Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
iScience ; 27(6): 109737, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38799564

ABSTRACT

Long-chain acyl-CoA synthetase family 4 (ACSL4) metabolizes long-chain polyunsaturated fatty acids (PUFAs), enriching cell membranes with phospholipids susceptible to peroxidation and drive ferroptosis. The role of ACSL4 and ferroptosis upon endoplasmic-reticulum (ER)-stress-induced acute kidney injury (AKI) is unknown. We used lipidomic, molecular, and cellular biology approaches along with a mouse model of AKI induced by ER stress to investigate the role of ACSL4 regulation in membrane lipidome remodeling in the injured tubular epithelium. Tubular epithelial cells (TECs) activate ACSL4 in response to STAT3 signaling. In this context, TEC membrane lipidome is remodeled toward PUFA-enriched triglycerides instead of PUFA-bearing phospholipids. TECs expressing ACSL4 in this setting are not vulnerable to ferroptosis. Thus, ACSL4 activity in TECs is driven by STAT3 signaling, but ACSL4 alone is not enough to sensitize ferroptosis, highlighting the significance of the biological context associated with the study model.

2.
JCI Insight ; 7(18)2022 09 22.
Article in English | MEDLINE | ID: mdl-35998043

ABSTRACT

Energy metabolism failure in proximal tubule cells (PTCs) is a hallmark of chronic kidney injury. We combined transcriptomic, metabolomic, and lipidomic approaches in experimental models and patient cohorts to investigate the molecular basis of the progression to chronic kidney allograft injury initiated by ischemia/reperfusion injury (IRI). The urinary metabolome of kidney transplant recipients with chronic allograft injury and who experienced severe IRI was substantially enriched with long chain fatty acids (FAs). We identified a renal FA-related gene signature with low levels of carnitine palmitoyltransferase 2 (Cpt2) and acyl-CoA synthetase medium chain family member 5 (Acsm5) and high levels of acyl-CoA synthetase long chain family member 4 and 5 (Acsl4 and Acsl5) associated with IRI, transition to chronic injury, and established chronic kidney disease in mouse models and kidney transplant recipients. The findings were consistent with the presence of Cpt2-Acsl4+Acsl5+Acsm5- PTCs failing to recover from IRI as identified by single-nucleus RNA-Seq. In vitro experiments indicated that ER stress contributed to CPT2 repression, which, in turn, promoted lipids' accumulation, drove profibrogenic epithelial phenotypic changes, and activated the unfolded protein response. ER stress through CPT2 inhibition and lipid accumulation engaged an auto-amplification loop leading to lipotoxicity and self-sustained cellular stress. Thus, IRI imprints a persistent FA metabolism disturbance in the proximal tubule, sustaining the progression to chronic kidney allograft injury.


Subject(s)
Carnitine O-Palmitoyltransferase , Kidney , Animals , Carnitine O-Palmitoyltransferase/genetics , Coenzyme A , Fatty Acids/metabolism , Kidney/metabolism , Ligases , Mice
3.
JCI Insight ; 7(1)2022 01 11.
Article in English | MEDLINE | ID: mdl-34793337

ABSTRACT

The biosynthetic routes leading to de novo nicotinamide adenine dinucleotide (NAD+) production are involved in acute kidney injury (AKI), with a critical role for quinolinate phosphoribosyl transferase (QPRT), a bottleneck enzyme of de novo NAD+ biosynthesis. The molecular mechanisms determining reduced QPRT in AKI, and the role of impaired NAD+ biosynthesis in the progression to chronic kidney disease (CKD), are unknown. We demonstrate that a high urinary quinolinate-to-tryptophan ratio, an indirect indicator of impaired QPRT activity and reduced de novo NAD+ biosynthesis in the kidney, is a clinically applicable early marker of AKI after cardiac surgery and is predictive of progression to CKD in kidney transplant recipients. We also provide evidence that the endoplasmic reticulum (ER) stress response may impair de novo NAD+ biosynthesis by repressing QPRT transcription. In conclusion, NAD+ biosynthesis impairment is an early event in AKI embedded with the ER stress response, and persistent reduction of QPRT expression is associated with AKI to CKD progression. This finding may lead to identification of noninvasive metabolic biomarkers of kidney injury with prognostic and therapeutic implications.


Subject(s)
Acute Kidney Injury/metabolism , Endoplasmic Reticulum Stress/physiology , Kidney/metabolism , NAD/biosynthesis , Animals , Cell Line , Male , Mice , Mice, Inbred C57BL , Pentosyltransferases/metabolism , Quinolinic Acid/urine , Tryptophan/urine
4.
Cell Mol Life Sci ; 78(5): 2157-2167, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32875355

ABSTRACT

Inherited fatty acid oxidation diseases in their mild forms often present as metabolic myopathies. Carnitine Palmitoyl Transferase 2 (CPT2) deficiency, one such prototypical disorder is associated with compromised myotube differentiation. Here, we show that CPT2-deficient myotubes exhibit defects in focal adhesions and redox balance, exemplified by increased SOD2 expression. We document unprecedented alterations in the cellular prion protein PrPC, which directly arise from the failure in CPT2 enzymatic activity. We also demonstrate that the loss of PrPC function in normal myotubes recapitulates the defects in focal adhesion, redox balance and differentiation hallmarks monitored in CPT2-deficient cells. These results are further corroborated by studies performed in muscles from Prnp-/- mice. Altogether, our results unveil a molecular scenario, whereby PrPC dysfunction governed by faulty CPT2 activity may drive aberrant focal adhesion turnover and hinder proper myotube differentiation. Our study adds a novel facet to the involvement of PrPC in diverse physiopathological situations.


Subject(s)
Carnitine O-Palmitoyltransferase/genetics , Focal Adhesions/genetics , Muscle Fibers, Skeletal/metabolism , Muscular Diseases/genetics , Prion Proteins/genetics , Animals , Carnitine O-Palmitoyltransferase/deficiency , Cells, Cultured , Focal Adhesions/metabolism , Humans , Mice, Knockout , Muscle Fibers, Skeletal/cytology , Muscular Diseases/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Oxidation-Reduction , Prion Diseases/genetics , Prion Diseases/metabolism , Prion Proteins/deficiency , RNA Interference , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
5.
Biochem J ; 477(20): 4037-4051, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33016323

ABSTRACT

Renal epithelial cells regulate the destructive activity of macrophages and participate in the progression of kidney diseases. Critically, the Unfolded Protein Response (UPR), which is activated in renal epithelial cells in the course of kidney injury, is required for the optimal differentiation and activation of macrophages. Given that macrophages are key regulators of renal inflammation and fibrosis, we suppose that the identification of mediators that are released by renal epithelial cells under Endoplasmic Reticulum (ER) stress and transmitted to macrophages is a critical issue to address. Signals leading to a paracrine transmission of ER stress (TERS) from a donor cell to a recipient cells could be of paramount importance to understand how ER-stressed cells shape the immune microenvironment. Critically, the vast majority of studies that have examined TERS used thaspigargin as an inducer of ER stress in donor cells in cellular models. By using multiple sources of ER stress, we evaluated if human renal epithelial cells undergoing ER stress can transmit the UPR to human monocyte-derived macrophages and if such TERS can modulate the inflammatory profiles of these cells. Our results indicate that carry-over of thapsigargin is a confounding factor in chemically based TERS protocols classically used to induce ER Stress in donor cells. Hence, such protocols are not suitable to study the TERS phenomenon and to identify its mediators. In addition, the absence of TERS transmission in more physiological models of ER stress indicates that cell-to-cell UPR transmission is not a universal feature in cultured cells.


Subject(s)
Culture Media, Conditioned/pharmacology , Endoplasmic Reticulum Stress/drug effects , Epithelial Cells/drug effects , Macrophages/drug effects , Paracrine Communication/drug effects , Thapsigargin/pharmacology , Unfolded Protein Response/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/metabolism , Endoplasmic Reticulum Stress/genetics , Epithelial Cells/metabolism , Humans , Kidney/drug effects , Kidney/metabolism , Macrophages/metabolism , Mass Spectrometry , Paracrine Communication/genetics , Unfolded Protein Response/genetics
6.
Cell Death Dis ; 11(4): 243, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32303684

ABSTRACT

Endoplasmic Reticulum (ER) stress underlies the pathogenesis of numerous kidney diseases. A better care of patients with kidney disease involves the identification and validation of ER stress biomarkers in the early stages of kidney disease. For the first time to our knowledge, we demonstrate that the prion protein PrPC is secreted in a conventional manner by ER-stressed renal epithelial cell under the control of the transcription factor x-box binding protein 1 (XBP1) and can serve as a sensitive urinary biomarker for detecting tubular ER stress. Urinary PrPC elevation occurs in patients with chronic kidney disease. In addition, in patients undergoing cardiac surgery, detectable urine levels of PrPC significantly increase after cardiopulmonary bypass, a condition associated with activation of the IRE1-XBP1 pathway in the kidney. In conclusion, our study has identified PrPC as a novel urinary ER stress biomarker with potential utility in early diagnosis of ongoing acute or chronic kidney injury.


Subject(s)
Biomarkers/urine , Endoplasmic Reticulum Stress/physiology , Kidney Diseases/metabolism , Kidney Diseases/urine , Kidney/injuries , Prion Proteins/metabolism , Animals , Cell Proliferation , Humans , Kidney Diseases/pathology , Male , Mice
7.
EBioMedicine ; 46: 94-104, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31377347

ABSTRACT

BACKGROUND: Comprehensive transcriptomic analyses have shown that colorectal cancer (CRC) is heterogeneous and have led to the definition of molecular subtypes among which the stem-cell, mesenchymal-like group is associated with poor prognosis. The molecular pathways orchestrating the emergence of this subtype are incompletely understood. In line with the contribution of the cellular prion protein PrPC to stemness, we hypothesize that deregulation of this protein could lead to a stem-cell, mesenchymal-like phenotype in CRC. METHODS: We assessed the distribution of the PrPC-encoding PRNP mRNA in two large CRC cohorts according to molecular classification and its association with patient survival. We developed cell-based assays to explore the impact of gain and loss of PrPC function on markers of the mesenchymal subtype and to delineate the signalling pathways recruited by PrPC. We measured soluble PrPC in the plasmas of 325 patients with metastatic CRC and probed associations with disease outcome. FINDINGS: We found that PRNP gene expression is enriched in tumours of the mesenchymal subtype and is associated with poor survival. Our in vitro analyses revealed that PrPC controls the expression of genes that specify the mesenchymal subtype through the recruitment of the Hippo pathway effectors YAP and TAZ and the TGFß pathway. We showed that plasma levels of PrPC are elevated in metastatic CRC and are associated with poor disease control. INTERPRETATION: Our findings define PrPC as a candidate driver of the poor-prognosis mesenchymal subtype of CRC. They suggest that PrPC may serve as a potential biomarker for patient stratification in CRC. FUNDING: Grant support was provided by the following: Cancéropôle Ile de France (grant number 2016-1-EMERG-36-UP 5-1), Association pour la Recherche sur le Cancer (grant number PJA 20171206220), SATT Ile de France Innov (grant number 415) as well as INSERM.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Prion Proteins/genetics , Adult , Aged , Aged, 80 and over , Animals , Biomarkers , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Female , Gene Expression , Hippo Signaling Pathway , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prion Proteins/metabolism , Prognosis , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , Signal Transduction , Transforming Growth Factor beta/metabolism
8.
Cell Cycle ; 17(15): 1901-1916, 2018.
Article in English | MEDLINE | ID: mdl-30109813

ABSTRACT

Adaptation to endoplasmic reticulum (ER) stress depends on the activation of the sensor inositol-requiring enzyme 1α (IRE1), an endoribonuclease that splices the mRNA of the transcription factor XBP1 (X-box-binding protein 1). To better understand the protein network that regulates the activity of the IRE1 pathway, we systematically screened the proteins that interact with IRE1 and identified a ribonuclease inhibitor called ribonuclease/angiogenin inhibitor 1 (RNH1). RNH1 is a leucine-rich repeat domains-containing protein that binds to and inhibits ribonucleases. Immunoprecipitation experiments confirmed this interaction. Docking experiments indicated that RNH1 physically interacts with IRE1 through its cytosolic RNase domain. Upon ER stress, the interaction of RNH1 with IRE1 in the ER increased at the expense of the nuclear pool of RNH1. Inhibition of RNH1 expression using siRNA mediated RNA interference upon ER stress led to an increased splicing activity of XBP1. Modulation of IRE1 RNase activity by RNH1 was recapitulated in a cell-free system, suggesting direct regulation of IRE1 by RNH. We conclude that RNH1 attenuates the activity of IRE1 by interacting with its ribonuclease domain. These findings have implications for understanding the molecular mechanism by which IRE1 signaling is attenuated upon ER stress.


Subject(s)
Carrier Proteins/metabolism , Endoribonucleases/antagonists & inhibitors , Enzyme Inhibitors/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Carrier Proteins/pharmacology , Cell Line, Transformed , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Humans , Models, Molecular , Protein Domains , Proteome , RNA Splicing , Uromodulin/metabolism
9.
Sci Rep ; 7(1): 10550, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28874730

ABSTRACT

Metabolic reprogramming is critical for T cell fate and polarization and is regulated by metabolic checkpoints, including Myc, HIF-1α, AMPK and mTORC1. Our objective was to determine the impact of mycophenolic acid (MPA) in comparison with rapamycin (Rapa), an inhibitor of mTORC1, on the metabolism of Jurkat T cells. We identified a drug-specific transcriptome signature consisting of the key enzymes and transporters involved in glycolysis, glutaminolysis or nucleotide synthesis. MPA produced an early and transient drop in the intracellular ATP content related to the inhibition of de novo synthesis of purines, leading to the activation of the energy sensor AMPK. MPA decreases glycolytic flux, consistent with a reduction in glucose uptake, but also in the oxidation of glutamine. Additionally, both drugs reduce aerobic glycolysis. The expression of HIF-1α and Myc, promoting the activation of glycolysis and glutaminolysis, was inhibited by MPA and Rapa. In conclusion, we report that MPA profoundly impacts the cellular metabolism of Jurkat T cells by generating an energetic distress, decreasing the glycolytic and glutaminolytic fluxes and by targeting HIF-1α and Myc. These findings open interesting perspectives for novel combinatorial therapeutic strategies targeting metabolic checkpoints to block the proliferation of T cells.


Subject(s)
Energy Metabolism/drug effects , Mycophenolic Acid/pharmacology , Transcriptome/drug effects , Glucose/metabolism , Glutamine/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Jurkat Cells , Proto-Oncogene Proteins c-myc/metabolism , Sirolimus/pharmacology
10.
Oncotarget ; 8(26): 43048-43060, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28574837

ABSTRACT

The anticancer drug 6-mercaptopurine (6-MP) inhibits de novo purine synthesis and acts as an antiproliferative agent by interfering with protein, DNA and RNA synthesis and promoting apoptosis. Metabolic reprogramming is crucial for tumor progression to foster cancer cells growth and proliferation, and is regulated by mechanistic target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) as well as the oncogenes Myc and hypoxia inducible factor 1α (HIF-1α). We hypothesized that 6-MP impacts metabolic remodeling through its action on nucleotide synthesis. The aim of our study is to provide a comprehensive characterization of the metabolic changes induced by 6-MP in leukemic T cells. Our results indicate that exposition to 6-MP rapidly reduces intracellular ATP concentration, leading to the activation of AMPK. In turn, mTOR, an AMPK target, was inhibited, and the expression of HIF-1α and Myc was reduced upon 6-MP incubation. As a consequence of these inhibitions, glucose and glutamine fluxes were strongly decreased. Notably, no difference was observed on glucose uptake upon exposition to 6-MP. In conclusion, our findings provide new insights into how 6-MP profoundly impacts cellular energetic metabolism by reducing ATP production and decreasing glycolytic and glutaminolytic fluxes, and how 6-MP modifies human leukemic T cells metabolism with potential antiproliferative effects.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Energy Metabolism/drug effects , Mercaptopurine/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/physiology , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Glycolysis/drug effects , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Metabolic Networks and Pathways/drug effects , Stress, Physiological/drug effects
11.
Biochimie ; 127: 23-36, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27126071

ABSTRACT

T lymphocytes undergo metabolic reprogramming to adapt to extracellular and intracellular cues. Specifically, T-cell metabolism results into ATP production, anabolism and catabolism pathways that not only support rapid cell growth and proliferation, but also differentiation and effector functions, recently referred as "immunometabolism". Quiescent naïve T cells rely on oxidative phosphorylation whereas aerobic glycolysis (Warburg effect) occurs in activated T cells (effector CD4(+) and CD8(+)). The molecular mechanisms that sense metabolic status and influence T-cell function require metabolic checkpoints including sensors of metabolic signals and transducers (Myc, HIF-1α, AMPK and mTOR). These metabolic checkpoints represent a novel therapeutic strategy for immune modulation. Interestingly, many immunosuppressive drugs including mTOR inhibitors (rapamycin), calcineurin inhibitors (tacrolimus, cyclosporine A) and inhibitors of de novo purine synthesis (6-mercaptopurine, mycophenolic acid and methotrexate) provide examples into how modulating these metabolic checkpoints can regulate T-cell activation, differentiation and function. In this Review we highlight emerging concepts about metabolic reprogramming in T-cell responses and we discuss the potential therapeutic interventions to influence T-cell fate and effector function.


Subject(s)
Immunosuppressive Agents/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Animals , Humans , T-Lymphocytes/immunology
12.
J Am Soc Nephrol ; 27(9): 2670-83, 2016 09.
Article in English | MEDLINE | ID: mdl-26823555

ABSTRACT

The ribonuclease angiogenin is a component of the mammalian stress response, and functions in both cell-autonomous and non-cell-autonomous ways to promote tissue adaptation to injury. We recently showed that angiogenin regulates tissue homeostasis during AKI associated with endoplasmic reticulum (ER) stress through the production of transfer RNA fragments that interfere with translation initiation and thereby alleviate ER stress. However, whether the paracrine signaling mediated by angiogenin secretion is a genuine component of the ER stress response to kidney injury is unknown. Here, we explored the molecular mechanisms by which angiogenin is secreted upon ER stress, and determined how it modulates the inflammatory microenvironment. In cultured renal epithelial cells, ER stress specifically induced angiogenin secretion under the selective control of inositol-requiring enzyme 1α, a key activator of the unfolded protein response. The transcription factors spliced X-box-binding protein 1 and p65, which are activated by inositol-requiring enzyme 1α upon ER stress, each bound the angiogenin promoter and controlled the amount of angiogenin secreted. Furthermore, p65 promoted angiogenin transcription in an ER stress-dependent manner. Similar to secretion of the ER stress-induced proinflammatory cytokine IL-6, secretion of angiogenin required the ER-Golgi pathway. Notably, incubation of human macrophages with angiogenin promoted macrophage reprogramming toward an activated and proinflammatory phenotype. In patients, angiogenin expression increased upon renal inflammation, and the urinary concentration of angiogenin correlated with the extent of immune-mediated kidney injury. Collectively, our data identify angiogenin as a mediator of the ER stress-dependent inflammatory response and as a potential noninvasive biomarker of AKI.


Subject(s)
Kidney/metabolism , Signal Transduction , Unfolded Protein Response/physiology , Animals , Cells, Cultured , Endoplasmic Reticulum Stress/physiology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Ribonuclease, Pancreatic/metabolism , Ribonuclease, Pancreatic/physiology
13.
Oncotarget ; 5(13): 4709-21, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25015210

ABSTRACT

Colorectal cancer (CRC) is a common tumor type with a high mortality rate, in part due to intrinsic drug resistance. Although bevacizumab, a VEGF-directed neutralizing antibody, is particularly active in this pathology, some patients never respond for reasons not well understood. We here wish to clarify the role of autocrine VEGF signaling in the response of CRC cells to angiogenesis inhibition. Our results show that CRC cells with intrinsic bevacizumab-resistance displayed pronounced upregulation of autocrine HIF-VEGF-VEGFR signaling in response to prolonged bevacizumab exposure whereas the same signaling pathway was downregulated in bevacizumab-sensitive xenografts. Importantly, both bevacizumab-sensitive and -resistant CRC xenografts were sensitive to nintedanib, a small molecule angiokinase inhibitor, which was associated with inhibition of mTORC1. In vitro studies revealed that bevacizumab-resistant cells displayed intrinsically higher HIF-VEGF signaling intensity and hypoxia tolerance compared to their bevacizumab-sensitive counterparts. Interestingly, although nintedanib showed comparable activity toward bevacizumab-sensitive cells under normoxia and hypoxia, the drug was three-fold more toxic to the resistant cells under hypoxia, suggesting that nintedanib attenuated the survival signaling that usually protects these cells from hypoxia-mediated cell death. In conclusion, our findings support a role for autocrine VEGF signaling in the survival of CRC cells to hypoxia and thus to angiogenesis inhibition. We further show that nintedanib, a small molecule angiokinase inhibitor, is active toward CRC models with intrinsic bevacizumab resistance supporting clinical trials of nintedanib in patients that do not respond to bevacizumab, alone or in combination with bevacizumab to increase angiogenesis inhibition.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Colorectal Neoplasms/drug therapy , Indoles/pharmacology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Bevacizumab , Blotting, Western , Cell Hypoxia , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Synergism , Enzyme Inhibitors/pharmacology , HT29 Cells , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Inbred Strains , Mice, Nude , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/prevention & control , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Small Molecule Libraries/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Xenograft Model Antitumor Assays
14.
Int J Oncol ; 42(5): 1644-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23546019

ABSTRACT

Irinotecan is a major anticancer agent specifically targeting DNA topoisomerase I. Its cytotoxicity is mediated via a two-step process involving accumulation of reversible DNA­topoisomerase I complexes associated with transient DNA single-strand breaks which subsequently are converted into permanent DNA double-strand breaks by the replication fork during S phase. Irinotecan may be selectively active for treatment of colorectal cancers that show microsatellite instability (MSI) due to deficiencies in mismatch repair enzymes, compared to tumors that are microsatellite stable but show chromosome instability (CIN). Although the clinical activity of irinotecan is principally limited by acquired drug resistance, surprisingly little is known about the influence of prolonged irinotecan exposure on the cell cycle dynamics. We have developed two colon cancer cell lines resistant to SN-38, the active metabolite of irinotecan, one derived from HT-29 (CIN), the other from HCT-116 (MSI). We here show that besides classical resistance mechanisms, SN-38 resistance is accompanied by an increased generation doubling time, a decreased S phase fraction and an increased G2 fraction in vitro as in tumor xenografts for both CIN and MSI models. As a consequence, SN-38-resistant cells and tumors show cross-resistance to the S-phase selective agent 5-fluorouracil. The resistance is accompanied by increased basal levels of γ-H2AX and phospho-Chk2 without notable changes in the levels of phospho-Chk1. Taken together, our results show that prolonged irinotecan exposure is accompanied by stable modifications of cell cycle dynamics which could have profound impact on tumor sensitivity to a wide range of antitumor agents and may influence tumor progression in patients.


Subject(s)
Camptothecin/analogs & derivatives , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Antineoplastic Agents/pharmacology , Camptothecin/pharmacology , Cell Cycle/drug effects , Cell Cycle/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Single-Stranded/drug effects , DNA Topoisomerases, Type I/drug effects , DNA Topoisomerases, Type I/genetics , HCT116 Cells , Humans , Irinotecan , Microsatellite Instability/drug effects , S Phase/drug effects , S Phase/genetics
15.
Clin Cancer Res ; 17(20): 6522-30, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21880790

ABSTRACT

PURPOSE: Epidermal growth factor receptor (EGFR) and VEGF(R) signaling show extensive cross-talk, providing a rationale for joint targeting of the two pathways. However, combinations of monoclonal antibodies (mAb) targeting EGFR and VEGF showed disappointing activity in patients with colorectal cancer (CRC). We speculated that inhibition of surface receptors and ligands might only partly prevent oncogenic signaling whereas small-molecule tyrosine kinase inhibitors (TKI) would also influence intracellular signaling. EXPERIMENTAL DESIGN: Mice with CRC xenografts were treated with two TKIs, vargatef and afatinib, or with two mAbs, bevacizumab and cetuximab, and their influence on tumor growth, viability, in vivo DNA synthesis, and the presence of phosphorylated EGFR and VEGFR was determined. The activity of the TKIs was further characterized in CRC cells with different KRAS status. RESULTS: Vargatef and afatinib together showed strong tumor growth inhibition toward HT-29 xenografts compared with either drug alone, which was associated with a 5-fold increase in apoptotic tumor cell death. In comparison, bevacizumab and cetuximab together were exclusively cytostatic with no more activity than either drug alone. Exposure to the two TKIs was accompanied by a marked decrease of tumor-associated intracellular phospho-VEGFR1 and phospho-EGFR, whereas similar exposure to the two mAbs had no detectable effect. A synergistic activity of vargatef plus afatinib was observed in all eight CRC cell lines examined, independent of KRAS status. CONCLUSIONS: Our results indicate that attenuation of intracellular EGFR and/or VEGF signaling is required for cytotoxic activity. These findings provide a rationale for trials of the TKIs, even in patients with mutant KRAS.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Afatinib , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Bevacizumab , Cell Proliferation/drug effects , Cell Survival/drug effects , Cetuximab , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Female , HT29 Cells , Humans , Indoles/pharmacology , Mice , Mice, Nude , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
16.
Mol Cancer Ther ; 10(8): 1481-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21622731

ABSTRACT

PM01183 is a novel marine-derived covalent DNA binder in clinical development. PM01183 is structurally similar to trabectedin (yondelis, ecteinascidin-743) except for the C subunit, and this modification is accompanied by different pharmacokinetics in cancer patients. We here characterize the interaction of PM01183 with the nucleotide excision repair (NER) pathway in comparison with trabectedin. Our results show for the first time that although neither PM01183 nor trabectedin is repaired by NER, both compounds are able to interfere with the NER machinery thereby attenuating the repair of specific NER substrates. We further show that the NER activity is increased in 3 of 4 cellular models with acquired resistance to cisplatin or oxaliplatin, confirming the involvement of NER in the resistance to platinum derivatives. Importantly, both PM01183 and trabectedin show unchanged or even enhanced activity toward all 4 cisplatin- and oxaliplatin-resistant cell lines. We finally show that combinations of PM01183 and cisplatin were mostly synergistic toward both parental and cisplatin-resistant ovarian carcinoma cells as indicated by Chou and Talalay analysis. These data show that the C subunit of trabectedin can be subjected to at least some structural modifications without loss of activity or NER interaction. While PM01183 and trabectedin appear functionally similar in cellular models, it is likely that the differences in pharmacokinetics may allow different dosing and scheduling of PM01183 in the clinic that could lead to novel and/or increased antitumor activity. Taken together, our results provide a mechanistic basis to support clinical trials of PM01183 alone or in combination with cisplatin.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Carbolines/pharmacology , Cisplatin/pharmacology , DNA Repair/drug effects , Dioxoles/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Tetrahydroisoquinolines/pharmacology , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/metabolism , Carbolines/chemistry , Cell Line, Tumor , Chromatin/metabolism , DNA Damage/drug effects , DNA Damage/radiation effects , Dioxoles/chemistry , Dioxoles/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Synergism , HCT116 Cells , HT29 Cells , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Neoplasms/metabolism , Neoplasms/pathology , Organoplatinum Compounds/pharmacology , Oxaliplatin , Tetrahydroisoquinolines/chemistry , Tetrahydroisoquinolines/metabolism , Trabectedin , Ultraviolet Rays/adverse effects
17.
Biochem Pharmacol ; 80(3): 335-43, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20399198

ABSTRACT

S23906 belongs to a novel class of alkylating anticancer agents forming bulky monofunctional DNA adducts. A unique feature of S23906 is its "helicase-like" activity leading to the destabilization of the surrounding duplex DNA. We here characterize the recognition and repair of S23906 adducts by the nucleotide excision repair (NER) machinery. All NER-deficient human cell lines tested showed increased sensitivity to S23906, which was particularly pronounced for cells deficient in XPC, CSB and XPA. In comparison, deficiencies in ERCC1 or XPF had lesser impact on the sensitivity to S23906. The sensitivity was, at least in part, linked to the conversion of unrepaired adducts into toxic DNA strand breaks as shown by single cell electrophoresis and gamma-H2AX formation. The pharmacological relevance of these findings was confirmed by the characterization of KB carcinoma cells with acquired S23906 resistance. These cells showed increased NER activity in vivo as well as toward damaged plasmid DNA in vitro. In particular, both global genome NER, as shown by unscheduled DNA synthesis, and transcription-coupled NER, as shown by transcriptional recovery, were up-regulated in the S23906-resistant cells. The increased NER activity was accompanied by up to 5-fold up-regulation of XPC, CSB and XPA proteins without detectable alterations of ERCC1 on the DNA, RNA or protein levels. Our results suggest that S23906 adducts are recognized and repaired by both NER sub-pathways in contrast to other members of this class, that are only recognized by transcription-coupled NER. We further show that NER activity can be up-regulated without changes in ERCC1 expression.


Subject(s)
Acronine/pharmacology , Antineoplastic Agents, Alkylating/metabolism , DNA Helicases/physiology , DNA Repair Enzymes/physiology , DNA-Binding Proteins/physiology , DNA/metabolism , Endonucleases/physiology , Acronine/metabolism , Alkylating Agents/chemistry , Alkylating Agents/metabolism , Antineoplastic Agents, Alkylating/chemistry , Binding Sites/physiology , Cell Line , Cell Line, Tumor , DNA Adducts/metabolism , Humans , Liver X Receptors , Orphan Nuclear Receptors/deficiency , Orphan Nuclear Receptors/physiology , Poly-ADP-Ribose Binding Proteins , Xeroderma Pigmentosum/metabolism
18.
J Cell Sci ; 121(Pt 8): 1275-83, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18388315

ABSTRACT

Transcription-coupled repair (TCR) plays a key role in the repair of DNA lesions induced by bulky adducts and is initiated when the elongating RNA polymerase II (Pol II) stalls at DNA lesions. This is accompanied by alterations in Pol II activity and stability. We have previously shown that the monofunctional adducts formed by irofulven (6-hydroxymethylacylfulvene) are exclusively recognized by TCR, without involvement of global genome repair (GGR), making irofulven a unique tool to characterize TCR-associated processes in vivo. Here, we characterize the influence of irofulven on Pol II activity, stability and mobility in living mammalian cells. Our results demonstrate that irofulven induces specific inhibition of nucleoplasmic RNA synthesis, an important decrease of Pol II mobility, coupled to the accumulation of initiating polymerase and a time-dependent loss of the engaged enzyme, associated with its polyubiquitylation. Both proteasome-mediated degradation of the stalled polymerase and new protein synthesis are necessary to allow Pol II recycling into preinitiating complexes. Together, our findings provide novel insights into the subsequent fate of the stalled RNA polymerase II and demonstrate the essential role of the recycling process for transcriptional reinitiation and viability of mammalian cells.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Repair , RNA Polymerase II/metabolism , Sesquiterpenes/pharmacology , Transcription, Genetic/drug effects , Chromatography, Affinity , HeLa Cells , Humans , Hydrolysis , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism
19.
Proc Natl Acad Sci U S A ; 104(32): 13062-7, 2007 Aug 07.
Article in English | MEDLINE | ID: mdl-17656556

ABSTRACT

Adducts induced by the antitumor alkylator ecteinascidin 743 (ET-743, Yondelis, trabectedin) represent a unique challenge to the DNA repair machinery because no pathway examined to date is able to remove the ET adducts, whereas cells deficient in nucleotide excision repair show increased resistance. We here describe the processing of the initial ET adducts into cytotoxic lesions and characterize the influence of cellular repair pathways on this process. Our findings show that exposure of proliferating mammalian cells to pharmacologically relevant concentrations of ET-743 is accompanied by rapid formation of DNA double-strand breaks (DSBs), as shown by the neutral comet assay and induction of focalized phosphorylated H2AX. The ET adducts are stable and can be converted into DSBs hours after the drug has been removed. Loss of homologous recombination repair has no influence on the initial levels of DSBs but is associated with the persistence of unrepaired DSBs after ET-743 is removed, resulting in extensive chromosomal abnormalities and pronounced sensitivity to the drug. In comparison, loss of nonhomologous end-joining had only modest effect on the sensitivity. The identification of DSB formation as a key step in the processing of ET-743 lesions represents a novel mechanism of action for the drug that is in agreement with its unusual potency. Because loss of repair proteins is common in human tumors, expression levels of selected repair factors may be useful in identifying patients particularly likely to benefit, or not, from treatment with ET-743.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Breaks, Double-Stranded/drug effects , DNA Repair , DNA Replication , Dioxoles/pharmacology , Recombination, Genetic , Tetrahydroisoquinolines/pharmacology , Cell Cycle/drug effects , Chromosome Aberrations , DNA-Binding Proteins/physiology , Dose-Response Relationship, Drug , HeLa Cells , Humans , Trabectedin
20.
Cancer Res ; 66(14): 7203-10, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16849567

ABSTRACT

S23906-1, a new DNA alkylating agent that reacts with the exocyclic 2-NH2 group of guanine residues yielding monofunctional adducts, is currently under clinical evaluation in phase I trials. To investigate the mechanism of action of S23906-1, we compared parental KB-3-1 cells and KB/S23-500 cells that are 15-fold resistant to S23906-1. Cell death induced by 1 micromol/L S23906-1 in KB-3-1 cells was associated with their irreversible arrest in the G2-M phases of the cell cycle followed by apoptosis, whereas a proportion of the resistant KB/S23-500 cells were able to exit from the G2 arrest and divide, leading to a significantly lower rate of apoptosis. The attenuated apoptotic response was associated with decreased Chk2 protein phosphorylation, indicating that the DNA damage signaling pathways are more potently activated in the sensitive cells. However, similar rates of adduct formation and repair were measured in both cell lines. Exposure to S23906-1 induced a higher formation of DNA breaks, measured by the comet assay, in sensitive cells. In agreement, a histone H2AX phosphorylation assay revealed that S23906-1 induced double-strand breaks (DSB) in a dose- and time-dependent manner and that these were more persistent in the parental cells. These DSBs were found mainly in S-phase cells and inhibited by aphidicolin, suggesting that they are DNA replication-mediated DSBs. These results suggest that secondary DNA lesions play an important role in the cytotoxicity of this compound and make histone H2AX phosphorylation an attractive marker for monitoring the efficacy of S23906-1.


Subject(s)
Acronine/analogs & derivatives , DNA Damage , DNA, Neoplasm/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Acronine/pharmacology , Alkylation , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Cycle/drug effects , Checkpoint Kinase 2 , DNA, Neoplasm/metabolism , Histones/metabolism , Humans , KB Cells , Phosphorylation , Protein Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...