Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Genes Dev ; 29(8): 817-31, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25846796

ABSTRACT

The bone microenvironment is composed of niches that house cells across variable oxygen tensions. However, the contribution of oxygen gradients in regulating bone and blood homeostasis remains unknown. Here, we generated mice with either single or combined genetic inactivation of the critical oxygen-sensing prolyl hydroxylase (PHD) enzymes (PHD1-3) in osteoprogenitors. Hypoxia-inducible factor (HIF) activation associated with Phd2 and Phd3 inactivation drove bone accumulation by modulating osteoblastic/osteoclastic cross-talk through the direct regulation of osteoprotegerin (OPG). In contrast, combined inactivation of Phd1, Phd2, and Phd3 resulted in extreme HIF signaling, leading to polycythemia and excessive bone accumulation by overstimulating angiogenic-osteogenic coupling. We also demonstrate that genetic ablation of Phd2 and Phd3 was sufficient to protect ovariectomized mice against bone loss without disrupting hematopoietic homeostasis. Importantly, we identify OPG as a HIF target gene capable of directing osteoblast-mediated osteoclastogenesis to regulate bone homeostasis. Here, we show that coordinated activation of specific PHD isoforms fine-tunes the osteoblastic response to hypoxia, thereby directing two important aspects of bone physiology: cross-talk between osteoblasts and osteoclasts and angiogenic-osteogenic coupling.


Subject(s)
Bone and Bones/enzymology , Homeostasis , Osteoprotegerin/metabolism , Oxygen/metabolism , Prolyl Hydroxylases/genetics , Prolyl Hydroxylases/metabolism , 3T3 Cells , Animals , Bone Resorption/genetics , Bone and Bones/cytology , Cell Communication , Cell Hypoxia/physiology , Cells, Cultured , Enzyme Activation , Female , Gene Silencing , Hypoxia-Inducible Factor 1/metabolism , Mice , Osteoblasts/metabolism , Osteoclasts/metabolism , Signal Transduction/genetics , Stem Cells/enzymology
2.
Nucleic Acids Res ; 45(7): 3674-3692, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28073943

ABSTRACT

The p53 tumor suppressor protein plays a critical role in orchestrating the genomic response to various stress signals by acting as a master transcriptional regulator. Differential gene activity is controlled by transcription factors but also dependent on the underlying chromatin structure, especially on covalent histone modifications. After screening different histone lysine methyltransferases and demethylases, we identified JMJD2B/KDM4B as a p53-inducible gene in response to DNA damage. p53 directly regulates JMJD2B gene expression by binding to a canonical p53-consensus motif in the JMJD2B promoter. JMJD2B induction attenuates the transcription of key p53 transcriptional targets including p21, PIG3 and PUMA, and this modulation is dependent on the catalytic capacity of JMJD2B. Conversely, JMJD2B silencing led to an enhancement of the DNA-damage driven induction of p21 and PIG3. These findings indicate that JMJD2B acts in an auto-regulatory loop by which p53, through JMJD2B activation, is able to influence its own transcriptional program. Functionally, exogenous expression of JMJD2B enhanced subcutaneous tumor growth of colon cancer cells in a p53-dependent manner, and genetic inhibition of JMJD2B impaired tumor growth in vivo. These studies provide new insights into the regulatory effect exerted by JMJD2B on tumor growth through the modulation of p53 target genes.


Subject(s)
DNA Damage , Epigenesis, Genetic , Jumonji Domain-Containing Histone Demethylases/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Cells, Cultured , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/biosynthesis , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mutagens/toxicity , Neoplasms/pathology , Promoter Regions, Genetic , Transcriptional Activation
3.
Proc Natl Acad Sci U S A ; 112(14): 4441-6, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25792458

ABSTRACT

Scaffold proteins are critical hubs within cells that have the ability to modulate upstream signaling molecules and their downstream effectors to fine-tune biological responses. Although they can serve as focal points for association of signaling molecules and downstream pathways that regulate tumorigenesis, little is known about how the tumor microenvironment affects the expression and activity of scaffold proteins. This study demonstrates that hypoxia, a common element of solid tumors harboring low oxygen levels, regulates expression of a specific variant of the scaffold protein AKAP12 (A-kinase anchor protein 12), AKAP12v2, in metastatic melanoma. In turn, through a kinome-wide phosphoproteomic and MS study, we demonstrate that this scaffolding protein regulates a shift in protein kinase A (PKA)-mediated phosphorylation events under hypoxia, causing alterations in tumor cell invasion and migration in vitro, as well as metastasis in an in vivo orthotopic model of melanoma. Mechanistically, the shift in AKAP12-dependent PKA-mediated phosphorylations under hypoxia is due to changes in AKAP12 localization vs. structural differences between its two variants. Importantly, our work defines a mechanism through which a scaffold protein can be regulated by the tumor microenvironment and further explains how a tumor cell can coordinate many critical signaling pathways that are essential for tumor growth through one individual scaffolding protein.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Cycle Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/pathology , Skin Neoplasms/metabolism , A Kinase Anchor Proteins/genetics , Amino Acid Sequence , Animals , Cell Cycle Proteins/genetics , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Humans , Melanoma/metabolism , Mice , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Oxygen/metabolism , Phosphoproteins/metabolism , Phosphorylation , Proteomics , Signal Transduction
5.
Proc Natl Acad Sci U S A ; 111(37): 13373-8, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25187556

ABSTRACT

Dysregulation of the von Hippel-Lindau/hypoxia-inducible transcription factor (HIF) signaling pathway promotes clear cell renal cell carcinoma (ccRCC) progression and metastasis. The protein kinase GAS6/AXL signaling pathway has recently been implicated as an essential mediator of metastasis and receptor tyrosine kinase crosstalk in cancer. Here we establish a molecular link between HIF stabilization and induction of AXL receptor expression in metastatic ccRCC. We found that HIF-1 and HIF-2 directly activate the expression of AXL by binding to the hypoxia-response element in the AXL proximal promoter. Importantly, genetic and therapeutic inactivation of AXL signaling in metastatic ccRCC cells reversed the invasive and metastatic phenotype in vivo. Furthermore, we define a pathway by which GAS6/AXL signaling uses lateral activation of the met proto-oncogene (MET) through SRC proto-oncogene nonreceptor tyrosine kinase to maximize cellular invasion. Clinically, AXL expression in primary tumors of ccRCC patients correlates with aggressive tumor behavior and patient lethality. These findings provide an alternative model for SRC and MET activation by growth arrest-specific 6 in ccRCC and identify AXL as a therapeutic target driving the aggressive phenotype in renal clear cell carcinoma.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Neoplasms/secondary , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , src-Family Kinases/metabolism , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/secondary , Cell Hypoxia , Cell Line, Tumor , Enzyme Activation , Hepatocyte Growth Factor/pharmacology , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/pathology , Models, Biological , Neoplasm Invasiveness , Phenotype , Proto-Oncogene Mas , Signal Transduction , Treatment Outcome , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Axl Receptor Tyrosine Kinase
6.
Cytotherapy ; 13(3): 329-40, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20849362

ABSTRACT

BACKGROUND AIMS: Bone marrow- and adipose tissue-derived mesenchymal stromal cells (MSC) represent promising sources for regenerative medicine. However, the precise molecular mechanisms underlying MSC stemness maintenance versus differentiation are not fully understood. The aim of this study was to compare the genome-wide expression profiles of bone marrow-and adipose tissue-derived MSC, in order to identify a common molecular stemness core. METHODS: Molecular profiling was carried out using Affymetrix microarray and relevant genes were further validated by Q-PCR. RESULTS: We identified an overlapping dataset of 190 transcripts commonly regulated in both cell populations, which included several genes involved in stemness regulation (i.e. self-renewal potential and the ability to generate differentiated cells), various signaling pathways and transcription factors. In particular, we identified a central role of the Kruppel-like factor 4 (KLF4) DNA-binding protein in regulating MSC transcriptional activity. CONCLUSIONS: Our results provide new insights toward understanding the molecular basis of MSC stemness maintenance and underline the ability of KLF4 to maintain cells in an undifferentiated state.


Subject(s)
Adipose Tissue/cytology , Bone Marrow Cells/metabolism , Cell Lineage/genetics , Gene Expression Profiling , Kruppel-Like Transcription Factors/metabolism , Mesenchymal Stem Cells/metabolism , Adult , Binding Sites , Bone Marrow Cells/cytology , Cell Differentiation/genetics , Chromatin Immunoprecipitation , Female , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Male , Mesenchymal Stem Cells/cytology , Middle Aged , Models, Biological , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Protein Binding , Reproducibility of Results , Stromal Cells/cytology , Stromal Cells/metabolism , Young Adult
7.
Antioxidants (Basel) ; 10(12)2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34943115

ABSTRACT

Free radicals play a role of paramount importance in the development of neonatal brain injury. Depending on the pathophysiological mechanisms underlying free radical overproduction and upon specific neonatal characteristics, such as the GA-dependent maturation of antioxidant defenses and of cerebrovascular autoregulation, different profiles of injury have been identified. The growing evidence on the detrimental effects of free radicals on the brain tissue has led to discover not only potential biomarkers for oxidative damage, but also possible neuroprotective therapeutic approaches targeting oxidative stress. While a more extensive validation of free radical biomarkers is required before considering their use in routine neonatal practice, two important treatments endowed with antioxidant properties, such as therapeutic hypothermia and magnesium sulfate, have become part of the standard of care to reduce the risk of neonatal brain injury, and other promising therapeutic strategies are being tested in clinical trials. The implementation of currently available evidence is crucial to optimize neonatal neuroprotection and to develop individualized diagnostic and therapeutic approaches addressing oxidative brain injury, with the final aim of improving the neurological outcome of this population.

8.
Nat Commun ; 12(1): 4308, 2021 07 14.
Article in English | MEDLINE | ID: mdl-34262028

ABSTRACT

Hypoxia plays a critical role in tumor progression including invasion and metastasis. To determine critical genes regulated by hypoxia that promote invasion and metastasis, we screen fifty hypoxia inducible genes for their effects on invasion. In this study, we identify v-maf musculoaponeurotic fibrosarcoma oncogene homolog F (MAFF) as a potent regulator of tumor invasion without affecting cell viability. MAFF expression is elevated in metastatic breast cancer patients and is specifically correlated with hypoxic tumors. Combined ChIP- and RNA-sequencing identifies IL11 as a direct transcriptional target of the heterodimer between MAFF and BACH1, which leads to activation of STAT3 signaling. Inhibition of IL11 results in similar levels of metastatic suppression as inhibition of MAFF. This study demonstrates the oncogenic role of MAFF as an activator of the IL11/STAT3 pathways in breast cancer.


Subject(s)
Breast Neoplasms/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-11/metabolism , MafF Transcription Factor/metabolism , Nuclear Proteins/metabolism , STAT3 Transcription Factor/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Hypoxia , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , MafF Transcription Factor/genetics , Mice , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Nuclear Proteins/genetics , Prognosis , Signal Transduction , Transcription, Genetic
9.
Clin Cancer Res ; 26(12): 2972-2985, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32098769

ABSTRACT

PURPOSE: To investigate how induced tumor heterogeneity influences immune responses to radiotherapy with different proportions of mixed immune-responsive and unresponsive tumor cells in a triple-negative breast cancer model. It is hypothesized that studying the immune environment of mixed tumors and responses to radiotherapy could nominate immune active therapies to enhance immune responses after radiotherapy. EXPERIMENTAL DESIGN: Evaluate efficacy and immune responses generated by radiotherapy in tumors with different proportions of immunologically responsive and unresponsive tumor cells. Then study the cellular responses and transcriptomic differences between the tumors to nominate immunotherapy combinations with radiotherapy and evaluate the combination. RESULTS: The addition of the responsive cells to unresponsive tumors led to a greater than expected therapeutic response to radiotherapy with both innate and adaptive immune components. There was a distinct change in myeloid cells, greater inflammatory macrophage activity, and enhanced antigen presentation with responsive cells after radiotherapy. Because differences in matrix components, cell adhesion biology, and innate immune signaling correlated with myeloid cell response and phenotype, we hypothesized that radiotherapy combined with CD40 agonist antibody would sensitize unresponsive tumors. The combination therapy resulted in improved innate and adaptive immune response. Importantly, CD40 treatment increased tumor response to radiotherapy and protected against metastatic spread in a metastatic model. CONCLUSIONS: These data combined with transcriptomics from human patients support radiotherapy and myeloid cell targeting for immunologically cold tumors. The established study model presents opportunities to investigate the complex overlapping biologic mechanisms that limit immunotherapy and to implement radiotherapy with different immunotherapy combinations.


Subject(s)
Breast Neoplasms/pathology , Immunotherapy/mortality , Radioimmunotherapy/mortality , Radiotherapy/mortality , Animals , Apoptosis , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Cell Proliferation , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Bone Res ; 7: 7, 2019.
Article in English | MEDLINE | ID: mdl-30792937

ABSTRACT

Osteoblasts, which are the bone-forming cells, operate in a hypoxic environment. The transcription factors hypoxia-inducible factor-1α (HIF1) and HIF2 are key mediators of the cellular response to hypoxia. Both are expressed in osteoblasts. HIF1 is known to be a positive regulator of bone formation. Conversely, the role of HIF2 in the control osteoblast biology is still poorly understood. In this study, we used mouse genetics to demonstrate that HIF2 is an inhibitor of osteoblastogenesis and bone mass accrual. Moreover, we provided evidence that HIF2 impairs osteoblast differentiation at least in part, by upregulating the transcription factor Sox9. Our findings constitute a paradigm shift, as activation of the hypoxia-signaling pathway has traditionally been associated with increased bone formation through HIF1. Inhibiting HIF2 could thus represent a therapeutic approach for the treatment of the low bone mass observed in chronic diseases, osteoporosis, or aging.

11.
J Neurochem ; 107(3): 871-82, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18786178

ABSTRACT

We recently reported that bcl-xL regulates interleukin 8 (CXCL8) protein expression and promoter activity in glioblastoma cells. In this paper we demonstrate that CXCL8 induction by bcl-xL is mediated through a nuclear factor-kappa B (NF-kB)-dependent mechanism. Mutational studies on the CXCL8 promoter showed that NF-kB binding site was required for bcl-xL-induced promoter activity and an enhanced nuclear expression of NF-kB subunits p65 and p50 was observed after bcl-xL over-expression. Electrophoretic mobility shift assay showed an increased DNA-binding activity of NF-kB in bcl-xL over-expressing cells and the use of specific antibodies confirmed the involvement of p65 and p50 in NF-kB activity on CXCL8 promoter sequence. NF-kB activity regulation by bcl-xL involved IkBalpha and IKK complex signaling pathway. In fact, bcl-xL over-expression induced a decrease of cytoplasmic expression of the IkBalpha protein, paralleled by an increase in the phosphorylation of the same IkBalpha and IKKalpha/beta. Moreover, the down-regulation of the ectopic or endogenous bcl-xL expression through RNA interference confirmed the ability of bcl-xL to modulate NF-kB pathway, and the transient expression of a degradation-resistant form of the cytoplasmic NF-kB inhibitor IkBalpha in bcl-xL transfectants confirmed the involvement of that inhibitor in bcl-xL-induced CXCL8 expression and promoter activity. In conclusion, our results demonstrate the role of NF-kB as the mediator of bcl-xL-induced CXCL8 up-regulation in glioblastoma cells.


Subject(s)
Gene Expression Regulation/physiology , Glioblastoma/metabolism , Interleukin-8/biosynthesis , NF-kappa B/metabolism , bcl-X Protein/metabolism , Blotting, Western , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-8/genetics , Neovascularization, Pathologic/metabolism , Promoter Regions, Genetic , Signal Transduction/physiology , Transfection , Up-Regulation
12.
Mol Cancer Res ; 5(8): 761-71, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17699103

ABSTRACT

In this paper, we investigated whether bcl-xL can be involved in the modulation of the angiogenic phenotype of human tumor cells. Using the ADF human glioblastoma and the M14 melanoma lines, and their derivative bcl-xL-overexpressing clones, we showed that the conditioned medium of bcl-xL transfectants increased in vitro endothelial cell functions, such as proliferation and morphogenesis, and in vivo vessel formation in Matrigel plugs, compared with the conditioned medium of control cells. Moreover, the overexpression of bcl-xL induced an increased expression of the proangiogenic interleukin-8 (CXCL8), both at the protein and mRNA levels, and an enhanced CXCL8 promoter activity. The role of CXCL8 on bcl-xL-induced angiogenesis was validated using CXCL8-neutralizing antibodies, whereas down-regulation of bcl-xL through antisense oligonucleotide or RNA interference strategies confirmed the involvement of bcl-xL on CXCL8 expression. Transient overexpression of bcl-xL led to extend this observation to other tumor cell lines with different origin, such as colon and prostate carcinoma. In conclusion, our results showed that CXCL8 modulation by bcl-xL regulates tumor angiogenesis, and they point to elucidate an additional function of bcl-xL protein.


Subject(s)
Glioblastoma/blood supply , Interleukin-8/metabolism , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/metabolism , Oligonucleotides, Antisense/pharmacology , bcl-X Protein/metabolism , Biomarkers, Tumor/metabolism , Blotting, Northern , Blotting, Western , Cells, Cultured , Collagen , Drug Combinations , Endothelium, Vascular , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Interleukin-8/genetics , Laminin , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Promoter Regions, Genetic , Protein Array Analysis , Proteoglycans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Umbilical Veins , bcl-X Protein/genetics
13.
Cancer Discov ; 7(10): 1184-1199, 2017 10.
Article in English | MEDLINE | ID: mdl-28790031

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most metastatic and deadly cancers. Despite the clinical significance of metastatic spread, our understanding of molecular mechanisms that drive PDAC metastatic ability remains limited. By generating a genetically engineered mouse model of human PDAC, we uncover a transient subpopulation of cancer cells with exceptionally high metastatic ability. Global gene expression profiling and functional analyses uncovered the transcription factor BLIMP1 as a driver of PDAC metastasis. The highly metastatic PDAC subpopulation is enriched for hypoxia-induced genes, and hypoxia-mediated induction of BLIMP1 contributes to the regulation of a subset of hypoxia-associated gene expression programs. These findings support a model in which upregulation of BLIMP1 links microenvironmental cues to a metastatic stem cell character.Significance: PDAC is an almost uniformly lethal cancer, largely due to its tendency for metastasis. We define a highly metastatic subpopulation of cancer cells, uncover a key transcriptional regulator of metastatic ability, and define hypoxia as an important factor within the tumor microenvironment that increases metastatic proclivity. Cancer Discov; 7(10); 1184-99. ©2017 AACR.See related commentary by Vakoc and Tuveson, p. 1067This article is highlighted in the In This Issue feature, p. 1047.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Gene Expression Profiling/methods , Pancreatic Neoplasms/pathology , Positive Regulatory Domain I-Binding Factor 1/genetics , Sequence Analysis, RNA/methods , Up-Regulation , Animals , Carcinoma, Pancreatic Ductal/genetics , Cell Hypoxia , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genetic Engineering , Humans , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Tumor Microenvironment
14.
Nat Commun ; 7: 13898, 2016 12 23.
Article in English | MEDLINE | ID: mdl-28008921

ABSTRACT

Increasing evidence suggests that ionizing radiation therapy (RT) in combination with checkpoint immunotherapy is highly effective in treating a subset of cancers. To better understand the limited responses to this combination we analysed the genetic, microenvironmental, and immune factors in tumours derived from a transgenic breast cancer model. We identified two tumours with similar growth characteristics but different RT responses primarily due to an antitumour immune response. The combination of RT and checkpoint immunotherapy resulted in cures in the responsive but not the unresponsive tumours. Profiling the tumours revealed that the Axl receptor tyrosine kinase is overexpressed in the unresponsive tumours, and Axl knockout resulted in slower growth and increased radiosensitivity. These changes were associated with a CD8+ T-cell response, which was improved in combination with checkpoint immunotherapy. These results suggest a novel role for Axl in suppressing antigen presentation through MHCI, and enhancing cytokine release, which promotes a suppressive myeloid microenvironment.


Subject(s)
Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/immunology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects , Animals , Cell Line, Tumor , Cell Proliferation , Combined Modality Therapy , Cytokines/metabolism , Histocompatibility Antigens Class I/metabolism , Immunity , Immunosuppression Therapy , Immunotherapy , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Radiation Tolerance/radiation effects , Axl Receptor Tyrosine Kinase
15.
Sci Rep ; 5: 9149, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25779766

ABSTRACT

Although the only effective drug against primary hepatocarcinoma, the multikinase inhibitor Sorafenib (SFB) usually fails to eradicate liver cancer. Since SFB targets mitochondria, cell metabolic reprogramming may underlie intrinsic tumor resistance. To characterize cancer cell metabolic response to SFB, we measured oxygen consumption, generation of reactive oxygen species (ROS) and ATP content in rat LCSC (Liver Cancer Stem Cells) -2 cells exposed to the drug. Genome wide analysis of gene expression was performed by Affymetrix technology. SFB cytotoxicity was evaluated by multiple assays in the presence or absence of metabolic inhibitors, or in cells genetically depleted of mitochondria. We found that low concentrations (2.5-5 µM) of SFB had a relatively modest effect on LCSC-2 or 293 T cell growth, but damaged mitochondria and increased intracellular ROS. Gene expression profiling of SFB-treated cells was consistent with a shift toward aerobic glycolysis and, accordingly, SFB cytotoxicity was dramatically increased by glucose withdrawal or the glycolytic inhibitor 2-DG. Under metabolic stress, activation of the AMP dependent Protein Kinase (AMPK), but not ROS blockade, protected cells from death. We conclude that mitochondrial damage and ROS drive cell killing by SFB, while glycolytic cell reprogramming may represent a resistance strategy potentially targetable by combination therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy , Cell Line, Tumor , Cell Respiration/drug effects , Cell Survival/drug effects , Deoxyglucose/pharmacology , Energy Metabolism/drug effects , Glycolysis/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Niacinamide/pharmacology , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sorafenib , TOR Serine-Threonine Kinases/metabolism
16.
Mol Cell Biol ; 34(4): 739-51, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24344197

ABSTRACT

The induction of hypoxia-inducible factors (HIFs) is essential for the adaptation of tumor cells to a low-oxygen environment. We found that the expression of the apoptosis inhibitor ARC (apoptosis repressor with a CARD domain) was induced by hypoxia in a variety of cancer cell types, and its induction is primarily HIF1 dependent. Chromatin immunoprecipitation (ChIP) and reporter assays also indicate that the ARC gene is regulated by direct binding of HIF1 to a hypoxia response element (HRE) located at bp -190 upstream of the transcription start site. HIFs play an essential role in the pathogenesis of renal cell carcinoma (RCC) under normoxic conditions, through the loss of the Von Hippel-Lindau gene (VHL). Accordingly, our results show that ARC is not expressed in normal renal tissue but is highly expressed in 65% of RCC tumors, which also express high levels of carbonic anhydrase IX (CAIX), a HIF1-dependent protein. Compared to controls, ARC-deficient RCCs exhibited decreased colony formation and increased apoptosis in vitro. In addition, loss of ARC resulted in a dramatic reduction of RCC tumor growth in SCID mice in vivo. Thus, HIF-mediated increased expression of ARC in RCC can explain how loss of VHL can promote survival early in tumor formation.


Subject(s)
Apoptosis/physiology , Carcinoma, Renal Cell/metabolism , Gene Expression Regulation, Neoplastic/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney Neoplasms/metabolism , Tumor Suppressor Proteins/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , AIDS-Related Complex/genetics , Animals , Apoptosis/genetics , Carcinoma, Renal Cell/genetics , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Proliferation , Cell Survival , Cells, Cultured , Gene Expression Regulation, Neoplastic/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mice , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
17.
Vet Microbiol ; 149(3-4): 330-8, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21216541

ABSTRACT

Hepatitis E is an acute disease of humans caused by a small RNA virus, Hepatitis E virus (HEV). In recent years, an increasing number of autochthonous human infections have been reported in industrialized countries. Genotype 3 is the main HEV type circulating in swine, and is also reported in sporadic cases of hepatitis E in humans worldwide. To date one serotype has been described. We have conducted a survey to detect antibodies against HEV in 48 swine at a slaughterhouse in Northern Italy, using ELISA test. Mean seroprevalence in the studied animal group was 87.0%. Bile, liver and feces from the 48 animals were also collected, and HEV RNA was detected by nested reverse transcription-polymerase chain reaction, amplifying a fragment of the ORF2. HEV genome was most frequently detected in bile samples (51.1%), followed by feces (33.3%) and liver (20.8%). Thirty-one out of 48 studied pigs (64.6%) were positive for HEV RNA in at least one sample. Overall, HEV RNA was found at a statistically higher rate in the 3-4-month-old than in 9-10-month-old animals (95.0% vs. 42.9%). Genetic characterization of swine strains identified was performed by sequencing and database alignment. Phylogenetic analysis on the nucleotide sequences from 14 positive PCR products indicated that all strains belonged to genotype 3, clustering in two branches subtypes g3c and g3f.


Subject(s)
Hepatitis Antibodies/blood , Hepatitis E virus/classification , Hepatitis E/veterinary , Swine Diseases/virology , Swine/virology , Abattoirs , Animals , Bile/virology , Enzyme-Linked Immunosorbent Assay , Feces/virology , Genotype , Hepatitis E/epidemiology , Hepatitis E/virology , Hepatitis E virus/genetics , Italy/epidemiology , Liver/virology , Phylogeny , Polymerase Chain Reaction , Prevalence , Seroepidemiologic Studies , Swine/blood , Swine Diseases/epidemiology
18.
Biochem Biophys Res Commun ; 303(3): 926-31, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12670500

ABSTRACT

Styrene is an important chemical extensively used in the petrochemical and polymer industries. In Pseudomonas fluorescens ST, styrene metabolism is controlled by a two-component regulatory system, very uncommon in the degradation of aromatic compounds. The two-component regulatory proteins StyS and StyR regulate the expression of the styABCD operon, which codes for styrene degradation. StyS corresponds to the sensor kinase and StyR to the response regulator, which is essential for the activation of PstyA, the promoter of the catabolic operon. In two-component systems, the response regulator is phosphorylated by the cognate sensor kinase. Phosphorylation activates the response regulator, inducing DNA-binding. The mechanism underlying this activation has been reported only for a very few response regulators. Here, the effect of phosphorylation on the oligomeric state and on the DNA-binding properties of StyR has been investigated. Phosphorylation induces dimerization of StyR, the affinity of dimeric StyR for the target DNA is higher than that of the monomer, moreover dimeric StyR binding to the DNA target is cooperative. Furthermore, StyR oligomerization may be driven by the DNA target. This is the first direct demonstration that StyR response regulator binds to the PstyA promoter.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , DNA, Bacterial/metabolism , Pseudomonas fluorescens/metabolism , Styrene/metabolism , Bacterial Proteins/genetics , Base Sequence , Biodegradation, Environmental , DNA, Bacterial/genetics , Dimerization , Environmental Pollutants/metabolism , Genes, Bacterial , Operon , Phosphorylation , Protein Structure, Quaternary , Pseudomonas fluorescens/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL