Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 76(21): 6351-6361, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27590741

ABSTRACT

The evolution of chemoresistance is a fundamental characteristic of cancer that ultimately hampers its clinical management. However, it may be possible to improve patient outcomes significantly by a better understanding of resistance mechanisms, which cancers rely upon during the evolution to an untreatable state. Here we report an essential role of the stem cell reprogramming factor, PBX1, in mediating chemoresistance in ovarian carcinomas. In the clinical setting, high levels of PBX1 expression correlated with shorter survival in post-chemotherapy ovarian cancer patients. In tumor cells with low endogenous levels of PBX1, its enforced expression promoted cancer stem cell-like phenotypes, including most notably an increase in resistance to platinum-based therapy used most commonly for treating this disease. Conversely, silencing PBX1 in platinum-resistant cells that overexpressed PBX1 sensitized them to platinum treatment and reduced their stem-like properties. An analysis of published genome-wide chromatin immunoprecipitation data indicated that PBX1 binds directly to promoters of genes involved in stem cell maintenance and the response to tissue injury. We confirmed direct regulation of one of these genes, STAT3, demonstrating that the PBX1 binding motif at its promoter acted to positively regulate STAT3 transcription. We further demonstrated that a STAT3/JAK2 inhibitor could potently sensitize platinum-resistant cells to carboplatin and suppress their growth in vivo Our findings offer a mechanistic rationale to target the PBX1/STAT3 axis to antagonize a key mechanism of chemoresistance in ovarian cancers and possibly other human cancers. Cancer Res; 76(21); 6351-61. ©2016 AACR.


Subject(s)
Cellular Reprogramming , DNA-Binding Proteins/physiology , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins/physiology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Janus Kinase 2/physiology , Ovarian Neoplasms/pathology , Pre-B-Cell Leukemia Transcription Factor 1 , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/physiology , Transcriptome
2.
Cell Rep ; 9(2): 674-87, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25373905

ABSTRACT

Sorting and degradation of receptors and associated signaling molecules maintain homeostasis of conserved signaling pathways during cell specification and tissue development. Yet, whether machineries that sort signaling proteins act preferentially on different receptors and ligands in different contexts remains mysterious. Here, we show that Vacuolar protein sorting 25, Vps25, a component of ESCRT-II (Endosomal Sorting Complex Required for Transport II), directs preferential endosome-mediated modulation of FGF signaling in limbs. By ENU-induced mutagenesis, we isolated a polydactylous mouse line carrying a hypomorphic mutation of Vps25 (Vps25(ENU)). Unlike Vps25-null embryos we generated, Vps25(ENU/ENU) mutants survive until late gestation. Their limbs display FGF signaling enhancement and consequent hyperactivation of the FGF-SHH feedback loop causing polydactyly, whereas WNT and BMP signaling remain unperturbed. Notably, Vps25(ENU/ENU) Mouse Embryonic Fibroblasts exhibit aberrant FGFR trafficking and degradation; however, SHH signaling is unperturbed. These studies establish that the ESCRT-II machinery selectively limits FGF signaling in vertebrate skeletal patterning.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/metabolism , Fibroblast Growth Factors/metabolism , Hedgehog Proteins/metabolism , Polydactyly/genetics , Signal Transduction , Vesicular Transport Proteins/genetics , Animals , Endosomal Sorting Complexes Required for Transport/genetics , Extremities/growth & development , Feedback, Physiological , Fibroblasts/metabolism , Mice , Mice, Inbred C57BL , Mutation , Polydactyly/metabolism , Vesicular Transport Proteins/metabolism
3.
J Reprod Immunol ; 90(1): 41-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21704384

ABSTRACT

The peroxisome proliferator-activated receptor-γ (PPARγ) is a member of the nuclear receptor superfamily that controls in a ligand-dependent manner the expression of a large array of genes involved in the control of energy homeostasis and in cell differentiation, proliferation, apoptosis, and the inflammatory process. Unexpectedly, genetic studies performed in mice established that PPARγ is essential for placental development. In the human placenta, PPARγ is specifically expressed in the trophoblast, both endocrine villous and invasive extravillous cytotrophoblasts (EVCT). Activation of PPARγ induces accumulation of lipids, villous trophoblast differentiation and inhibits trophoblast invasiveness. Oxidized LDLs that contain potential PPARγ ligands, but not native LDL, induce PPARγ transcriptional activity and inhibit trophoblast invasion in vitro. Recently, human cytomegalovirus (HCMV) was shown to activate trophoblastic PPARγ for its own replication and consequently inhibits invasiveness of infected cytotrophoblasts. Analysis of PPARγ target genes revealed trophoblastic factors described to control trophoblast invasiveness and surprisingly chorionic gonadotropin hormone (hCG), known to be mainly produced by the endocrine villous trophoblast. Analysis of hCG gene expression revealed opposite regulation by PPARγ in the two trophoblast subtypes. Finally, a hyperglycosylated form of hCG (hCG-H) only produced by invasive EVCT was shown to promote trophoblast invasion. Together, these data underscore the major role of PPARγ and its target genes, such as hCG, in the control of human trophoblast differentiation and invasion, and suggest that over-activation of this nuclear receptor following HCMV infection or by excess of ligands at the maternal-fetal interface could impair implantation and placentation and therefore embryonic development.


Subject(s)
Cell Differentiation , PPAR gamma/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism , Animals , Cells, Cultured , Chorionic Gonadotropin/biosynthesis , Cytomegalovirus/metabolism , Embryo Implantation , Female , Gene Expression Regulation, Developmental , Humans , Lipids/biosynthesis , Mice , Peroxisome Proliferator-Activated Receptors/metabolism , Placentation/physiology , Pregnancy
4.
Dev Dyn ; 240(5): 1173-89, 2011 May.
Article in English | MEDLINE | ID: mdl-21455939

ABSTRACT

Genes expressed in the somatopleuric mesoderm, the embryonic domain giving rise to the vertebrate pelvis, appear important for pelvic girdle formation. Among such genes, Pbx family members and Emx2 were found to genetically interact in hindlimb and pectoral girdle formation. Here, we generated compound mutant embryos carrying combinations of mutated alleles for Pbx1, Pbx2, and Pbx3, as well as Pbx1 and Emx2, to examine potential genetic interactions during pelvic development. Indeed, Pbx genes share overlapping functions and Pbx1 and Emx2 genetically interact in pelvic formation. We show that, in compound Pbx1;Pbx2 and Pbx1;Emx2 mutants, pelvic mesenchymal condensation is markedly perturbed, indicative of an upstream control by these homeoproteins. We establish that expression of Tbx15, Prrx1, and Pax1, among other genes involved in the specification and development of select pelvic structures, is altered in our compound mutants. Lastly, we identify potential Pbx1-Emx2-regulated enhancers for Tbx15, Prrx1, and Pax1, using bioinformatics analyses.


Subject(s)
Pelvis/embryology , Animals , Computational Biology , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization , Male , Mice , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Pre-B-Cell Leukemia Transcription Factor 1 , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Curr Med Chem ; 15(28): 3011-24, 2008.
Article in English | MEDLINE | ID: mdl-19075649

ABSTRACT

During pregnancy, the placenta ensures multiple functions, which are directly involved in the initiation, outcome of gestation and fetal growth. Human implantation involves a major invasion of the uterus wall and a complete remodeling of the uterine arteries by the extravillous cytotrophoblasts (EVCT) during the first trimester of pregnancy. Abnormality of these early steps of placental development leads to poor placentation, fetal growth defects and is very often associated with preeclampsia, a major and frequent complication of human pregnancy. Unexpectedly, genetic studies performed in mice established that the peroxisome proliferator-activated receptor-gamma (PPARgamma) is essential for placental development. In the human placenta, PPARgamma is specifically expressed in the villous cytotrophoblast (VCT) and the syncytiotrophoblast (ST) as well as in the EVCT along their invasive pathway. To study the mechanisms that control human trophoblast invasion during early placental development and to provide new insight in the understanding of preeclampsia, we have developed in vitro models of human invasive trophoblasts. We observed that activation of the ligand-activated nuclear receptor PPARgamma agonists inhibits the trophoblastic invasion process in a concentration-dependent manner. Analysis of PPARgamma-target genes revealed that placental growth hormone, the protease PAPP-A and the human chorionic gonadotropin hormone (hCG) might be involved in the PPARgamma-mediated effect in an autocrine manner. The presence of oxidized-LDLs at the maternofetal interface suggests that oxidized-LDLs from maternal sera might be a source of potential PPARgamma ligands for the trophoblasts. Indeed, oxidized-LDLs decrease trophoblast invasion in vitro and analysis of their content revealed that they contain potent PPARgamma agonists such as eicosanoids, but also oxysterols, which are ligands for another nuclear receptor, the liver X Receptor (LXR). LXRbeta was found to be expressed in trophoblast and LXR agonists shown to inhibit trophoblast invasion. Together, these data underscore a major role for PPARgamma in the control of human trophoblast invasion during early placental development and suggest that ligands such as oxidized-LDLs at the implantation site might contribute to the modulation of trophoblast invasion through activation of PPARgamma and LXRbeta, two nuclear receptors that modulate the human trophoblastic cell invasion process.


Subject(s)
Gene Expression Regulation, Developmental , PPAR gamma/physiology , Placentation , Cell Movement , Female , Humans , Immunohistochemistry , Ligands , Lipoproteins, LDL/chemistry , Models, Chemical , Oxygen/chemistry , PPAR gamma/metabolism , Pre-Eclampsia/diagnosis , Pregnancy , Trophoblasts/metabolism
6.
Retrovirology ; 5: 6, 2008 Jan 23.
Article in English | MEDLINE | ID: mdl-18215254

ABSTRACT

Human trophoblast expresses two fusogenic retroviral envelope proteins, the widely studied syncytin 1, encoded by HERV-W and the recently characterized syncytin 2 encoded by HERV-FRD. Here we studied syncytin 2 in normal and Trisomy 21-affected placenta associated with abnormal trophoblast differentiation. Syncytin 2 immunolocalization was restricted throughout normal pregnancy to some villous cytotrophoblastic cells (CT). During the second trimester of pregnancy, syncytin 2 was immunolocalized in some cuboidal CT in T21 placentas, whereas in normal placentas it was observed in flat CT, extending into their cytoplasmic processes. In vitro, CT isolated from normal placenta fuse and differentiate into syncytiotrophoblast. At the same time, syncytin 2 transcript levels decreased significantly with syncytiotrophoblast formation. In contrast, CT isolated from T21-affected placentas fused and differentiated poorly and no variation in syncytin 2 transcript levels was observed. Syncytin 2 expression illustrates the abnormal trophoblast differentiation observed in placenta of fetal T21-affected pregnancies.


Subject(s)
Down Syndrome/metabolism , Endogenous Retroviruses/metabolism , Placenta/metabolism , Pregnancy Proteins/metabolism , Cell Differentiation , Cells, Cultured , Female , Humans , Pregnancy , Pregnancy Trimester, First , Pregnancy Trimester, Second , Trophoblasts/cytology , Trophoblasts/metabolism
7.
J Reprod Immunol ; 77(2): 161-70, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17706792

ABSTRACT

Human implantation involves a major invasion of the uterine wall and complete remodelling of uterine arteries by extravillous cytotrophoblasts (EVCT). Abnormality in these early steps of placental development leads to poor placentation, fetal growth defects and is often associated with preeclampsia, a major and frequent complication of human pregnancy. To study the mechanisms that control trophoblast invasion during early placental development and provide new insight in the understanding of preeclampsia, we have developed in vitro models of human invasive trophoblasts. We have shown that activation of the ligand-activated nuclear receptor PPARgamma with synthetic (rosiglitazone) or natural (15deoxyPGJ(2)) agonists inhibits the trophoblastic invasion process. Analysis of PPARgamma-target genes revealed that placental growth hormone and the protease PAPP-A might be involved in the PPARgamma-mediated effect in an autocrine manner. We next investigated PPARgamma ligands at the materno-fetal interface and have shown that oxidized LDLs are present in EVCT in situ and decrease trophoblast invasion in vitro. Analysis of oxidized LDLs revealed that they contain potent PPARgamma agonists such as eicosanoids and also high levels of oxysterols, which are specific ligands for the liver X receptor (LXR). The isoform beta of LXR was found in EVCT in situ, and activation of LXRbeta with synthetic or natural ligands inhibits trophoblast invasion in vitro. Together, our data underscore a major role for PPARgamma and LXRbeta in the control of human trophoblast invasion and suggest that excess ligands such as oxidized LDLs at the implantation site might contribute to the development of preeclampsia.


Subject(s)
DNA-Binding Proteins/metabolism , Embryo Implantation , PPAR gamma/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism , Animals , DNA-Binding Proteins/agonists , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Developmental , Humans , Ligands , Lipoproteins, LDL/metabolism , Liver X Receptors , Orphan Nuclear Receptors , PPAR gamma/agonists , PPAR gamma/genetics , Placenta/cytology , Placenta/metabolism , Placental Hormones/metabolism , Pre-Eclampsia/etiology , Pre-Eclampsia/pathology , Pregnancy , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics
8.
Endocrinology ; 148(10): 5011-9, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17628005

ABSTRACT

A critical step in the establishment of human pregnancy is the invasion of the uterus wall by extravillous cytotrophoblasts (EVCTs) during the first trimester. It is well established that human chorionic gonadotropin hormone (hCG) is secreted by the endocrine syncytiotrophoblast (ST) into the maternal compartment. We recently reported that invasive EVCTs also produce hCG, suggesting an autocrine role in the modulation of trophoblast invasion. Here we analyzed the role of hCG secreted in vitro by primary cultures of invasive EVCT and noninvasive ST. We first demonstrated that LH/CG receptor was present in EVCTs in situ and in vitro as well as in an EVCT cell line (HIPEC65). We next showed that hCG secreted by EVCTs stimulated progesterone secretion by MA10 cells in a concentration-dependent manner. Incubation of HIPEC65 with EVCT supernatants induced a 10-fold increase in cell invasion, whereas ST supernatants had no effect. This stimulating effect was strongly decreased when hCG was depleted from EVCT supernatants containing a large amount of the hyperglycosylated form of hCG, which is almost undetectable in ST supernatants. Finally, we investigated the regulation of hCG expression by peroxisome proliferator-activated receptor (PPAR)-gamma, a nuclear receptor shown to inhibit trophoblast invasion. Activation of PPARgamma decreased alpha- and beta-subunit transcript levels and total hCG secretion in primary EVCTs. Our results offer the first evidence that hCG secreted by the invasive trophoblast, likely the hyperglycosylated form of hCG, but not by the syncytiotrophoblast, promotes trophoblast invasion and may be a PPARgamma target gene in trophoblast invasion process.


Subject(s)
Chorionic Gonadotropin/physiology , PPAR gamma/physiology , Trophoblasts/physiology , Uterus/physiology , Animals , Cell Line , Chorionic Gonadotropin/metabolism , Down-Regulation , Female , Humans , Immunoblotting , Immunohistochemistry , Mice , Pregnancy , Receptors, LH/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...