Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters

Publication year range
1.
Mem Inst Oswaldo Cruz ; 117: e210395, 2022.
Article in English | MEDLINE | ID: mdl-35239842

ABSTRACT

Transforming growth factor beta (TGF-ß) is deeply involved on the pathogenesis of Chagas disease. Our group has been investigating the participation of this pleiotropic cytokine in different aspects of Chagas disease over the last 20 years. Important observations have been made, such as: (i) the ability of Trypanosoma cruzi in activating latent TGF-ß; (ii) the potential involvement of TGF-ß pathway on T. cruzi invasion of host cells; (iii) association of TGF-ß with parasite intracellular replication; (iv) cardiac fibrosis development and maintenance; (v) disruption of Connexin-43 plaque structures and (vi) inflammation and immune response. In this perspective article we intend to discuss the advances of the potential use of new therapies targeting TGF-ß to treat the cardiac alterations of Chagas disease-affected patients.


Subject(s)
Chagas Cardiomyopathy , Trypanosoma cruzi , Chagas Cardiomyopathy/drug therapy , Chagas Cardiomyopathy/metabolism , Heart , Humans , Myocardium/pathology , Transforming Growth Factor beta/antagonists & inhibitors , Trypanosoma cruzi/physiology
2.
Circ Res ; 124(6): 846-855, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30636542

ABSTRACT

RATIONALE: Although many familial cases of pulmonary arterial hypertension exhibit an autosomal dominant mode of inheritance with the majority having mutations in essential constituents of the BMP (bone morphogenetic protein) signaling, the specific contribution of the long-term loss of signal transduction triggered by the BMPR2 (type 2 BMP receptor) remains poorly characterized. OBJECTIVE: To investigate the role of BMP9, the main ligand of ALK1 (Activin receptor-like kinase 1)/BMPR2 heterocomplexes, in pulmonary hypertension. METHOD AND RESULTS: The absence of BMP9 in Bmp9-/- mice and its inhibition in C57BL/6 mice using neutralizing anti-BMP9 antibodies substantially prevent against chronic hypoxia-induced pulmonary hypertension judged by right ventricular systolic pressure measurement, right ventricular hypertrophy, and pulmonary distal arterial muscularization. In agreement with these observations, we found that the BMP9/BMP10 ligand trap ALK1ECD administered in monocrotaline or Sugen/hypoxia (SuHx) rats substantially attenuate proliferation of pulmonary vascular cells, inflammatory cell infiltration, and regresses established pulmonary hypertension in rats. Our data obtained in human pulmonary endothelial cells derived from controls and pulmonary arterial hypertension patients indicate that BMP9 can affect the balance between endothelin-1, apelin, and adrenomedullin. We reproduced these in vitro observations in mice chronically exposed to hypoxia, with Bmp9-/- mice exhibiting lower mRNA levels of the vasoconstrictor peptide ET-1 (endothelin-1) and higher levels of the 2 potent vasodilator factors apelin and ADM (adrenomedullin) compared with Bmp9+/+ littermates. CONCLUSIONS: Taken together, our data indicate that the loss of BMP9, by deletion or inhibition, has beneficial effects against pulmonary hypertension onset and progression.


Subject(s)
Growth Differentiation Factor 2/antagonists & inhibitors , Hypertension, Pulmonary/prevention & control , Activin Receptors, Type II/pharmacology , Animals , Cells, Cultured , Endothelin-1/genetics , Growth Differentiation Factor 2/physiology , Humans , Hypoxia/complications , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar
3.
Hepatology ; 70(4): 1392-1408, 2019 10.
Article in English | MEDLINE | ID: mdl-30964206

ABSTRACT

Bone morphogenetic protein 9 (BMP9) is a circulating factor produced by hepatic stellate cells that plays a critical role in vascular quiescence through its endothelial receptor activin receptor-like kinase 1 (ALK1). Mutations in the gene encoding ALK1 cause hereditary hemorrhagic telangiectasia type 2, a rare genetic disease presenting hepatic vessel malformations. Variations of both the circulating levels and the hepatic mRNA levels of BMP9 have been recently associated with various forms of hepatic fibrosis. However, the molecular mechanism that links BMP9 with liver diseases is still unknown. Here, we report that Bmp9 gene deletion in 129/Ola mice triggers hepatic perisinusoidal fibrosis that was detectable from 15 weeks of age. An inflammatory response appeared within the same time frame as fibrosis, whereas sinusoidal vessel dilation developed later on. Proteomic and mRNA analyses of primary liver sinusoidal endothelial cells (LSECs) both revealed that the expression of the LSEC-specifying transcription factor GATA-binding protein 4 was strongly reduced in Bmp9 gene knockout (Bmp9-KO) mice as compared with wild-type mice. LSECs from Bmp9-KO mice also lost the expression of several terminal differentiation markers (Lyve1, Stab1, Stab2, Ehd3, Cd209b, eNos, Maf, Plvap). They gained CD34 expression and deposited a basal lamina, indicating that they were capillarized. Another main characteristic of differentiated LSECs is the presence of permeable fenestrae. LSECs from Bmp9-KO mice had a significantly reduced number of fenestrae. This was already observable in 2-week-old pups. Moreover, we could show that addition of BMP9 to primary cultures of LSECs prevented the loss of their fenestrae and maintained the expression levels of Gata4 and Plvap. Conclusion: Taken together, our observations show that BMP9 is a key paracrine regulator of liver homeostasis, controlling LSEC fenestration and protecting against perivascular hepatic fibrosis.


Subject(s)
Activin Receptors, Type II/genetics , Endothelial Cells/metabolism , Gene Expression Regulation , Growth Differentiation Factor 2/genetics , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Animals , Cells, Cultured , Disease Models, Animal , Endothelial Cells/cytology , Growth Differentiation Factor 2/metabolism , Hepatic Stellate Cells/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proteomics , RNA, Messenger/genetics , Random Allocation , Statistics, Nonparametric , Tissue Culture Techniques/methods
4.
J Biol Chem ; 293(28): 10963-10974, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29789425

ABSTRACT

Bone morphogenetic protein 9 (BMP9) and BMP10 are the two high-affinity ligands for the endothelial receptor activin receptor-like kinase 1 (ALK1) and are key regulators of vascular remodeling. They are both present in the blood, but their respective biological activities are still a matter of debate. The aim of the present work was to characterize their circulating forms to better understand how their activities are regulated in vivo First, by cotransfecting BMP9 and BMP10, we found that both can form a disulfide-bonded heterodimer in vitro and that this heterodimer is functional on endothelial cells via ALK1. Next, we developed an ELISA that could specifically recognize the BMP9-BMP10 heterodimer and which indicated its presence in both human and mouse plasma. In addition to using available Bmp9-KO mice, we generated a conditional Bmp10-KO mouse strain. The plasma from Bmp10-KO mice, similarly to that of Bmp9-KO mice, completely lacked the ability to activate ALK1-transfected 3T3 cells or phospho-Smad1-5 on endothelial cells, indicating that the circulating BMP activity is mostly due to the BMP9-BMP10 heterodimeric form. This result was confirmed in human plasma that had undergone affinity chromatography to remove BMP9 homodimer. Finally, we provide evidence that hepatic stellate cells in the liver could be the source of the BMP9-BMP10 heterodimer. Together, our findings demonstrate that BMP9 and BMP10 can heterodimerize and that this heterodimer is responsible for most of the biological BMP activity found in plasma.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Endothelium, Vascular/metabolism , Growth Differentiation Factor 2/metabolism , Growth Differentiation Factors/metabolism , Protein Multimerization , 3T3 Cells , Animals , Bone Morphogenetic Proteins/blood , Bone Morphogenetic Proteins/chemistry , Endothelium, Vascular/cytology , Growth Differentiation Factor 2/blood , Growth Differentiation Factor 2/chemistry , Growth Differentiation Factors/blood , Growth Differentiation Factors/chemistry , Humans , Mice , Mice, Knockout , Signal Transduction
5.
Int J Cancer ; 143(4): 944-957, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29516499

ABSTRACT

Adrenocortical carcinoma (ACC) is a tumor with poor prognosis in which overexpression of a panel of microRNAs has been associated with malignancy but a very limited number of investigations on their role in ACC pathogenesis have been conducted. We examined the involvement of miR-483-5p and miR-139-5p in adrenocortical cancer aggressiveness. Using bioinformatics predictions and mRNA/miRNA expression profiles, we performed an integrated analysis to identify inversely correlated miRNA-mRNA pairs in ACC. We identified N-myc downstream-regulated gene family members 2 and 4 (NDRG2 and NDRG4) as targets of miR-483-5p and miR-139-5p, respectively. NDRG2 and NDRG4 expressions were inversely correlated respectively with miR-483-5p and miR-139-5p levels in aggressive ACC samples from two independent cohorts of 20 and 44 ACC. Moreover, upregulation of miR-139-5p and downregulation of NDRG4 demonstrated a striking prognostic value. A direct interaction between miR-483-5p or miR-139-5p and their targets was demonstrated in reporter assays. Downregulation of miR-483-5p or miR-139-5p in the ACC cell lines NCI-H295R and SW13 increased NDRG2 or NDRG4 mRNA and protein expression, compromised adrenocortical cancer cell invasiveness and anchorage-independent growth. MiR-483-5p or miR-139-5p overexpression and NDRG2 or NDRG4 inhibition produce similar changes, which are rescued by NDRG2 or NDRG4 ectopic expression. We established that key factors mediating epithelial-to-mesenchymal transition are downstream effectors of miR-483-5p/NDRG2 and miR-139-5p/NDRG4 pathways. Collectively, our data show for the first time that miR-483-5p/NDRG2 and miR-139-5p/NDRG4 axes promote ACC aggressiveness, with potential implications for prognosis and therapeutic interventions in adrenocortical malignancies.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Genes, myc , MicroRNAs/physiology , Multigene Family , 3' Untranslated Regions , Apoptosis/physiology , Cell Adhesion/physiology , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Down-Regulation , Epithelial-Mesenchymal Transition , HEK293 Cells , Humans , MicroRNAs/genetics , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neoplasm Staging , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Prognosis , RNA, Messenger/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation
6.
Proc Natl Acad Sci U S A ; 112(25): E3207-15, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26056270

ABSTRACT

The transition to pulmonary respiration after birth requires rapid alterations in the structure of the mammalian cardiovascular system. One dramatic change that occurs is the closure of the ductus arteriosus (DA), an arterial connection in the fetus that directs blood flow away from the pulmonary circulation. Two members of the TGFß family, bone morphogenetic protein 9 (BMP9) and BMP10, have been recently involved in postnatal angiogenesis, both being necessary for remodeling of newly formed microvascular beds. The aim of the present work was to study whether BMP9 and BMP10 could be involved in closure of the DA. We found that Bmp9 knockout in mice led to an imperfect closure of the DA. Further, addition of a neutralizing anti-BMP10 antibody at postnatal day 1 (P1) and P3 in these pups exacerbated the remodeling defect and led to a reopening of the DA at P4. Transmission electron microscopy images and immunofluorescence stainings suggested that this effect could be due to a defect in intimal cell differentiation from endothelial to mesenchymal cells, associated with a lack of extracellular matrix deposition within the center of the DA. This result was supported by the identification of the regulation by BMP9 and BMP10 of several genes known to be involved in this process. The involvement of these BMPs was further supported by human genomic data because we could define a critical region in chromosome 2 encoding eight genes including BMP10 that correlated with the presence of a patent DA. Together, these data establish roles for BMP9 and BMP10 in DA closure.


Subject(s)
Bone Morphogenetic Proteins/physiology , Ductus Arteriosus/physiology , Growth Differentiation Factor 2/physiology , Animals , Bone Morphogenetic Proteins/genetics , Ductus Arteriosus/pathology , Growth Differentiation Factor 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Hum Mol Genet ; 24(4): 1142-54, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25312062

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant inheritable vascular dysplasia caused by mutations in genes encoding either endoglin or activin receptor-like kinase-1 (ALK1). Functional significance of endoglin missense mutations remains largely unknown leading to a difficult discrimination between polymorphisms and pathogenic mutations. In order to study the functional significance of endoglin mutations and to help HHT1 diagnosis, we developed a cellular assay based on the ability of endoglin to enhance ALK1 response to bone morphogenetic protein 9 (BMP9). We generated and characterized 31 distinct ENG mutants reproducing human HHT1 missense mutations identified in patients of the Molecular Genetics Department in Lyon. We found that 16 mutants behaved like wild-type (WT) endoglin, and thus corresponded to benign rare variants. The 15 other variants showed defects in BMP9 response and were identified as pathogenic mutations. Interestingly, two mutants (S278P and F282V) had lost their ability to bind BMP9, identifying two crucial amino acids for BMP9 binding to endoglin. For all the others, the functional defect was correlated with a defective trafficking to the cell surface associated with retention in the endoplasmic reticulum. Further, we demonstrated that some intracellular mutants dimerized with WT endoglin and impaired its cell-surface expression thus acting as dominant-negatives. Taken together, we show that endoglin loss-of-function can result from different mechanisms in HHT1 patients. We also provide a diagnostic tool helping geneticists in screening for novel or conflicting ENG mutations.


Subject(s)
Antigens, CD/genetics , Antigens, CD/metabolism , Mutation , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/metabolism , Animals , Antigens, CD/chemistry , Cell Line , Cell Membrane/metabolism , Endoglin , Gene Expression , Growth Differentiation Factor 2 , Growth Differentiation Factors/metabolism , Humans , Mice , Phenotype , Protein Binding , Protein Multimerization , Protein Transport , Receptors, Cell Surface/chemistry , Telangiectasia, Hereditary Hemorrhagic/diagnosis
9.
Am J Physiol Lung Cell Mol Physiol ; 309(3): L314-22, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26047640

ABSTRACT

Enhanced lung angiogenesis has been reported in cystic fibrosis (CF). Recently, two highly homologous ligands, endocrine gland vascular endothelial growth factor (EG-VEGF) and mammalian Bv8, have been described as new angiogenic factors. Both ligands bind and activate two closely related G protein-coupled receptors, the prokineticin receptor (PROKR) 1 and 2. Yet, the expression, regulation, and potential role of EG-VEGF, BV8, and their receptors in normal and CF lung are still unknown. The expression of the receptors and their ligands was examined using molecular, biochemical, and immunocytochemistry analyses in lungs obtained from CF patients vs. control and in normal and CF bronchial epithelial cells. Cystic fibrosis transmembrane conductance regulator (CFTR) activity was evaluated in relation to both ligands, and concentrations of EG-VEGF were measured by ELISA. At the mRNA level, EG-VEGF, BV8, and PROKR2 gene expression was, respectively, approximately five, four, and two times higher in CF lungs compared with the controls. At the cellular level, both the ligands and their receptors showed elevated expressions in the CF condition. Similar results were observed at the protein level. The EG-VEGF secretion was apical and was approximately two times higher in CF compared with the normal epithelial cells. This secretion was increased following the inhibition of CFTR chloride channel activity. More importantly, EG-VEGF and BV8 increased the intracellular concentration of Ca(2+) and cAMP and stimulated CFTR-chloride channel activity. Altogether, these data suggest local roles for epithelial BV8 and EG-VEGF in the CF airway peribronchial vascular remodeling and highlighted the role of CFTR activity in both ligand biosynthesis and secretion.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/metabolism , Gastrointestinal Hormones/metabolism , Neuropeptides/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism , Adult , Aged , Calcium Signaling , Case-Control Studies , Cell Line, Tumor , Chlorides/metabolism , Cystic Fibrosis/genetics , Epithelial Cells/metabolism , Female , Gastrointestinal Hormones/genetics , Gene Expression , Humans , Lung/metabolism , Lung/pathology , Male , Middle Aged , Neuropeptides/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Sequence Deletion , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics , Young Adult
10.
Am J Physiol Endocrinol Metab ; 309(4): E357-69, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26081281

ABSTRACT

PPARγ-deficient mice die at E9.5 due to placental abnormalities. The mechanism by which this occurs is unknown. We demonstrated that the new endocrine factor EG-VEGF controls the same processes as those described for PPARγ, suggesting potential regulation of EG-VEGF by PPARγ. EG-VEGF exerts its functions via prokineticin receptor 1 (PROKR1) and 2 (PROKR2). This study sought to investigate whether EG-VEGF mediates part of PPARγ effects on placental development. Three approaches were used: 1) in vitro, using human primary isolated cytotrophoblasts and the extravillous trophoblast cell line (HTR-8/SVneo); 2) ex vivo, using human placental explants (n = 46 placentas); and 3) in vivo, using gravid wild-type PPARγ(+/-) and PPARγ(-/-) mice. Major processes of placental development that are known to be controlled by PPARγ, such as trophoblast proliferation, migration, and invasion, were assessed in the absence or presence of PROKR1 and PROKR2 antagonists. In both human trophoblast cell and placental explants, we demonstrated that rosiglitazone, a PPARγ agonist, 1) increased EG-VEGF secretion, 2) increased EG-VEGF and its receptors mRNA and protein expression, 3) increased placental vascularization via PROKR1 and PROKR2, and 4) inhibited trophoblast migration and invasion via PROKR2. In the PPARγ(-/-) mouse placentas, EG-VEGF levels were significantly decreased, supporting an in vivo control of EG-VEGF/PROKRs system during pregnancy. The present data reveal EG-VEGF as a new mediator of PPARγ effects during pregnancy and bring new insights into the fine mechanism of trophoblast invasion.


Subject(s)
PPAR gamma/physiology , Placentation , Pregnancy Outcome/genetics , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics , Animals , Benzamides/pharmacology , Cells, Cultured , Cricetinae , Embryo Implantation/drug effects , Embryo Implantation/genetics , Embryo, Mammalian , Female , Humans , Male , Mice , Mice, Transgenic , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , Placenta/metabolism , Pregnancy , Pyridines/pharmacology , Rosiglitazone , Thiazolidinediones/pharmacology , Transcriptional Activation/drug effects , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism
11.
Mol Med ; 21(1): 645-656, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26208047

ABSTRACT

Idiopathic fetal growth restriction (FGR) is frequently associated with placental insufficiency. Previous reports have provided evidence that endocrine gland-derived vascular endothelial growth factor (EG-VEGF), a placental secreted protein, is expressed during the first trimester of pregnancy, controls both trophoblast proliferation and invasion, and its increased expression is associated with human FGR. In this study, we hypothesize that EG-VEGF-dependent changes in placental homeobox gene expressions contribute to trophoblast dysfunction in idiopathic FGR. The changes in EG-VEGF-dependent homeobox gene expressions were determined using a homeobox gene cDNA array on placental explants of 8-12 wks gestation after stimulation with EG-VEGF in vitro for 24 h. The homeobox gene array identified a greater-than-five-fold increase in HOXA9, HOXC8, HOXC10, HOXD1, HOXD8, HOXD9 and HOXD11, while NKX 3.1 showed a greater-than-two-fold decrease in mRNA expression compared with untreated controls. Homeobox gene NKX3.1 was selected as a candidate because it is a downstream target of EG-VEGF and its expression and functional roles are largely unknown in control and idiopathic FGR-affected placentae. Real-time PCR and immunoblotting showed a significant decrease in NKX3.1 mRNA and protein levels, respectively, in placentae from FGR compared with control pregnancies. Gene inactivation in vitro using short-interference RNA specific for NKX3.1 demonstrated an increase in BeWo cell differentiation and a decrease in HTR-8/SVneo proliferation. We conclude that the decreased expression of homeobox gene NKX3.1 downstream of EG-VEGF may contribute to the trophoblast dysfunction associated with idiopathic FGR pregnancies.

12.
Blood ; 122(4): 598-607, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23741013

ABSTRACT

Lymphatic vessels are critical for the maintenance of tissue fluid homeostasis and their dysfunction contributes to several human diseases. The activin receptor-like kinase 1 (ALK1) is a transforming growth factor-ß family type 1 receptor that is expressed on both blood and lymphatic endothelial cells (LECs). Its high-affinity ligand, bone morphogenetic protein 9 (BMP9), has been shown to be critical for retinal angiogenesis. The aim of this work was to investigate whether BMP9 could play a role in lymphatic development. We found that Bmp9 deficiency in mice causes abnormal lymphatic development. Bmp9-knockout (KO) pups presented hyperplastic mesenteric collecting vessels that maintained LYVE-1 expression. In accordance with this result, we found that BMP9 inhibited LYVE-1 expression in LECs in an ALK1-dependent manner. Bmp9-KO pups also presented a significant reduction in the number and in the maturation of mesenteric lymphatic valves at embryonic day 18.5 and at postnatal days 0 and 4. Interestingly, the expression of several genes known to be involved in valve formation (Foxc2, Connexin37, EphrinB2, and Neuropilin1) was upregulated by BMP9 in LECS. Finally, we demonstrated that Bmp9-KO neonates and adult mice had decreased lymphatic draining efficiency. These data identify BMP9 as an important extracellular regulator in the maturation of the lymphatic vascular network affecting valve development and lymphatic vessel function.


Subject(s)
Growth Differentiation Factor 2/physiology , Lymphangiogenesis/genetics , Lymphatic Vessels/physiology , Mesentery/embryology , Animals , Animals, Newborn , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/physiology , Gene Expression Regulation, Developmental , Glycoproteins/genetics , Glycoproteins/metabolism , Growth Differentiation Factor 2/genetics , Growth Differentiation Factor 2/metabolism , Humans , Lymphangiogenesis/physiology , Lymphatic Vessels/metabolism , Membrane Transport Proteins , Mesentery/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
PLoS Genet ; 8(5): e1002700, 2012.
Article in English | MEDLINE | ID: mdl-22589739

ABSTRACT

Sporadic adrenocortical carcinomas (ACC) are rare endocrine neoplasms with a dismal prognosis. By contrast, benign tumors of the adrenal cortex are common in the general population. Whether benign tumors represent a separate entity or are in fact part of a process of tumor progression ultimately leading to an ACC is still an unresolved issue. To this end, we have developed a mouse model of tumor progression by successively transducing genes altered in adrenocortical tumors into normal adrenocortical cells. The introduction in different orders of the oncogenic allele of Ras (H-Ras(G12V)) and the mutant p53(DD) that disrupts the p53 pathway yielded tumors displaying major differences in histological features, tumorigenicity, and metastatic behavior. Whereas the successive expression of Ras(G12V) and p53(DD) led to highly malignant tumors with metastatic behavior, reminiscent of those formed after the simultaneous introduction of p53(DD) and Ras(G12V), the reverse sequence gave rise only to benign tumors. Microarray profiling revealed that 157 genes related to cancer development and progression were differentially expressed. Of these genes, 40 were up-regulated and 117 were down-regulated in malignant cell populations as compared with benign cell populations. This is the first evidence-based observation that ACC development follows a multistage progression and that the tumor phenotype is directly influenced by the order of acquisition of genetic alterations.


Subject(s)
Adrenal Cortex Neoplasms , Adrenal Cortex , Cell Transformation, Neoplastic/genetics , Oncogene Protein p21(ras)/genetics , Tumor Suppressor Protein p53/genetics , Adrenal Cortex/cytology , Adrenal Cortex/metabolism , Adrenal Cortex Neoplasms/genetics , Animals , Cell Proliferation , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, SCID , Transduction, Genetic
14.
Blood ; 119(25): 6162-71, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22566602

ABSTRACT

ALK1 is a type I receptor of the TGF-ß family that is involved in angiogenesis. Circulating BMP9 was identified as a specific ligand for ALK1 inducing vascular quiescence. In this work, we found that blocking BMP9 with a neutralizing antibody in newborn mice significantly increased retinal vascular density. Surprisingly, Bmp9-KO mice did not show any defect in retinal vascularization. However, injection of the extracellular domain of ALK1 impaired retinal vascularization in Bmp9-KO mice, implicating another ligand for ALK1. Interestingly, we detected a high level of circulating BMP10 in WT and Bmp9-KO pups. Further, we found that injection of a neutralizing anti-BMP10 antibody to Bmp9-KO pups reduced retinal vascular expansion and increased vascular density, whereas injection of this antibody to WT pups did not affect the retinal vasculature. These data suggested that BMP9 and BMP10 are important in postnatal vascular remodeling of the retina and that BMP10 can substitute for BMP9. In vitro stimulation of endothelial cells by BMP9 and BMP10 increased the expression of genes involved in the Notch signaling pathway (Jagged1, Dll4, Hey1, Hey2, Hes1) and decreased apelin expression, suggesting a possible cross-talk between these pathways and the BMP pathway.


Subject(s)
Bone Morphogenetic Proteins/physiology , Growth Differentiation Factor 2/physiology , Retinal Vessels/physiology , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/pharmacology , Activin Receptors, Type II , Animals , Animals, Newborn , Antibodies/pharmacology , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Count , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Growth Differentiation Factor 2/antagonists & inhibitors , Growth Differentiation Factor 2/genetics , Growth Differentiation Factor 2/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Protein Structure, Tertiary , Recombinant Proteins/pharmacology , Retinal Vessels/cytology , Retinal Vessels/drug effects , Retinal Vessels/metabolism
15.
Cell Mol Life Sci ; 70(12): 2031-44, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22968342

ABSTRACT

Members of the tristetraprolin (TTP/TIS11) family are important RNA-binding proteins initially characterized as mediators of mRNA degradation. They act via their interaction with AU-rich elements present in the 3'UTR of regulated transcripts. However, it is progressively appearing that the different steps of mRNA processing and fate including transcription, splicing, polyadenylation, translation, and degradation are coordinately regulated by multifunctional integrator proteins that possess a larger panel of functions than originally anticipated. Tristetraprolin and related proteins are very good examples of such integrators. This review gathers the present knowledge on the functions of this family of RNA-binding proteins, including their role in AU-rich element-mediated mRNA decay and focuses on recent advances that support the concept of their broader involvement in distinct steps of mRNA biogenesis and degradation.


Subject(s)
Gene Expression Regulation/physiology , Multigene Family/genetics , Protein Biosynthesis/physiology , RNA Stability/physiology , RNA, Messenger/biosynthesis , RNA-Binding Proteins/metabolism , Signal Transduction/physiology , Tristetraprolin/metabolism , 3' Untranslated Regions/genetics , 3' Untranslated Regions/physiology , AU Rich Elements/genetics , AU Rich Elements/physiology , Amino Acid Sequence , Animals , Gene Components , Gene Expression Regulation/genetics , Mice , Models, Biological , Molecular Sequence Data , Phenotype , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Signal Transduction/genetics , Tristetraprolin/genetics
16.
Med Sci (Paris) ; 30(3): 274-9, 2014 Mar.
Article in French | MEDLINE | ID: mdl-24685218

ABSTRACT

During the last decade, there has been growing evidence for the involvement of prokineticins and their receptors (PROK/PROKR) in human reproduction, with multiple roles in the female and male reproductive systems. The PROK/PROKR signalling complex has been reported as a new actor in ovary, uterus, placenta, and testis physiology, with marked dysfunction in various pathological conditions such as polycystic ovary syndrome, recurrent pregnancy loss, preeclampsia, and ectopic pregnancy. Altogether, the results strongly suggest the involvement of prokineticins in spermatogenesis, oocyte competence, embryo implantation, pregnancy, and delivery, and argue for the clinical relevance of these cytokines and their receptors as diagnostic markers for several reproductive diseases.


Subject(s)
Gastrointestinal Hormones/physiology , Neuropeptides/physiology , Reproduction/physiology , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/physiology , Animals , Female , Humans , Male , Neuropeptides/isolation & purification , Pregnancy , Receptors, G-Protein-Coupled/physiology , Snake Venoms/chemistry
17.
Cell Mol Life Sci ; 69(2): 313-24, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21710321

ABSTRACT

Bone Morphogenetic Protein 9 (BMP9) has been recently found to be the physiological ligand for the activin receptor-like kinase 1 (ALK1), and to be a major circulating vascular quiescence factor. Moreover, a soluble chimeric ALK1 protein (ALK1-Fc) has recently been developed and showed powerful anti-tumor growth and anti-angiogenic effects. However, not much is known concerning BMP9. This prompted us to investigate the human endogenous sources of this cytokine and to further characterize its circulating form(s) and its function. Analysis of BMP9 expression reveals that BMP9 is produced by hepatocytes and intrahepatic biliary epithelial cells. Gel filtration analysis combined with ELISA and biological assays demonstrate that BMP9 circulates in plasma (1) as an unprocessed inactive form that can be further activated by furin a serine endoprotease, and (2) as a mature and fully active form (composed of the mature form associated with its prodomain). Analysis of BMP9 circulating levels during mouse development demonstrates that BMP9 peaks during the first 3 weeks after birth and then decreases to 2 ng/mL in adulthood. We also show that circulating BMP9 physiologically induces a constitutive Smad1/5/8 phosphorylation in endothelial cells. Taken together, our results argue for the role of BMP9 as a hepatocyte-derived factor, circulating in inactive (40%) and active (60%) forms, the latter constantly activating endothelial cells to maintain them in a resting state.


Subject(s)
Growth Differentiation Factor 2/blood , Growth Differentiation Factors/biosynthesis , Hepatocytes/metabolism , Adult , Animals , Aorta/metabolism , Bile Ducts, Intrahepatic/metabolism , Epithelial Cells/metabolism , Female , Growth Differentiation Factors/blood , Humans , Male , Mice , Middle Aged , NIH 3T3 Cells , Proprotein Convertases/metabolism , Rats , Rats, Wistar , Serine Endopeptidases/metabolism , Smad Proteins/metabolism
18.
J Cell Physiol ; 227(11): 3593-602, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22287283

ABSTRACT

The lymphatic vasculature is essential for the maintenance of tissue fluid, immune surveillance, and dissemination of metastasis. Recently, several models for lymphatic vascular research and markers specific for lymphatic endothelium have been characterized. Despite these significant achievements, our understanding of the early lymphatic development is still rather limited. The purpose of the study was to further define early lymphatic differentiation regulatory pathways. In the present study, we have developed conditions leading to lymphatic endothelial cell differentiation under both serum-rich and serum-free conditions, using the coculture system of Flk-1-positive vascular precursors derived from murine embryonic stem (ES) cells grown on an OP9 stromal cell layer. In this work, we also identified Transforming Growth Factor-ß1 (TGFß1) as a negative regulator of lymphvasculogenesis from ES-derived vascular progenitors. Finally, we could show that TGFß1 addition decreases COUP-TFII and Sox18 mRNA levels, which are two transcription factors known to be involved in early lymphatic endothelial differentiation. Taken together these findings support the concept that manipulating the TGFß signaling pathway may represent an interesting target to favor lymphatic endothelial cell expansion for cell replacement strategies.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Endothelial Cells , Transforming Growth Factor beta1/metabolism , Animals , COUP Transcription Factor II/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cell Lineage/genetics , Coculture Techniques , Culture Media, Serum-Free , Embryonic Stem Cells/physiology , Endothelial Cells/cytology , Endothelial Cells/physiology , Gene Expression Regulation, Developmental/drug effects , Glycoproteins/metabolism , Membrane Transport Proteins , Mice , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , SOXF Transcription Factors/metabolism , Signal Transduction , Stromal Cells/cytology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
Blood ; 116(9): 1604-12, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20501893

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant genetically inheritable vascular dysplasia caused by mutations in genes encoding receptors of the transforming growth factor-beta (TGF-beta) family: ENG, encoding endoglin (HHT1), and ACVRL1, encoding activin receptor-like kinase-1 (ALK1; HHT2). Our recent discovery of bone morphogenetic protein 9 (BMP9) as the specific ligand for ALK1 allowed us to reevaluate the functional significance of ACVRL1 mutations. We generated 19 ALK1 mutants reproducing HHT2 mutations (4 were novel mutations) found throughout the protein. We show that all ALK1 mutant proteins were expressed by transfected cells; most of them were present at the cell surface and retained their ability to bind BMP9 (except for the extracellular mutants). However, most were defective in BMP9 signaling. None of the ALK1 mutants had a dominant negative effect on wild-type ALK1 activity. These data demonstrate that mutations of ACVRL1 fit with a functional haploinsufficiency model affecting BMP9 signaling. Our study also identified 4 ACVRL1 mutations (D179A, R386C, R454W, and A482V) that did not alter the BMP9 responses that are polymorphisms and 2 novel mutations that are pathogenic (L381P and I485F). This demonstrates that the analysis of BMP9 responses can be used as a diagnostic tool by geneticists confronted with novel or conflicting ACVRL1 mutations.


Subject(s)
Activin Receptors, Type II/genetics , Growth Differentiation Factors/metabolism , Mutation/genetics , Telangiectasia, Hereditary Hemorrhagic/diagnosis , Telangiectasia, Hereditary Hemorrhagic/genetics , Animals , Blotting, Western , Flow Cytometry , Growth Differentiation Factor 2 , Humans , Immunoprecipitation , Luciferases/metabolism , Mice , NIH 3T3 Cells , Prognosis
20.
Front Cell Infect Microbiol ; 12: 1017040, 2022.
Article in English | MEDLINE | ID: mdl-36530434

ABSTRACT

Chronic Chagasic cardiomyopathy (CCC), a progressive inflammatory and fibrosing disease, is the most prominent clinical form of Chagas disease, a neglected tropical disease caused by Trypanosoma cruzi infection. During CCC, the parasite remains inside the cardiac cells, leading to tissue damage, involving extensive inflammatory response and irregular fibrosis. Among the fibrogenic factors is transforming growth factor-ß (TGF-ß), a key cytokine controlling extracellular matrix synthesis and degradation. TGF-ß is involved in CCC onset and progression, with increased serum levels and activation of its signaling pathways in the cardiac tissue, which crucially contributes to fibrosis. Inhibition of the TGF-ß signaling pathway attenuates T. cruzi infection and prevents cardiac damage in an experimental model of acute Chagas disease. The aim of this study was to investigate the effect of TGF-ß neutralization on T. cruzi infection in both in vitro and in vivo pre-clinical models, using the 1D11 monoclonal antibody. To this end, primary cultures of cardiac cells were infected with T. cruzi trypomastigote forms and treated with 1D11. For in vivo studies, 1D11 was administered in different schemes for acute and chronic phase models (Swiss mice infected with 104 parasites from the Y strain and C57BL/6 mice infected with 102 parasites from the Colombian strain, respectively). Here we show that the addition of 1D11 to cardiac cells greatly reduces cardiomyocyte invasion by T. cruzi and the number of parasites per infected cell. In both acute and chronic experimental models, T. cruzi infection altered the electrical conduction, decreasing the heart rate, increasing the PR interval and the P wave duration. The treatment with 1D11 reduced cardiac fibrosis and reversed electrical abnormalities improving cardiac performance. Taken together, these data further support the major role of the TGF-ß signaling pathways in T. cruzi-infection and their biological consequences on parasite/host interactions. The therapeutic effects of the 1D11 antibody are promising and suggest a new possibility to treat cardiac fibrosis in the chronic phase of Chagas' heart disease by TGF-ß neutralization.


Subject(s)
Chagas Cardiomyopathy , Chagas Disease , Trypanosoma cruzi , Mice , Animals , Transforming Growth Factor beta/metabolism , Chagas Cardiomyopathy/drug therapy , Trypanosoma cruzi/metabolism , Mice, Inbred C57BL , Chagas Disease/drug therapy , Chagas Disease/parasitology , Fibrosis
SELECTION OF CITATIONS
SEARCH DETAIL