells/pathology , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/genetics , Mice , Models, Biological , Transfection
13.
J Neurosci ; 36(40): 10337-10355, 2016 10 05.
Article En | MEDLINE | ID: mdl-27707970

Axon-Schwann cell interactions are crucial for the development, function, and repair of the peripheral nervous system, but mechanisms underlying communication between axons and nonmyelinating Schwann cells are unclear. Here, we show that ER81 is functionally required in a subset of mouse RET+ mechanosensory neurons for formation of Pacinian corpuscles, which are composed of a single myelinated axon and multiple layers of nonmyelinating Schwann cells, and Ret is required for the maintenance of Er81 expression. Interestingly, Er81 mutants have normal myelination but exhibit deficient interactions between axons and corpuscle-forming nonmyelinating Schwann cells. Finally, ablating Neuregulin-1 (Nrg1) in mechanosensory neurons results in no Pacinian corpuscles, and an Nrg1 isoform not required for communication with myelinating Schwann cells is specifically decreased in Er81-null somatosensory neurons. Collectively, our results suggest that a RET-ER81-NRG1 signaling pathway promotes axon communication with nonmyelinating Schwann cells, and that neurons use distinct mechanisms to interact with different types of Schwann cells. SIGNIFICANCE STATEMENT: Communication between neurons and Schwann cells is critical for development, normal function, and regeneration of the peripheral nervous system. Despite many studies about axonal communication with myelinating Schwann cells, mostly via a specific isoform of Neuregulin1, the molecular nature of axonal communication with nonmyelinating Schwann cells is poorly understood. Here, we described a RET-ER81-Neuregulin1 signaling pathway in neurons innervating Pacinian corpuscle somatosensory end organs, which is essential for communication between the innervating axon and the end organ nonmyelinating Schwann cells. We also showed that this signaling pathway uses isoforms of Neuregulin1 that are not involved in myelination, providing evidence that neurons use different isoforms of Neuregulin1 to interact with different types of Schwann cells.


DNA-Binding Proteins/physiology , Neuregulin-1/physiology , Pacinian Corpuscles/growth & development , Pacinian Corpuscles/physiology , Proto-Oncogene Proteins c-ret/physiology , Signal Transduction/physiology , Transcription Factors/physiology , Animals , Axons/physiology , DNA-Binding Proteins/genetics , Mechanotransduction, Cellular/genetics , Mechanotransduction, Cellular/physiology , Mice , Mice, Inbred C57BL , Mutation/genetics , Myelin Sheath/physiology , Neuregulin-1/genetics , Neurons/physiology , Peripheral Nerve Injuries/physiopathology , Proto-Oncogene Proteins c-ret/genetics , Schwann Cells/physiology , Transcription Factors/genetics
14.
PLoS Biol ; 14(2): e1002382, 2016 Feb.
Article En | MEDLINE | ID: mdl-26894589

Branching morphogenesis of the epithelial ureteric bud forms the renal collecting duct system and is critical for normal nephron number, while low nephron number is implicated in hypertension and renal disease. Ureteric bud growth and branching requires GDNF signaling from the surrounding mesenchyme to cells at the ureteric bud tips, via the Ret receptor tyrosine kinase and coreceptor Gfrα1; Ret signaling up-regulates transcription factors Etv4 and Etv5, which are also critical for branching. Despite extensive knowledge of the genetic control of these events, it is not understood, at the cellular level, how renal branching morphogenesis is achieved or how Ret signaling influences epithelial cell behaviors to promote this process. Analysis of chimeric embryos previously suggested a role for Ret signaling in promoting cell rearrangements in the nephric duct, but this method was unsuited to study individual cell behaviors during ureteric bud branching. Here, we use Mosaic Analysis with Double Markers (MADM), combined with organ culture and time-lapse imaging, to trace the movements and divisions of individual ureteric bud tip cells. We first examine wild-type clones and then Ret or Etv4 mutant/wild-type clones in which the mutant and wild-type sister cells are differentially and heritably marked by green and red fluorescent proteins. We find that, in normal kidneys, most individual tip cells behave as self-renewing progenitors, some of whose progeny remain at the tips while others populate the growing UB trunks. In Ret or Etv4 MADM clones, the wild-type cells generated at a UB tip are much more likely to remain at, or move to, the new tips during branching and elongation, while their Ret-/- or Etv4-/- sister cells tend to lag behind and contribute only to the trunks. By tracking successive mitoses in a cell lineage, we find that Ret signaling has little effect on proliferation, in contrast to its effects on cell movement. Our results show that Ret/Etv4 signaling promotes directed cell movements in the ureteric bud tips, and suggest a model in which these cell movements mediate branching morphogenesis.


Kidney/embryology , Morphogenesis , Proto-Oncogene Proteins c-ets/physiology , Proto-Oncogene Proteins c-ret/physiology , Stem Cells/physiology , Animals , Cell Movement , Female , Male , Mice , Organ Culture Techniques
15.
Crit Rev Clin Lab Sci ; 53(4): 217-27, 2016 08.
Article En | MEDLINE | ID: mdl-26678667

Thyroid cancer is the most common endocrine malignancy and accounts for nearly 1% of all of human cancer. Thyroid cancer has four main histological types: papillary, follicular, medullary, and anaplastic. Papillary, follicular, and anaplastic thyroid carcinomas are derived from follicular thyroid cells, whereas medullary thyroid carcinoma (MTC) originates from the neural crest parafollicular cells or C-cells of the thyroid gland. MTC represents a neuroendocrine tumor and differs considerably from differentiated thyroid carcinoma. MTC is one of the aggressive types of thyroid cancer, which represents 3-10% of all thyroid cancers. It occurs in hereditary (25%) and sporadic (75%) forms. The hereditary form of MTC has an autosomal dominant mode of inheritance. According to the present classification, hereditary MTC is classified as a multiple endocrine neoplasi type 2 A & B (MEN2A & MEN2B) and familial MTC (FMTC). The RET proto-oncogene is located on chromosome 10q11.21. It is composed of 21 exons and encodes a transmembrane receptor tyrosine kinase. RET regulates a complex network of signal transduction pathways during development, survival, proliferation, differentiation, and migration of the enteric nervous system progenitor cells. Gain of function mutations in RET have been well demonstrated in MTC development. Variants of MTC result from different RET mutations, and they have a good genotype-phenotype correlation. Various MTC related mutations have been reported in different exons of the RET gene. We proposed that RET genetic mutations may be different in distinct populations. Therefore, the aim of this study was to find a geographical pattern of RET mutations in different populations.


Carcinoma, Neuroendocrine , Mutation , Proto-Oncogene Proteins c-ret , Thyroid Neoplasms , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/physiopathology , Humans , Mutation/genetics , Mutation/physiology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Proto-Oncogene Proteins c-ret/physiology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/physiopathology
16.
J Am Soc Nephrol ; 27(2): 417-27, 2016 Feb.
Article En | MEDLINE | ID: mdl-26047795

Hypoplastic and/or cystic kidneys have been found in both LDL receptor-related protein 6 (Lrp6)- and ß-catenin-mutant mouse embryos, and these proteins are key molecules for Wnt signaling. However, the underlying mechanisms of Lrp6/ß-catenin signaling in renal development and cystic formation remain poorly understood. In this study, we found evidence that diminished cell proliferation and increased apoptosis occur before cystic dysplasia in the renal primordia of Lrp6-deficient mouse embryos. The expression of Ret proto-oncogene (Ret), a critical receptor for the growth factor glial cell line-derived neurotrophic factor (GDNF), which is required for early nephrogenesis, was dramatically diminished in the mutant renal primordia. The activities of other representative nephrogenic genes, including Lim1, Pax2, Pax8, GDNF, and Wnt11, were subsequently diminished in the mutant renal primordia. Molecular biology experiments demonstrated that Ret is a novel transcriptional target of Wnt/ß-catenin signaling. Wnt agonist lithium promoted Ret expression in vitro and in vivo. Furthermore, Lrp6-knockdown or lithium treatment in vitro led to downregulation or upregulation, respectively, of the phosphorylated mitogen-activated protein kinases 1 and 3, which act downstream of GDNF/Ret signaling. Mice with single and double mutations of Lrp6 and Ret were perinatal lethal and demonstrated gene dosage-dependent effects on the severity of renal hypoplasia during embryogenesis. Taken together, these results suggest that Lrp6-mediated Wnt/ß-catenin signaling modulates or interacts with a signaling network consisting of Ret cascades and related nephrogenic factors for renal development, and the disruption of these genes or signaling activities may cause a spectrum of hypoplastic and cystic kidney disorders.


Kidney/growth & development , Low Density Lipoprotein Receptor-Related Protein-6/physiology , Multicystic Dysplastic Kidney/etiology , Proto-Oncogene Proteins c-ret/physiology , Signal Transduction , Animals , Mice , Mice, Knockout , Multicystic Dysplastic Kidney/genetics
17.
J Smooth Muscle Res ; 51: 82-94, 2015.
Article En | MEDLINE | ID: mdl-26658112

Two-photon microscopy (2PM) can enable high-resolution deep imaging of thick tissue by exciting a fluorescent dye and protein at anastomotic sites in the mouse small intestine in vivo. We performed gut surgery and transplanted neural stem cells (NSC) from the embryonic central nervous system after marking them with the fluorescent cell linker, PKH26. We found that neurons differentiated from transplanted NSC (PKH [+]) and newborn enteric neurons differentiated from mobilized (host) NSC (YFP [+]) could be localized within the granulation tissue of anastomoses. A 5-HT4-receptor agonist, mosapride citrate (MOS), significantly increased the number of PKH (+) and YFP (+) neurons by 2.5-fold (P<0.005). The distribution patterns of PKH (+) neurons were similar to those of YFP (+) neurons. On the other hand, the 5-HT4-receptor antagonist, SB-207266 abolished these effects of MOS. These results indicate that neurogenesis from transplanted NSC is facilitated by activation of 5-HT4-receptors. Thus, a combination of drug administration and cell transplantation could be more beneficial than exclusive cell transplantation in treating Hirschsprung's disease and related disorders including post rectal cancer surgery. The underlying mechanisms for its action were explored using immunohistochemistry of the longitudinal mouse ileum and rat rectal preparations including an anastomosis. MOS significantly increased the number of new neurons, but not when co-administered with either of a protein tyrosine kinase receptor, c-RET two inhibitors. The c-RET signaling pathway contributes to enteric neurogenesis facilitated by MOS. In the future, we would perform functional studies of new neurons over the thick granulation tissue at anastomoses, using in vivo imaging with 2PM and double transgenic mice expressing a calcium indicator such as GCaMP6 and channelrhodopsin.


Anastomosis, Surgical , Enteric Nervous System/physiology , Granulation Tissue/innervation , Intestine, Small/innervation , Neural Stem Cells/cytology , Neurogenesis/genetics , Neurons/physiology , Receptors, Serotonin, 5-HT4/physiology , Animals , Benzamides/pharmacology , Cell Differentiation/genetics , Guinea Pigs , Hirschsprung Disease/therapy , Humans , Intestine, Small/cytology , Mice , Morpholines/pharmacology , Neural Stem Cells/transplantation , Neurogenesis/drug effects , Proto-Oncogene Proteins c-ret/physiology , Serotonin 5-HT4 Receptor Agonists/pharmacology , Signal Transduction/physiology
18.
FEBS Lett ; 589(24 Pt A): 3760-72, 2015 Dec 21.
Article En | MEDLINE | ID: mdl-26555190

Glial cell line-derived neurotrophic factor (GDNF) and its canonical receptor Ret can signal together or independently to fulfill many important functions in the midbrain dopaminergic (DA) system. While Ret signaling clearly impacts on the development, maintenance and regeneration of the mesostriatal DA system, the physiological functions of GDNF for the DA system are still unclear. Nevertheless, GDNF is still considered to be an excellent candidate to protect and/or regenerate the mesostriatal DA system in Parkinson disease (PD). Clinical trials with GDNF on PD patients are, however, so far inconclusive. Here, we review the current knowledge of GDNF and Ret signaling and function in the midbrain DA system, and their crosstalk with proteins and signaling pathways associated with PD.


Dopaminergic Neurons/physiology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Parkinson Disease/metabolism , Proto-Oncogene Proteins c-ret/physiology , Animals , Dopamine/physiology , Humans , Mesencephalon/metabolism , Mesencephalon/pathology , Parkinson Disease/pathology , Synaptic Transmission
19.
J Clin Invest ; 125(5): 1873-85, 2015 May.
Article En | MEDLINE | ID: mdl-25822020

Parkin and the glial cell line-derived neurotrophic factor (GDNF) receptor RET have both been independently linked to the dopaminergic neuron degeneration that underlies Parkinson's disease (PD). In the present study, we demonstrate that there is genetic crosstalk between parkin and the receptor tyrosine kinase RET in two different mouse models of PD. Mice lacking both parkin and RET exhibited accelerated dopaminergic cell and axonal loss compared with parkin-deficient animals, which showed none, and RET-deficient mice, in which we found moderate degeneration. Transgenic expression of parkin protected the dopaminergic systems of aged RET-deficient mice. Downregulation of either parkin or RET in neuronal cells impaired mitochondrial function and morphology. Parkin expression restored mitochondrial function in GDNF/RET-deficient cells, while GDNF stimulation rescued mitochondrial defects in parkin-deficient cells. In both cases, improved mitochondrial function was the result of activation of the prosurvival NF-κB pathway, which was mediated by RET through the phosphoinositide-3-kinase (PI3K) pathway. Taken together, these observations indicate that parkin and the RET signaling cascade converge to control mitochondrial integrity and thereby properly maintain substantia nigra pars compacta dopaminergic neurons and their innervation in the striatum. The demonstration of crosstalk between parkin and RET highlights the interplay in the protein network that is altered in PD and suggests potential therapeutic targets and strategies to treat PD.


Dopaminergic Neurons/pathology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Nerve Degeneration/pathology , Parkinsonian Disorders/genetics , Proto-Oncogene Proteins c-ret/physiology , Ubiquitin-Protein Ligases/physiology , Adenosine Triphosphate/biosynthesis , Animals , Anxiety/genetics , Cell Line , Cell Size , Disease Progression , Exploratory Behavior , Glial Cell Line-Derived Neurotrophic Factor/deficiency , Glial Cell Line-Derived Neurotrophic Factor/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/pathology , NF-kappa B/physiology , Parkinsonian Disorders/pathology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-ret/deficiency , Proto-Oncogene Proteins c-ret/genetics , Recombinant Fusion Proteins/metabolism , Rotarod Performance Test , Signal Transduction , Substantia Nigra/pathology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
20.
J Clin Invest ; 125(3): 899-907, 2015 Mar 02.
Article En | MEDLINE | ID: mdl-25664848

The enteric nervous system (ENS) is sometimes called the "second brain" because of the diversity of neuronal cell types and complex, integrated circuits that permit the ENS to autonomously regulate many processes in the bowel. Mechanisms supporting ENS development are intricate, with numerous proteins, small molecules, and nutrients that affect ENS morphogenesis and mature function. Damage to the ENS or developmental defects cause vomiting, abdominal pain, constipation, growth failure, and early death. Here, we review molecular mechanisms and cellular processes that govern ENS development, identify areas in which more investigation is needed, and discuss the clinical implications of new basic research.


Colon/innervation , Enteric Nervous System/embryology , Animals , Body Patterning , Cell Movement , Cell Proliferation , Enteric Nervous System/cytology , Enteric Nervous System/physiology , Humans , Neural Crest/cytology , Proto-Oncogene Proteins c-ret/physiology , Retinoids/physiology , Signal Transduction
...