Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Oncotarget ; 8(34): 56228-56242, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28915586

ABSTRACT

Hepatitis C virus (HCV) is a leading cause of liver diseases including the development of hepatocellular carcinoma (HCC). Particularly, core protein has been involved in HCV-related liver pathologies. However, the impact of HCV core on signaling pathways supporting the genesis of HCC remains largely elusive. To decipher the host cell signaling pathways involved in the oncogenic potential of HCV core, a global quantitative phosphoproteomic approach was carried out. This study shed light on novel differentially phosphorylated proteins, in particular several components involved in translation. Among the eukaryotic initiation factors that govern the translational machinery, 4E-BP1 represents a master regulator of protein synthesis that is associated with the development and progression of cancers due to its ability to increase protein expression of oncogenic pathways. Enhanced levels of 4E-BP1 in non-modified and phosphorylated forms were validated in human hepatoma cells and in mouse primary hepatocytes expressing HCV core, in the livers of HCV core transgenic mice as well as in HCV-infected human primary hepatocytes. The contribution of HCV core in carcinogenesis and the status of 4E-BP1 expression and phosphorylation were studied in HCV core/Myc double transgenic mice. HCV core increased the levels of 4E-BP1 expression and phosphorylation and significantly accelerated the onset of Myc-induced tumorigenesis in these double transgenic mice. These results reveal a novel function of HCV core in liver carcinogenesis potentiation. They position 4E-BP1 as a tumor-specific target of HCV core and support the involvement of the 4E-BP1/eIF4E axis in hepatocarcinogenesis.

2.
Cancer Res ; 75(22): 4753-65, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26404004

ABSTRACT

Primary myelofibrosis is a myeloproliferative neoplasm that is a precursor to myeloid leukemia. Dysmegakaryopoiesis and extramedullary hematopoiesis characterize primary myelofibrosis, which is also associated with bone marrow stromal alterations marked by fibrosis, neoangiogenesis, and osteomyelosclerosis. In particular, contributions to primary myelofibrosis from mesenchymal stromal cells (MSC) have been suggested by mouse studies, but evidence in humans remains lacking. In this study, we show that bone marrow MSCs from primary myelofibrosis patients exhibit unique molecular and functional abnormalities distinct from other myeloproliferative neoplasms and these abnormalities are maintained stably ex vivo in the absence of leukemic cells. Primary myelofibrosis-MSC overexpressed heparin-binding cytokines, including proinflammatory TGFß1 and osteogenic BMP-2, as well as glycosaminoglycans such as heparan sulfate and chondroitin sulfate. Transcriptome and functional analyses revealed alterations in MSC differentiation characterized by an increased osteogenic potential and a TGFß1 signaling signature. Accordingly, phospho-Smad2 levels were intrinsically increased in primary myelofibrosis-MSC along with enhanced expression of the master bone regulator RUNX2, while inhibition of the endogenous TGFß1 receptor TGFßR1 impaired osteogenic differentiation in these MSCs. Taken together, our results define the source of a critical osteogenic function in primary myelofibrosis that supports its pathophysiology, suggesting that combined targeting of both the hematopoietic and stromal cell compartments in primary myelofibrosis patients may heighten therapeutic efficacy.


Subject(s)
Cell Differentiation/physiology , Mesenchymal Stem Cells/pathology , Ossification, Heterotopic/physiopathology , Primary Myelofibrosis/physiopathology , Adult , Aged , Aged, 80 and over , Animals , Cells, Cultured , Female , Heterografts , Humans , Male , Mice , Mice, Nude , Middle Aged , Polymerase Chain Reaction
3.
PLoS One ; 10(6): e0130559, 2015.
Article in English | MEDLINE | ID: mdl-26110787

ABSTRACT

BACKGROUND AND AIMS: The prognosis of hepatocellular carcinoma (HCC) is hampered by frequent tumour recurrence and metastases. Epithelial-Mesenchymal Transition (EMT) is now recognized as a key process in tumour invasion, metastasis and the generation of cancer initiating cells. The morphological identification of EMT in tumour samples from the expression of novel mesenchymal markers could provide relevant prognostic information and aid in understanding the metastatic process. METHODS: The expression of Smooth Muscle Actins was studied using immunofluorescence and immunohistochemistry assays in cultured liver cells during an induced EMT process and in liver specimens from adult and paediatric HCC series. RESULTS: We report here that in HCC cell lines treated with TGF-ß and in HCC specimens, the expression of αSMA, a known mesenchymal marker of EMT, could never be detected. In addition, our in vitro studies identified the enteric form of SMA, γSMA, as being a marker of EMT. Moreover, this SMA isoform was expressed in 46% of 58 tumours from 42 adult HCC patients and in 90% of 16 tumours from 12 paediatric HCC patients. Interestingly, this expression was significantly correlated with poor tumour differentiation and progenitor cell features characterized by the expression of EpCAM and K19. CONCLUSION: Taken together, our results support the conclusion that γSMA expression in HCC is strongly correlated with the EMT process, HCC aggressiveness and the identification of cancer stem cells. This correlation suggests that γSMA represents a novel and powerful marker to predict HCC progression.


Subject(s)
Actins/metabolism , Carcinoma, Hepatocellular/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Adult , Aged , Antigens, Neoplasm/metabolism , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cell Line, Tumor , Cohort Studies , Epithelial Cell Adhesion Molecule , Female , Humans , Keratin-19/metabolism , Male , Microscopy, Fluorescence , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Odds Ratio , Prognosis , Recombinant Proteins/chemistry , Stem Cells/cytology , Transforming Growth Factor beta/metabolism
4.
J Appl Toxicol ; 35(12): 1520-30, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25663515

ABSTRACT

Aristolochic acids (AA) are nephrotoxic and profibrotic agents, leading to chronic kidney disease. As some controversial studies have reported a nephroprotective effect of exogenous recombinant human bone morphogenetic protein (rhBMP)-7 in several models of renal fibrosis, we investigated the putative effect of rhBMP-7 to prevent progressive tubulointerstitial damage after AA intoxication in vitro and in vivo. In vitro, the toxicity of AA on renal tubular cells was demonstrated by an increase in vimentin as well as a decrease in ß-catenin expressions, reflecting a dedifferentiation process. Increased fibronectin and interleukin-6 levels were measured in the supernatants. Enhanced α-SMA mRNA levels associated to decreased E-cadherin mRNA levels were also measured. Incubation with rhBMP-7 only prevented the increase in vimentin and the decrease in ß-catenin expressions. In vivo, in a rat model of AA nephropathy, severe tubulointerstitial lesions induced by AA after 10 and 35 days (collagen IV deposition and tubular atrophy), were not prevented by the rhBMP-7 treatment. Similarly, rhBMP-7 did not ameliorate the significant increase in urinary concentrations of transforming growth factor-ß. In summary, our in vitro data demonstrated a poor beneficial effect of rhBMP-7 to reverse cell toxicity while, in vivo, there was no beneficial effect of rhBMP-7. Therefore, further investigations are needed to confirm the exact role of BMP-7 in progressive chronic kidney disease.


Subject(s)
Aristolochic Acids/toxicity , Bone Morphogenetic Protein 7/therapeutic use , Kidney/drug effects , Renal Insufficiency, Chronic/prevention & control , Animals , Bone Morphogenetic Protein 7/administration & dosage , Cell Line , Fibronectins/metabolism , Fibrosis , Humans , Kidney/metabolism , Kidney/pathology , Male , Rats, Wistar , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/urine , Treatment Outcome , Vimentin/biosynthesis , beta Catenin/metabolism
5.
Article in English | MEDLINE | ID: mdl-24798596

ABSTRACT

Diagnosis of endemic nephropathy (EN) is based on the combination of several clinical and laboratory criteria. Despite extensive research no specific diagnostic biomarker for EN has yet been identified. The aim of the study was to evaluate the diagnostic significance of the variables previously proposed as diagnostic criteria, but also new ones. After an extended questionnaire, the clinical and laboratory examination population in EN villages was classified according to the modified WHO criteria. The urinary active form of TGF-ß was measured with a bioassay using a cell line which expresses luciferase activity. In the study we used ROC analysis to examine the predictive value of the tested variables. In the study there was no difference in haemoglobin level between the study subgroups. Leucine aminopeptidase (LAP) in urine and active urinary TGF-ß levels were increased in the EN diseased group when compared to other subgroups, but they did not fulfil the statistical criteria needed for differentiating a diseased form from other study subgroups. Both kidney length and parenchima thickness, alfa1-microglobulinuria, and kidney function assessed by MDRD formula were the variables that differentiated the study subgroups well. Based on our results the cut-off value of alfa1-microglobulin for screening should be 23.5 mg/g creatinine instead of 15 mg/g creatinine in the present criteria, and for making a diagnosis of EN 31,5 mg/g creatinine. Persons with a positive family history for EN had a 5.8 times greater risk of developing EN when compared to a negative one. Taken together, the above-mentioned variables should be implemented in new uniform diagnostic criteria for EN.


Subject(s)
Balkan Nephropathy/diagnosis , Kidney/pathology , Adolescent , Adult , Aristolochic Acids/metabolism , Balkan Nephropathy/metabolism , Biomarkers/metabolism , Diagnosis, Differential , Female , Humans , Kidney/metabolism , Male , Transforming Growth Factor beta/metabolism , Young Adult
6.
J Hepatol ; 59(6): 1160-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23928402

ABSTRACT

BACKGROUND & AIMS: The mechanisms by which fibrosis, cirrhosis, and hepatocellular carcinoma (HCC) develop during chronic hepatitis C virus (HCV) infection are not fully understood. We previously observed that HCV core protein induced a TGF-ß-dependent epithelial mesenchymal transition, a process contributing to the promotion of cell invasion and metastasis by impacting TGF-ß1 signalling. Here we investigated HCV core capacity to drive increased expression of the active form of TGF-ß1n transgenic mice and hepatoma cell lines. METHODS: We used an in vivo model of HCV core expressing transgenic mice. RESULTS: We observed that about 50% of genes deregulated by core protein expression were TGF-ß1 target genes. Active TGF-ß levels were increased in HCV core transgenic mouse livers. Overexpression of core protein in hepatoma cells increased active TGF-ß levels in culture supernatants and induced Smad2/3 phosphorylation, thus reflecting activation of the TGF-ß signaling pathway. Moreover, our data showed the implication of thrombospondin-1 in core-dependent TGF-ß activation. Finally, hepatoma cells expressing HCV core could activate stellate cells in co-culture and this activation was TGF-ß dependent. CONCLUSIONS: Collectively, these data delineate a novel paradigm where HCV may be related to liver pathogenesis through its ability to induce a local, intrahepatic TGF-ß activation. They argue for a dual impact of HCV core on liver fibrosis and liver carcinogenesis: HCV core could act both as autocrine and paracrine factor modulating TGF-ß responses within hepatocytes and in stromal environment through TGF-ß activation.


Subject(s)
Hepacivirus/physiology , Hepatocytes/physiology , Thrombospondin 1/physiology , Transforming Growth Factor beta/physiology , Animals , Humans , Mice , Mice, Transgenic
7.
Cell Rep ; 4(3): 530-41, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23911286

ABSTRACT

The homeodomain protein TGIF (TG-interacting factor) restricts TGF-ß/Smad cytostatic signaling by interfering with the nucleocytoplasmic transit of the tumor suppressor cPML. Here, we identify PHRF1 as a ubiquitin ligase that enforces TGIF decay by driving its ubiquitination at lysine 130. In so doing, PHRF1 ensures redistribution of cPML into the cytoplasm, where it associates with SARA and coordinates activation of Smad2 by the TGF-ß receptor. The PHRF1 gene resides within the tumor suppressor locus 11p15.5, which displays frequent loss in a wide variety of malignancies, including breast cancer. Remarkably, we found that the PHRF1 gene is deleted or silenced in a high proportion of human breast cancer samples and cancer cell lines. Reconstitution of PHRF1 into deficient cells impeded their propensity to form tumors in vivo, most likely because of the reemergence of TGF-ß responsiveness. These findings unveil a paradigm behind inactivation of the cPML tumor suppressor network in human malignancies.


Subject(s)
Breast Neoplasms/genetics , Interferon Regulatory Factor-7/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/genetics , Animals , Breast Neoplasms/metabolism , Dogs , Female , Genes, Tumor Suppressor , Hep G2 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Interferon Regulatory Factor-7/metabolism , Madin Darby Canine Kidney Cells , Nuclear Proteins/genetics , Phosphorylation , Promyelocytic Leukemia Protein , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription, Genetic , Transfection , Transforming Growth Factor beta/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitination
8.
Cancer Res ; 71(8): 2901-15, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21487043

ABSTRACT

Primary myelofibrosis (PMF) is characterized by increased number of hematopoietic progenitors and a dysmegakaryopoiesis which supports the stromal reaction defining this disease. We showed that increased ligand (FL) levels in plasma, hematopoietic progenitors, and stromal cells from PMF patients were associated with upregulation of the cognate Flt3 receptor on megakaryocytic (MK) cells. This connection prompted us to study a functional role for the FL/Flt3 couple in PMF dysmegakaryopoiesis, as a route to reveal insights into pathobiology and therapy in this disease. Analysis of PMF CD34(+) and MK cell transcriptomes revealed deregulation of the mitogen-activated protein kinase (MAPK) pathway along with Flt3 expression. In PMF patients, a higher proportion of circulating Flt3(+)CD34(+)CD41(+) cells exhibited an increased MAPK effector phosphorylation independently of Jak2(V617F) mutation. Activation of FL/Flt3 axis in PMF MK cell cultures, in response to FL, induced activation of the p38-MAPK cascade, which is known to be involved in inflammation, also increasing expression of its target genes (NFATC4, p53, AP-1, IL-8). Inhibiting Flt3 or MAPK or especially p38 by chemical, antibody, or silencing strategies restored megakaryopoiesis and reduced phosphorylation of Flt3 and p38 pathway effectors, confirming the involvement of Flt3 in PMF dysmegakaryopoiesis via p38 activation. In addition, in contrast to healthy donors, MK cells derived from PMF CD34(+) cells exhibited an FL-induced migration that could be reversed by p38 inhibition. Taken together, our results implicate the FL/Flt3 ligand-receptor complex in PMF dysmegakaryopoiesis through persistent p38-MAPK activation, with implications for therapeutic prospects to correct altered megakaryopoiesis in an inflammatory context.


Subject(s)
Megakaryocytes/metabolism , Mitogen-Activated Protein Kinases/metabolism , Primary Myelofibrosis/metabolism , fms-Like Tyrosine Kinase 3/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Antigens, CD34/biosynthesis , Enzyme Activation , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , MAP Kinase Signaling System , Megakaryocytes/enzymology , Megakaryocytes/pathology , Membrane Proteins/blood , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Phosphorylation , Primary Myelofibrosis/blood , Primary Myelofibrosis/enzymology , Stromal Cells/enzymology , Stromal Cells/metabolism , Stromal Cells/pathology , fms-Like Tyrosine Kinase 3/biosynthesis
10.
PLoS One ; 4(2): e4355, 2009.
Article in English | MEDLINE | ID: mdl-19190755

ABSTRACT

BACKGROUND: Chronic hepatitis C virus (HCV) infection and associated liver cirrhosis represent a major risk factor for hepatocellular carcinoma (HCC) development. TGF-beta is an important driver of liver fibrogenesis and cancer; however, its actual impact in human cancer progression is still poorly known. The aim of this study was to investigate the role of HCC-derived HCV core natural variants on cancer progression through their impact on TGF-beta signaling. PRINCIPAL FINDINGS: We provide evidence that HCC-derived core protein expression in primary human or mouse hepatocyte alleviates TGF-beta responses in terms or growth inhibition or apoptosis. Instead, in these hepatocytes TGF-beta was still able to induce an epithelial to mesenchymal transition (EMT), a process that contributes to the promotion of cell invasion and metastasis. Moreover, we demonstrate that different thresholds of Smad3 activation dictate the TGF-beta responses in hepatic cells and that HCV core protein, by decreasing Smad3 activation, may switch TGF-beta growth inhibitory effects to tumor promoting responses. CONCLUSION/SIGNIFICANCE: Our data illustrate the capacity of hepatocytes to develop EMT and plasticity under TGF-beta, emphasize the role of HCV core protein in the dynamic of these effects and provide evidence for a paradigm whereby a viral protein implicated in oncogenesis is capable to shift TGF-beta responses from cytostatic effects to EMT development.


Subject(s)
Epithelium/pathology , Liver Neoplasms/pathology , Liver Neoplasms/virology , Mesoderm/pathology , Transforming Growth Factor beta/pharmacology , Viral Core Proteins/metabolism , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelium/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Mesoderm/drug effects , Mice , Mutant Proteins/metabolism , Smad3 Protein/metabolism
11.
Mol Cell ; 36(6): 1073-85, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-20064471

ABSTRACT

The homeodomain protein TGIF functions as a negative regulator of multiple classes of transcription factors. Here we report on the characterization of TGIF as an essential component of the tumor necrosis factor alpha (TNF-alpha) cytotoxic program. This proapoptotic role of TGIF does not appear to rely on transcriptional modulation but instead is executed in conjunction with Itch/AIP4, an E3 ubiquitin ligase operating in TNF-alpha-induced apoptosis through its ability to target the caspase antagonist cFlip(L) for degradation. Notably, we found that activation of TNF-alpha signaling induced the association of TGIF with Itch/AIP4, resulting in increased accessibility of cFlip(L) for association and ubiquitination by Itch/AIP4. Moreover, we show that Itch/AIP4 can also stabilize the TGIF protein in response to TNF-alpha by triggering its monoubiquitination at lysine 259, thereby revealing the existence of a functional network that can evolve into a positive feedback loop for ensuring effective execution of the TNF-alpha apoptotic signaling.


Subject(s)
Cell Death/drug effects , Homeodomain Proteins/pharmacology , Repressor Proteins/pharmacology , Ubiquitin-Protein Ligases/metabolism , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Line , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lysine/metabolism , Mice , Mice, Knockout , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitination
12.
Stem Cells ; 26(12): 3150-61, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18757300

ABSTRACT

Cell cycle regulation plays a fundamental role in stem cell biology. A balance between quiescence and proliferation of hematopoietic stem cells in interaction with the microenvironment is critical for sustaining long-term hematopoiesis and for protection against stress. We analyzed the molecular mechanisms by which stromal cell-derived factor-1 (SDF-1) exhibited a cell cycle-promoting effect and interacted with transforming growth factor-beta (TGF-beta), which has negative effects on cell cycle orchestration of human hematopoietic CD34(+) progenitor cells. We demonstrated that a low concentration of SDF-1 modulated the expression of key cell cycle regulators such as cyclins, cyclin-dependent kinase inhibitors, and TGF-beta target genes, confirming its cell cycle-promoting effect. We showed that a cross-talk between SDF-1- and TGF-beta-related signaling pathways involving phosphatidylinositol 3-kinase (PI3K)/Akt phosphorylation participated in the control of CD34(+) cell cycling. We demonstrated a pivotal role of two downstream effectors of the PI3K/Akt pathway, FoxO3a and mammalian target of rapamycin, as connectors in the SDF-1-/TGF-beta-induced control of the cycling/quiescence switch and proposed a model integrating a dialogue between the two molecules in cell cycle progression. Our data shed new light on the signaling pathways involved in SDF-1 cell cycle-promoting activity and suggest that the balance between SDF-1- and TGF-beta-activated pathways is critical for the regulation of hematopoietic progenitor cell cycle status.


Subject(s)
Chemokine CXCL12/metabolism , Forkhead Transcription Factors/metabolism , Protein Kinases/metabolism , Transforming Growth Factor beta/metabolism , Antigens, CD34/biosynthesis , Cell Cycle , Forkhead Box Protein O3 , Hematopoiesis , Hematopoietic Stem Cells/cytology , Humans , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stem Cells/cytology , TOR Serine-Threonine Kinases
13.
EMBO J ; 27(13): 1804-15, 2008 Jul 09.
Article in English | MEDLINE | ID: mdl-18511908

ABSTRACT

The TGIF homoeodomain protein functions as an important negative regulator in the TGF-beta signalling pathway. The inhibitory function of TGIF is executed in part through its ability to sequester the tumour suppressor cytoplasmic promyelocytic leukaemia (cPML) in the nucleus, thereby preventing the phosphorylation of Smad2 by the activated TGF-beta type I receptor. Here, we report on the identification of PCTA (PML competitor for TGIF association), a TGIF antagonist that promotes TGF-beta-induced transcriptional and cytostatic responses. We provide evidence that PCTA functions in TGF-beta signalling by relieving the suppression of Smad2 phosphorylation by TGIF. Furthermore, we demonstrate that PCTA selectively competes with cPML for TGIF association, resulting in the accumulation of cPML in the cytoplasm, where it associates with SARA and coordinates the access of Smad2 for phosphorylation by the activated TGF-beta type I receptor. Thus, our findings on the mode of action of PCTA provide new and important insights into the molecular mechanism underlying the antagonistic interplay between TGIF and cPML in the TGF-beta signalling network.


Subject(s)
Carrier Proteins/metabolism , Homeodomain Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Smad2 Protein/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line , DNA, Complementary , Dogs , Female , Gene Library , Humans , Phosphorylation , Placenta/metabolism , Promyelocytic Leukemia Protein , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases
14.
Nephrol Dial Transplant ; 23(8): 2480-91, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18385385

ABSTRACT

BACKGROUND: Interstitial inflammation is a prominent feature associated with the severity of renal injury and progressive kidney failure. We utilized an animal model of aristolochic acid (AA)-induced nephropathy (AAN) to assess patterns of infiltration and inflammation during the evolution of tubulointerstitial damage and to relate them to the development of fibrosis. METHODS: Male Wistar rats receiving sc daily AA or vehicle were sacrificed between Days 1 and 35. Infiltrating mononuclear cells were characterized by immunohistochemistry. The kidney infiltrating T lymphocytes were phenotyped by flow cytometry. Urinary levels of Th-1/ Th-2 cytokines, of monocyte chemoattractant protein-1 and of active transforming growth factor-beta (TGF-beta) were measured. Tissue expression of phosphorylated smad 2/3 protein was used to examine the TGF-beta signalling pathway. RESULTS: In AA rats, monocytes/macrophages and T lymphocytes predominantly infiltrated areas of necrotic proximal tubular cells. The coexpressions of ED1 and/or Ki-67/MHCII by infiltrating cells reflected monocyte/macrophage proliferation and their activation, respectively. The accumulation of cytotoxic T lymphocytes was attested by severe signs of CD8+ cell tubulitis. The CD8/E-cadherin costaining confirmed intrarenal homing of CD8+CD103+ cells. Urinary levels of proinflammatory cytokines and of active TGF-beta significantly increased at Days 10 and 35. An early and persistent nuclear overexpression of phosphorylated smad 2/3 protein was detected in tubular and interstitial compartments. CONCLUSION: An early and massive interstitial inflammation characterized by activated monocytes/macrophages and cytotoxic CD8+CD103+ T lymphocytes is demonstrated for the first time during the progression of experimental AAN. The involvement in an interstitial fibrosis onset of active TGF-beta is highly suggested, at least via the psmad 2/3 intracellular signalling pathway.


Subject(s)
Aristolochic Acids/toxicity , Kidney/drug effects , Kidney/pathology , Animals , Disease Models, Animal , Fibrosis , Kidney/physiopathology , Kidney Failure, Chronic/chemically induced , Kidney Failure, Chronic/pathology , Kidney Failure, Chronic/physiopathology , Macrophages/immunology , Macrophages/pathology , Male , Monocytes/immunology , Monocytes/pathology , Rats , Rats, Wistar , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transforming Growth Factor beta/metabolism
15.
Mol Cell ; 23(4): 547-59, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16916642

ABSTRACT

The homeodomain protein TGIF has been implicated in the negative regulation of TGF-beta signaling. In this study, we report an unexpected role of TGIF in the inhibition of Smad2 phosphorylation, which occurs by a mechanism independent of its association with Smad2. This inhibitory function of TGIF is executed in concert with c-Jun, which facilitates the interaction of TGIF with cPML, resulting in the nuclear sequestration of cPML and the disruption of the cPML-SARA complex. Notably, knockdown of TGIF by siRNA caused increased association of cPML with SARA and cytoplasmic accumulation of cPML. Furthermore, c-Jun(-/-) fibroblasts exhibit enhanced association of cPML with SARA. c-Jun(-/-) fibroblasts also lose their sensitivity to TGIF-mediated disruption of the cPML-SARA complex and of nuclear sequestration of cPML. We suggest that the interaction of TGIF with cPML through c-Jun may negatively regulate TGF-beta signaling through controlling the localization of cPML and, consequently, the assembly of the cPML-SARA complex.


Subject(s)
Cell Nucleus/metabolism , Homeodomain Proteins/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Dogs , Down-Regulation/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Humans , Mice , Phosphorylation/drug effects , Promyelocytic Leukemia Protein , Protein Binding , Proto-Oncogene Proteins c-jun/deficiency , Proto-Oncogene Proteins c-jun/metabolism , Smad2 Protein/metabolism , Transcription, Genetic/drug effects
16.
Kidney Int ; 68(4): 1875-83, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16164666

ABSTRACT

BACKGROUND: Transforming growth factor-beta (TGF-beta) plays an important role in renal fibrosis. Measurement of the concentration of the active form of TGF-beta particularly in urine may help our understanding of the mechanism of chronic allograft nephropathy and could be used as a diagnostic tool. However, TGF-beta release and activation are complex and, consequently, there is currently no accurate way to measure TGF-beta activity. METHODS: TGF-beta-sensitive BL41 cells were stably transfected with a reporter plasmid harboring a synthetic TGF-beta-inducible DNA sequence upstream from the luciferase gene. Cells were incubated with urine samples from normal donors or transplanted recipients with or without patent nephropathy, and the active form of TGF-beta was determined as luciferase activity. RESULTS: We have established a cell line which expresses luciferase activity in response to active TGF-beta in a dose-dependent manner. Moreover, the use of a histone deacetylase inhibitor greatly increased sensitivity to TGF-beta and also stabilized luciferase inductibility. This test is highly specific to active TGF-beta. Detectable levels of TGF-beta were found in urine from patients with renal dysfunction due to acute or chronic renal allograft rejection (P < 0.001), but not in that from patients with stable, correctly functional kidneys. CONCLUSION: We describe a highly sensitive and specific assay for active TGF-beta. We also show that, in cases of renal allograft, TGF-beta expression is highly and significantly correlated with acute or chronic rejections.


Subject(s)
Biological Assay/methods , Graft Rejection/diagnosis , Graft Rejection/urine , Kidney Transplantation , Transforming Growth Factor beta/urine , Adult , Animals , Cell Line , Dogs , Enzyme Inhibitors/pharmacology , Evaluation Studies as Topic , Gene Expression/drug effects , Humans , Hydroxamic Acids/pharmacology , Kidney/cytology , Kidney Diseases/diagnosis , Kidney Diseases/urine , Luciferases/genetics , Middle Aged , Sensitivity and Specificity , Transfection , Transplantation, Homologous
17.
EMBO Rep ; 6(10): 985-91, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16113647

ABSTRACT

The fusion of Abl with either Bcr or Tel in human leukaemia leads to the constitutive activation of Abl tyrosine kinase, which in turn induces growth-factor-independent proliferation and cell survival. However, the mechanism by which Bcr-Abl induces cellular transformation has not yet been well characterized. Here, we show that Bcr-Abl-expressing cells are resistant to growth inhibition and apoptosis mediated by transforming growth factor-beta (TGF-beta). Interestingly, we observed that the suppressive effects of Bcr-Abl on TGF-beta responses were not mediated by an impairment of Smad signalling, which is believed to act as the principal mediator of TGF-beta responses. In contrast, we found that Bcr-Abl can target the protein kinase AKT and the transcription factor Fox O3 to interfere with growth inhibition and apoptosis in response to TGF-beta. Our results show a novel mechanism of cellular transformation by the oncogenic fusion protein Bcr-Abl through suppression of the cytostatic actions of TGF-beta.


Subject(s)
Forkhead Transcription Factors/physiology , Fusion Proteins, bcr-abl/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Animals , Apoptosis , Blotting, Western , Cell Line , Cell Proliferation , Fluorescent Antibody Technique , Humans , Mice , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Transforming Growth Factor beta/biosynthesis
18.
J Immunol ; 175(5): 2968-73, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16116183

ABSTRACT

EBV infects a large proportion of the human population worldwide and is one of the major viruses with human B lymphocyte tropism. It can immortalize human B lymphocytes and controls their resistance to apoptosis. EBV infection is associated with several lymphomas, including Burkitt's lymphoma. In this report we show that EBV infection leads to the post-transcriptional down-regulation of expression of the proapoptotic protein Bim. This process involves the phosphorylation of BimEL by the constitutive EBV-activated kinase ERK1/2, followed by its degradation through the proteasome pathway. We also show that ectopic expression of BimEL in EBV-positive Burkitt's lymphoma cells can enhance the sensitivity of these cells to serum deprivation-dependent apoptosis. Thus, EBV-mediated resistance to growth factor deprivation in human B lymphocytes is dependent on BimEL expression. Our data suggest that this regulatory pathway is an important contributor to the oncogenic potential of EBV.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis , B-Lymphocytes/virology , Gene Expression Regulation , Herpesvirus 4, Human/pathogenicity , Apoptosis Regulatory Proteins , B-Lymphocytes/pathology , Bcl-2-Like Protein 11 , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Membrane Proteins , Proto-Oncogene Proteins
19.
Oncogene ; 24(20): 3268-73, 2005 May 05.
Article in English | MEDLINE | ID: mdl-15782135

ABSTRACT

The cysteine protease caspase-8 plays a pivotal role in the initiation of different apoptotic pathways and controls the maturation and differentiation of various cell types including neurons, fibroblasts and lymphocytes. Specific substrates of caspase-8 are present in both the cytoplasm and the nucleus, which may determine the ultimate biological effect of caspase-8. However, the mechanisms regulating the cellular localization of caspase-8 are still unknown. We show here that, in contrast to other caspases such as caspase-9 and -3, caspase-8 can be sumoylated at lysine 156. This sumoylation (i) is associated with the nuclear localization of caspase-8 and (ii) did not impair caspase-8 activation.


Subject(s)
Caspases/metabolism , Cell Nucleus/metabolism , SUMO-1 Protein/physiology , Amino Acid Sequence , Anisomycin/pharmacology , Apoptosis , Blotting, Western , Caspase 3 , Caspase 8 , Caspase 9 , Cell Line, Tumor , Cysteine/metabolism , Cytoplasm/metabolism , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Fibroblasts/metabolism , Gene Library , Humans , Immunoprecipitation , Jurkat Cells , Lymphocytes/metabolism , Lysine/chemistry , Mass Spectrometry , Molecular Sequence Data , Neurons/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Structure, Tertiary , SUMO-1 Protein/metabolism , Subcellular Fractions/metabolism , Two-Hybrid System Techniques
20.
Oncogene ; 24(5): 737-45, 2005 Jan 27.
Article in English | MEDLINE | ID: mdl-15580299

ABSTRACT

Two cell lines that exemplify erythropoietin (EPO) receptor-positive tumors, human renal carcinoma cell lines RCC and the myelomonocytic leukemia cell line U937, were investigated for the apoptosis-modulatory potential of EPO. Cells cultured in the presence of EPO exhibited an elevated apoptotic response to cancer chemotherapeutic agents such as daunorubicin (Dauno) and vinblastine (VBL). Chemosensitization by EPO did not involve an increase in p53 activation, yet correlated with enhanced Bax/Bak-dependent mitochondrial membrane perturbation and caspase maturation. In vitro monotherapy with Dauno or VBL induced the degradation of IkappaBalpha, provoked the translocation of NF-kappaB p65/50 to the nucleus and stimulated the expression of an NF-kappaB-activatable reporter gene. All these signs of NF-kappaB activation were perturbed in the presence of EPO. Inhibition of JAK2, one of the receptor-proximal elements of EPO-mediated signal transduction, greatly diminished the EPO-mediated chemosensitization and NF-kappaB inhibition. EPO lost its death-facilitating effects in the presence of an NF-kappaB inhibitor, underscoring the cause-effect relationship between EPO-mediated chemosensitization and NF-kappaB inhibition. Altogether, these results suggest that, at least in a specific subset of tumors, EPO receptor agonists can prevent activation of the NF-kappaB pathway, thereby enhancing the propensity of EPO receptor-positive tumor cells to undergo apoptosis.


Subject(s)
Erythropoietin/pharmacology , NF-kappa B/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Renal Cell , Cell Death/drug effects , Cell Line, Tumor , Daunorubicin/pharmacology , Humans , Janus Kinase 2 , Kidney Neoplasms , Protein Transport/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Receptors, Erythropoietin/drug effects , Receptors, Erythropoietin/physiology , Signal Transduction/drug effects , U937 Cells , Vinblastine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...