Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Gut ; 71(8): 1656-1668, 2022 08.
Article in English | MEDLINE | ID: mdl-34588223

ABSTRACT

OBJECTIVE: Hepatocellular carcinoma (HCC) has high intratumoral heterogeneity, which contributes to therapeutic resistance and tumour recurrence. We previously identified Prominin-1 (PROM1)/CD133 as an important liver cancer stem cell (CSC) marker in human HCC. The aim of this study was to investigate the heterogeneity and properties of Prom1+ cells in HCC in intact mouse models. DESIGN: We established two mouse models representing chronic fibrotic HCC and rapid steatosis-related HCC. We performed lineage tracing post-HCC induction using Prom1C-L/+; Rosa26tdTomato/+ mice, and targeted depletion using Prom1C-L/+; Rosa26DTA/+ mice. Single-cell RNA sequencing (scRNA-seq) was carried out to analyse the transcriptomic profile of traced Prom1+ cells. RESULTS: Prom1 in HCC tumours marks proliferative tumour-propagating cells with CSC-like properties. Lineage tracing demonstrated that these cells display clonal expansion in situ in primary tumours. Labelled Prom1+ cells exhibit increasing tumourigenicity in 3D culture and allotransplantation, as well as potential to form cancers of differential lineages on transplantation. Depletion of Prom1+ cells impedes tumour growth and reduces malignant cancer hallmarks in both HCC models. scRNA-seq analysis highlighted the heterogeneity of Prom1+ HCC cells, which follow a trajectory to the dedifferentiated status with high proliferation and stem cells traits. Conserved gene signature of Prom1 linage predicts poor prognosis in human HCC. The activated oxidant detoxification underlies the protective mechanism of dedifferentiated transition and lineage propagation. CONCLUSION: Our study combines in vivo lineage tracing and scRNA-seq to reveal the heterogeneity and dynamics of Prom1+ HCC cells, providing insights into the mechanistic role of malignant CSC-like cells in HCC progression.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , AC133 Antigen/genetics , AC133 Antigen/therapeutic use , Animals , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Mice , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Single-Cell Analysis
2.
J Hepatol ; 69(4): 826-839, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29885413

ABSTRACT

BACKGROUND & AIMS: Advanced hepatocellular carcinoma (HCC) is a lethal malignancy with limited treatment options. Sorafenib is the only FDA-approved first-line targeted drug for advanced HCC, but its effect on patient survival is limited. Further, patients ultimately present with disease progression. A better understanding of the causes of sorafenib resistance, enhancing the efficacy of sorafenib and finding a reliable predictive biomarker are crucial to achieve efficient control of HCC. METHODS: The functional effects of ANXA3 in conferring sorafenib resistance to HCC cells were analyzed in apoptotic and tumorigenicity assays. The role of ANXA3/PKCδ-mediated p38 signaling, and subsequently altered autophagic and apoptotic events, was assessed by immunoprecipitation, immunoblotting, immunofluorescence and transmission electron microscopy assays. The prognostic value of ANXA3 in predicting response to sorafenib was evaluated by immunohistochemistry. The therapeutic value of targeting ANXA3 to combat HCC with anti-ANXA3 monoclonal antibody alone or in combination with sorafenib/regorafenib was investigated ex vivo and in vivo. RESULTS: ANXA3 conferred HCC cells with resistance to sorafenib. ANXA3 was found enriched in sorafenib-resistant HCC cells and patient-derived xenografts. Mechanistically, overexpression of ANXA3 in sorafenib-resistant HCC cells suppressed PKCδ/p38 associated apoptosis and activated autophagy for cell survival. Clinically, ANXA3 expression correlated positively with the autophagic marker LC3B in HCC and was associated with a worse overall survival in patients who went on to receive sorafenib treatment. Anti-ANXA3 monoclonal antibody therapy combined with sorafenib/regorafenib impaired tumor growth in vivo and significantly increased survival. CONCLUSION: Anti-ANXA3 therapy in combination with sorafenib/regorafenib represents a novel therapeutic strategy for HCC treatment. ANXA3 represents a useful predictive biomarker to stratify patients with HCC for sorafenib treatment. LAY SUMMARY: This study represents the most extensive pre-clinical characterization of anti-ANXA3 monoclonal antibodies for the treatment of hepatocellular carcinoma to date. These results support the clinical trial development of anti-ANXA3 antibodies in combination with sorafenib/regorafenib. Further studies will optimize patient target selection and identify the best treatment combinations.


Subject(s)
Annexin A3/antagonists & inhibitors , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Phenylurea Compounds/therapeutic use , Pyridines/therapeutic use , Sorafenib/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Drug Resistance, Neoplasm , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/physiology
3.
Genome Biol ; 18(1): 48, 2017 03 08.
Article in English | MEDLINE | ID: mdl-28274275

ABSTRACT

BACKGROUND: Hirschsprung disease (HSCR), which is congenital obstruction of the bowel, results from a failure of enteric nervous system (ENS) progenitors to migrate, proliferate, differentiate, or survive within the distal intestine. Previous studies that have searched for genes underlying HSCR have focused on ENS-related pathways and genes not fitting the current knowledge have thus often been ignored. We identify and validate novel HSCR genes using whole exome sequencing (WES), burden tests, in silico prediction, unbiased in vivo analyses of the mutated genes in zebrafish, and expression analyses in zebrafish, mouse, and human. RESULTS: We performed de novo mutation (DNM) screening on 24 HSCR trios. We identify 28 DNMs in 21 different genes. Eight of the DNMs we identified occur in RET, the main HSCR gene, and the remaining 20 DNMs reside in genes not reported in the ENS. Knockdown of all 12 genes with missense or loss-of-function DNMs showed that the orthologs of four genes (DENND3, NCLN, NUP98, and TBATA) are indispensable for ENS development in zebrafish, and these results were confirmed by CRISPR knockout. These genes are also expressed in human and mouse gut and/or ENS progenitors. Importantly, the encoded proteins are linked to neuronal processes shared by the central nervous system and the ENS. CONCLUSIONS: Our data open new fields of investigation into HSCR pathology and provide novel insights into the development of the ENS. Moreover, the study demonstrates that functional analyses of genes carrying DNMs are warranted to delineate the full genetic architecture of rare complex diseases.


Subject(s)
Exome , Genetic Predisposition to Disease , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Hirschsprung Disease/genetics , Alleles , Animals , Case-Control Studies , Computational Biology/methods , DNA Mutational Analysis , Disease Models, Animal , Gene Knockout Techniques , Genotype , Humans , Mutation , Phenotype , Zebrafish
4.
Cell Death Differ ; 24(1): 38-48, 2017 01.
Article in English | MEDLINE | ID: mdl-27636107

ABSTRACT

The mechanisms of how signaling pathways are coordinated and integrated for the maintenance of the self-renewal of human embryonic stem cells (hESCs) and the acquisition of pluripotency in reprogramming are still only partly understood. CDK1 is a key regulator of mitosis. Recently, CDK1 has been shown to be involved in regulating self-renewal of stem cells, even though the mechanistic role of how CDK1 regulates pluripotency is unknown. In this report, we aim to understand how CDK1 can control pluripotency by reducing CDK1 activity to a level that has no effect on cell cycle progression. We demonstrated that high levels of CDK1 is associated with the pluripotency stage of hESCs; and decreased CDK1 activity to a level without perturbing the cell cycle is sufficient to induce differentiation. CDK1 specifically targets the phosphorylation of PDK1 and consequently the activity of PI3K/Akt and its effectors ERK and GSK3ß. Evidence of the reversion of inactive CDK1-mediated differentiation by the inhibition of Akt signaling effectors suggests that the CDK1-PDK1-PI3K/Akt kinase cascade is a functional signaling pathway for the pluripotency of hESCs. Moreover, cyclin B1-CDK1 complexes promote somatic reprogramming efficiency, probably by regulating the maturation of induced pluripotent stem cells (iPSCs), as cyclin B1 stimulates a higher cellular level of LIN28A, suggesting that monitoring iPSC factors could be a new path for the enhancement of reprogramming efficiency. Together, we demonstrate an essential role for the CDK1-PDK1-PI3K/Akt kinase signaling pathway in the regulation of self-renewal, differentiation, and somatic reprogramming, which provides a novel kinase cascade mechanism for pluripotency control and acquisition.


Subject(s)
CDC2 Protein Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/genetics , Cell Differentiation/drug effects , Cell Self Renewal , Cellular Reprogramming , Down-Regulation/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Human Embryonic Stem Cells , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Nanog Homeobox Protein/antagonists & inhibitors , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/antagonists & inhibitors , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Quinolines/pharmacology , RNA Interference , Signal Transduction/drug effects , Thiazoles/pharmacology , Tretinoin/pharmacology
5.
Birth Defects Res A Clin Mol Teratol ; 94(1): 47-51, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22131258

ABSTRACT

BACKGROUND: Hirschsprung disease (HSCR) is a congenital disease characterized by the absence of ganglion cells in various length of distal digestive tract. The rearranged during transfection gene (RET) is considered the major gene in HSCR. Although an increasing number of HSCR-associated RET coding sequence (CDS) mutations have been identified in recent years, not many have been investigated for functional consequence on the RET protein. METHODS AND RESULTS: We examined the functional implications of the de novo RET-CDS mutations V145G, Y483X, V636fsX1, and F961L that we first identified in sporadic Chinese patients with HSCR. The V145G disrupted RET glycosylation and F961L RET phosphorylation. Presumably, the truncation mutations would affect the translocation or the anchoring of the RET protein onto the cellular membrane. CONCLUSION: The study of RET-CDS mutations that appear de novo is essential not only for understanding the mechanistic of the disease but also for penetrance and recurrence risk estimations, being the ultimate goal for the improvement in disease management and counseling.


Subject(s)
Asian People/genetics , Hirschsprung Disease/genetics , Mutation , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Cell Line, Tumor , Female , Humans , Male , Penetrance
6.
J Pediatr Surg ; 44(10): 1904-12, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19853745

ABSTRACT

BACKGROUND: The rearranged during transfection (RET) gene encodes a single-pass receptor whose proper expression and function are essential for the development of enteric nervous system. Mutations in RET regulatory regions are also associated with Hirschsprung disease (HSCR) (aganglionosis of the colon). We previously showed that 2 polymorphisms in RET promoter are associated with the increased risk of HSCR. These single nucleotide polymorphisms overlap with the NK2 homeobox 1 (Nkx2-1) binding motif interrupting the physical interaction of NKX2-1 with the RET promoter and result in reduced RET transcription. In this study, we further delineated Nkx2-1-mediated RET Transcription. METHODS AND RESULTS: First, we demonstrated that PHOX2B, like SOX10 and NKX2-1, is expressed in the mature enteric ganglions of human gut by immunohistochemistry. Second, subsequent dual-luciferase-reporter studies indicated that Nkx2-1 indeed works coordinately with Phox2b and Sox10, but not Pax3, to mediate RET transcription. In addition, identification of Phox2b responsive region in RET promoter further provides solid evidence of the potential functional interaction between Phox2b and RET. CONCLUSION: In sum, Phox2b and Sox10 act together with Nkx2.1 to modify RET signaling and this interaction may also contribute to HSCR susceptibility.


Subject(s)
Hirschsprung Disease/genetics , Homeodomain Proteins/genetics , Proto-Oncogene Proteins c-ret/genetics , Transcription Factors/genetics , Animals , Cell Line, Tumor/metabolism , Enteric Nervous System/metabolism , Epistasis, Genetic/genetics , Gene Expression Regulation/genetics , Genetic Predisposition to Disease/genetics , Genotype , Homeodomain Proteins/metabolism , Humans , Mice , Mutation/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Proto-Oncogene Proteins c-ret/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Signal Transduction/genetics , Thyroid Nuclear Factor 1 , Transcription Factors/metabolism , Transcriptional Activation/genetics , Transfection/statistics & numerical data
7.
J Natl Cancer Inst ; 101(3): 162-75, 2009 Feb 04.
Article in English | MEDLINE | ID: mdl-19176457

ABSTRACT

BACKGROUND: The genetic factors that determine the risk of papillary thyroid carcinoma (PTC) among patients with multinodular goiter (MNG) remain undefined. Because thyroid transcription factor-1 (TTF-1) is important to thyroid development, we evaluated whether the gene that encodes it, TITF-1/NKX2.1, is a genetic determinant of MNG/PTC predisposition. METHODS: Twenty unrelated PTC patients with a history of MNG (MNG/PTC), 284 PTC patients without a history of MNG (PTC), and 349 healthy control subjects were screened for germline mutation(s) in TITF-1/NKX2.1 by sequencing of amplified DNA from blood. The effects of the mutation on the growth and differentiation of thyroid cells were demonstrated by ectopic expression of wild-type (WT) and mutant proteins in PCCL3 normal rat thyroid cells, followed by tests of cell proliferation, activation of cell growth pathways, and transcription of TTF-1 target genes. All statistical tests were two-sided. RESULTS: A missense mutation (1016C>T) was identified in TITF-1/NKX2.1 that led to a mutant TTF-1 protein (A339V) in four of the 20 MNG/PTC patients (20%). These patients developed substantially more advanced tumors than MNG/PTC or PTC patients without the mutation (P = .022, Fisher exact test). Notably, this germline mutation was dominantly inherited in two families, with some members bearing the mutation affected with MNG, associated with either PTC or colon cancer. The mutation encoding the A339V substitution was not found among the 349 healthy control subjects nor among the 284 PTC patients who had no history of MNG. Overexpression of A339V TTF-1 in PCCL3 cells, as compared with overexpression of WT TTF-1, was associated with increased cell proliferation including thyrotropin-independent growth (average A339V proliferation rate = 134.27%, WT rate = 104.43%, difference = 34.3%, 95% confidence interval = 12.0% to 47.7%, P = .010), enhanced STAT3 activation, and impaired transcription of the thyroid-specific genes Tg, TSH-R, and Pax-8. CONCLUSION: This is the first germline mutation identified in MNG/PTC patients. It could contribute to predisposition for MNG and/or PTC and to the pathogenesis of PTC.


Subject(s)
Carcinoma, Papillary/genetics , Cell Transformation, Neoplastic/genetics , Germ-Line Mutation , Goiter, Nodular/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Thyroid Neoplasms/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Alanine , Animals , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Case-Control Studies , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Child , Electrophoretic Mobility Shift Assay , Female , Gene Expression Regulation, Neoplastic , Goiter, Nodular/metabolism , Goiter, Nodular/pathology , Humans , Immunoblotting , Luciferases , Male , Middle Aged , Neoplasm Staging , Rats , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Thyroid Gland/cytology , Thyroid Gland/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Thyroid Nuclear Factor 1 , Transcription, Genetic , Up-Regulation , Valine , Young Adult
8.
Int J Biochem Cell Biol ; 40(9): 1679-84, 2008.
Article in English | MEDLINE | ID: mdl-18440852

ABSTRACT

Prokineticin signaling comprises two secreted proteins (Prok-1 and Prok-2) and two cognate G-protein coupled receptors (PK-R1 and PK-R2) that are widely expressed in different tissues and of great versatility. Prokineticins were originally identified as the potent agents mediating gut motility in the digestive system, but were later shown to promote angiogenesis in steroidgenic glands, heart and reproductive organs. Prokineticins also modulate neurogenesis, circadian rhythms, nociception, haematopoiesis as well as immune response. Their diverse biological functions and functional complexity are exquisitely mediated by the distinct expression pattern and the multiple G-protein coupling of the receptors and ligands. Emerging evidence indicated that prokineticins are also associated with pathologies of the reproductive and nervous systems, myocardial infarction and tumorigenesis. The physiological and patho-physiological roles of prokineticin signaling are just beginning to be revealed and a better understanding of the system should lead to the development of useful therapies for various diseases.


Subject(s)
Signal Transduction , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism , Animals , Hirschsprung Disease/metabolism , Humans , Kallmann Syndrome/metabolism , Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism
9.
Biochim Biophys Acta ; 1783(3): 467-78, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18006159

ABSTRACT

Enteric neural crest cells (NCC) are multipotent progenitors which give rise to neurons and glia of the enteric nervous system (ENS) during fetal development. Glial cell line-derived neurotrophic factor (GDNF)/RET receptor tyrosine kinase (Ret) signaling is indispensable for their survival, migration and differentiation. Using microarray analysis and isolated NCCs, we found that 45 genes were differentially expressed after GDNF treatment (16 h), 29 of them were up-regulated including 8 previously undescribed genes. Prokineticin receptor 1 (PK-R1), a receptor for Prokineticins (Prok), was identified in our screen and shown to be consistently up-regulated by GDNF in enteric NCCs. Further, PK-R1 was persistently expressed at a lower level in the enteric ganglions of the c-Ret deficient mice when compared to that of the wild-type littermates. Subsequent functional analysis showed that GDNF potentiated the proliferative and differentiation effects of Prok-1 by up-regulating PK-R1 expression in enteric NCCs. In addition, expression analysis and gene knock-down experiments indicated that Prok-1 and GDNF signalings shared some common downstream targets. More importantly, Prok-1 could induce both proliferation and expression of differentiation markers of c-Ret deficient NCCs, suggesting that Prok-1 may also provide a complementary pathway to GDNF signaling. Taken together, these findings provide evidence that Prok-1 crosstalks with GDNF/Ret signaling and probably provides an additional layer of signaling refinement to maintain proliferation and differentiation of enteric NCCs.


Subject(s)
Cell Differentiation , Cell Proliferation , Enteric Nervous System/embryology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Neural Crest/physiology , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/physiology , Animals , Cell Differentiation/genetics , Embryo, Mammalian , Extracellular Matrix Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glycoproteins/genetics , Mice , Mice, Transgenic , Models, Biological , Neural Crest/metabolism , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-ret/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism , Vasoactive Intestinal Peptide/genetics
10.
Clin Cancer Res ; 13(3): 868-75, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17289879

ABSTRACT

PURPOSE: Neuroblastoma is a common pediatric tumor that is derived from improperly differentiated neural crest cells (NCC). We recently revealed that endocrine gland-derived vascular endothelial growth factor/prokineticin-1 (EG-VEGF/Prok-1) is a key factor mediating the growth and differentiation of enteric NCCs during development. In this report, we further elucidate its role in neuroblastoma progression. EXPERIMENTAL DESIGN: We studied the expression and copy number of EG-VEGF/Prok-1 receptors (PK-R1 and PK-R2) in 26 neuroblastoma tumors by real-time reverse transcription-PCR and immunohistochemical analysis. Implication of EG-VEGF/Prok-1 signaling in neuroblastoma progression was further shown in a neuroblastoma cell line (SK-N-SH). RESULTS: We found that all neuroblastoma samples from stages II to IV expressed both PK-R1 and PK-R2. Kruskall-Wallis signed rank tests revealed that the expression level of PK-R1 transcript is associated with the stages and metastasis of the neuroblastoma (P<0.05), and PK-R2 is persistently higher in advanced-stage neuroblastoma samples. About 38% of the neuroblastoma tumors (10:26) possessed MYCN amplification, whereas no PK-R1 and PK-R2 amplifications were detected, suggesting that the overexpression of the receptors was not due to gene amplification. Subsequent functional studies showed that EG-VEGF/Prok-1 activates the Akt pathway to induce the proliferation of neuroblastoma cells. Targeted down-regulation studies revealed that EG-VEGF/Prok-1-mediated proliferation requires the presence of these two receptors, and that PK-R2 is essential for inhibiting apoptosis. In vitro migration and invasion assays also indicated that EG-VEGF/Prok-1 significantly enhances the cell migration/invasion of SK-N-SH. CONCLUSIONS: Our study has shown for the first time that aberrant EG-VEGF/Prok-1 signaling favors neuroblastoma progression and could be a potential target for future neuroblastoma treatment.


Subject(s)
Endocrine Glands/metabolism , Gene Expression Regulation, Neoplastic , Neuroblastoma/metabolism , Signal Transduction , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism , Cell Movement , Cell Proliferation , Child , Child, Preschool , Disease Progression , Female , Humans , Infant , Male , Neoplasm Invasiveness , Neuroblastoma/pathology , Reverse Transcriptase Polymerase Chain Reaction
11.
Biochim Biophys Acta ; 1773(4): 536-45, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17324478

ABSTRACT

Prokineticins (Prok-1 and Prok-2) belong to a newly identified AVIT protein family. They are involved in variety of activities in various tissues, including smooth muscle contraction of the gastrointestinal tract and promoting proliferation of endothelial cells derived from adrenal gland. Importantly, they also act as the survival factors to modulate growth and survival of neurons and hematopoietic stem cells. In this study we demonstrated that Prok-1 (but not Prok-2) protein is expressed in the mucosa and mesenchyme of the mouse embryonic gut during enteric nervous system development. Its receptor, PK-R1 is expressed in the enteric neural crest cells (NCCs). To elucidate the physiological role(s) of Prok-1 in NCCs, we isolated the NCCs from the mouse embryonic gut (E11.5) and cultured them in the form of neurospheres. In an in vitro NCC culture, Prok-1 was able to activate both Akt and MAPK pathways and induce the proliferation and differentiation (but not migration) of NCCs via PK-R1. Knock-down of PK-R1 using siRNA resulted in a complete abolishment of Prok-1 induced proliferation. Taken together, it is the first report demonstrating that Prok-1 acts as a gut mucosa/mesenchyme-derived factor and maintains proliferation and differentiation of enteric NCCs.


Subject(s)
Cell Differentiation , Gastrointestinal Tract/cytology , Neural Crest/cytology , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism , Animals , Cell Movement , Cell Proliferation , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Gastrointestinal Tract/embryology , Gastrointestinal Tract/metabolism , Gene Expression Regulation , Humans , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Inbred ICR , Mitogen-Activated Protein Kinases/metabolism , Neural Crest/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics
12.
Endocrinology ; 147(1): 88-95, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16210375

ABSTRACT

Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) is a newly identified angiogenic and permeability-enhancing factor, predominantly expressed in steroidogenic tissues. Recently, we found that EG-VEGF is also expressed in the normal peri-implantation endometrial samples from patients of reproductive ages (80%). Immunohistochemistry analysis showed that EG-VEGF is predominantly expressed in the glandular epithelial cells and its expression is dynamic during the menstrual cycle with a peak expression at the mid-luteal phase. We also found that EG-VEGF transcripts are up-regulated in all the peri-implantation endometrial samples from the patients after the ovulating dose of human chorionic gonadotropin in gonadotropin-stimulated cycles and patients receiving hormone replacement therapy. In in vitro endometrial cell culture, EG-VEGF mRNA was detected in endometrial cells only in the presence of steroids, suggesting that EG-VEGF expression is highly dependent on the steroid hormones. Subsequent expression analyses on the EG-VEGF receptors showed that hPK-R1 and hPK-R2 are differentially expressed in human endometrium, but show no significant correlation with the hormonal treatments. On the other hand, EG-VEGF transcript was rarely detected in the endometrial samples from the postmenopausal patients and patients with endometrial carcinoma. It may imply that EG-VEGF may only play a role in vascular function of peri-implantation endometrium, but is unlikely to be associated with the etiology of endometrial cancer development.


Subject(s)
Endometrial Neoplasms/genetics , Endometrium/physiology , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics , Cells, Cultured , Embryo Implantation , Female , Humans , In Situ Hybridization , Menstrual Cycle , Pregnancy , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Vascular Endothelial Growth Factor A/genetics
13.
Oncogene ; 22(5): 734-9, 2003 Feb 06.
Article in English | MEDLINE | ID: mdl-12569365

ABSTRACT

Cdc25B is a dual-specific phosphatase that mediates cell cycle progression by activating the cyclin-dependent kinases. It has been shown to possess oncogenic potential. To elucidate its potential contribution to human prostate cancer development, the expression profile of Cdc25B protein in human patients was analysed by immunohistocytochemistry. Cdc25B is frequently overexpressed in human prostate cancer tissues (29 of 30; 97%). In addition, the overexpression is more profound in the tumors of high combined Gleason scores and in late stages. Subsequently, we demonstrated that Cdc25B acts as a coactivator for AR in a hormone-dependent manner in the prostate cancer cell line, LNCaP. This coactivator function, surprisingly, is independent of its cell cycle functions. Cdc25B, on the other hand, directly interacts with AR as evidenced in GST-pull down and mammalian two-hybrid assays. In addition, it is also able to enhance AR-mediated transcription in synergy with other coactivators, including CREB-binding protein (CBP) and p300/CBP associated factor. Therefore, upregulation of Cdc25B in human prostate cancer and its interplay with AR may contribute to prostate cancer development.


Subject(s)
Cell Cycle Proteins/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/metabolism , cdc25 Phosphatases/genetics , Adult , Aged , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Humans , Immunohistochemistry , Male , Middle Aged , Mutation , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Transcription, Genetic , Up-Regulation , cdc25 Phosphatases/biosynthesis , cdc25 Phosphatases/metabolism
14.
Endocrinology ; 144(2): 518-27, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12538612

ABSTRACT

Differential usage of several transcription start sites in the human GnRH receptor gene was evident in human brain and pituitary. To locate the promoter responsible for a cluster of the 3' CAP sites from -635 to -578 (relative to ATG) found in the pituitary, a proximal promoter element was identified at -677/-558 by 5' and 3' deletion mutant analysis. The promoter element drove a 13.1 +/- 0.6-fold increase in reporter gene activity in an orientation-dependent manner in the mouse gonadotrope-derived alphaT3-1 cells. Within the core promoter element, two functional AT-rich Inr motifs, interacting with the same protein factor with different affinities, were identified. By Southwestern blot analysis and competitive gel mobility shift assays, multiple nuclear factors (36-150 kDa) were found to interact specifically with the core promoter element. Interestingly, these nuclear proteins also interacted with a previously identified distal promoter of the human GnRH receptor gene. Taken together, our studies suggested that these two promoters share common protein factors to regulate transcription initiations at two different regions. Additional mechanisms are needed to modulate the efficiencies of individual promoters for developmental and/or tissue-specific regulations.


Subject(s)
Promoter Regions, Genetic/genetics , Receptors, LHRH/genetics , Animals , Blotting, Southwestern , Cell Line , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Erythroid-Specific DNA-Binding Factors , Gene Deletion , Humans , Mice , Mutagenesis, Site-Directed , Nuclear Proteins/metabolism , Transcription Factor TFIID/metabolism , Transcription Factors/metabolism , Transcription Initiation Site , Transcription, Genetic/genetics
15.
Semin Cell Dev Biol ; 13(2): 143-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12240599

ABSTRACT

The mifepristone (Mfp)-inducible gene regulatory system is designed to allow control of the spatiotemporal expression of transgenes in vivo in a ligand-dependent manner. This regulatory system is composed of two components: (1) a chimeric transactivator protein that activates transgene transcription only in the presence of the progesterone antagonist Mfp, and (2) a target transgene placed in the context of a promoter which is responsive only to the Mfp-bound chimeric transactivator. Incorporation of the components of the Mfp-inducible gene regulatory system into transgenic mice has resulted in the establishment of several novel, Mfp-dependent models of disease. Similarly, adaptation of the Mfp-inducible system for use in gene knockout models has resulted in the development of new gene ablation technology which is both tissue-specific and Mfp-dependent. Additionally, the Mfp-inducible gene regulatory system has been used in animal experiments involving somatic gene therapy, where it has shown considerable promise in the regulation of both reporter and therapeutic gene expression. This review focuses on recent application of the Mfp-inducible system to transgenic models, gene knockout models, and somatic gene therapy experiments. In so doing, it demonstrates the considerable promise that future use of this system holds for better understanding and treatment of human disease.


Subject(s)
Gene Transfer Techniques , Genetic Techniques , Genetic Therapy/methods , Mifepristone/pharmacology , Animals , Disease Models, Animal , Hormone Antagonists/pharmacology , Ligands , Mice , Models, Genetic , Transgenes
16.
Proc Natl Acad Sci U S A ; 99(17): 11187-92, 2002 Aug 20.
Article in English | MEDLINE | ID: mdl-12169667

ABSTRACT

Fibroblast growth factor-3 (FGF-3) is a crucial developmental regulator. Aberrant activation of this gene by mouse mammary tumor virus insertion results in pregnancy-responsive mammary tumorigenesis. To characterize better FGF-3 function in postnatal mammary gland development and cancer initiation/progression, we used a mifepristone (RU486)-inducible regulatory system to express conditionally FGF-3 in the mammary epithelium of transgenic mice. Ectopic overexpression of FGF-3 in pubescent mammary glands elicited severe perturbations in early mammary gland development leading to mammary hyperplasia. Ductal elongation was retarded, multiple cysts persisted in the virgin ducts, and ductal epithelium was expanded and multilayered. The altered ductal architecture and the persistence of hyperplastic multilayered epithelium reflect a defect in growth regulation, which resulted from an imbalance between mitogenic and apoptotic signals. By altering the duration of RU486 treatment, we showed that the persistence of mitogenic signal elicited by FGF-3 is crucial for the initiation, progression, and maintenance of the hyperplastic characteristic of the mammary epithelium. The manifestations elicited by FGF-3 could be reversed by RU486 withdrawal. In addition, synergism between the stimulus from estrogen and FGF-3 mitogenic pathways was evident and likely contributes to the pregnancy-dependent tumorigenesis of FGF-3. Taken together, the mifepristone-inducible regulatory system provides a powerful means for understanding the diverse roles of FGF-3 and its interactions with hormones in mammary gland tumorigenesis.


Subject(s)
Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental/physiology , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Proto-Oncogene Proteins/genetics , Animals , Cell Adhesion Molecules/genetics , Disease Progression , Epithelial Cells/physiology , Female , Fibroblast Growth Factor 3 , Hyperplasia , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/pathology , Mice , Mice, Transgenic , Mifepristone/pharmacology , Ovariectomy , Sexual Maturation
SELECTION OF CITATIONS
SEARCH DETAIL
...