Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3016, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38589367

ABSTRACT

Myelodysplastic syndromes (MDS) with mutated SF3B1 gene present features including a favourable outcome distinct from MDS with mutations in other splicing factor genes SRSF2 or U2AF1. Molecular bases of these divergences are poorly understood. Here we find that SF3B1-mutated MDS show reduced R-loop formation predominating in gene bodies associated with intron retention reduction, not found in U2AF1- or SRSF2-mutated MDS. Compared to erythroblasts from SRSF2- or U2AF1-mutated patients, SF3B1-mutated erythroblasts exhibit augmented DNA synthesis, accelerated replication forks, and single-stranded DNA exposure upon differentiation. Importantly, histone deacetylase inhibition using vorinostat restores R-loop formation, slows down DNA replication forks and improves SF3B1-mutated erythroblast differentiation. In conclusion, loss of R-loops with associated DNA replication stress represents a hallmark of SF3B1-mutated MDS ineffective erythropoiesis, which could be used as a therapeutic target.


Subject(s)
Myelodysplastic Syndromes , R-Loop Structures , Humans , Splicing Factor U2AF/genetics , Serine-Arginine Splicing Factors/genetics , RNA Splicing Factors/genetics , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Mutation , Transcription Factors/genetics , Phosphoproteins/genetics
2.
Haematologica ; 107(1): 268-283, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-33241676

ABSTRACT

The gene CXXC5, encoding a Retinoid-Inducible Nuclear Factor (RINF), is located within a region at 5q31.2 commonly deleted in myelodysplastic syndrome (MDS) and adult acute myeloid leukemia (AML). RINF may act as an epigenetic regulator and has been proposed as a tumor suppressor in hematopoietic malignancies. However, functional studies in normal hematopoiesis are lacking, and its mechanism of action is unknow. Here, we evaluated the consequences of RINF silencing on cytokineinduced erythroid differentiation of human primary CD34+ progenitors. We found that RINF is expressed in immature erythroid cells and that RINF-knockdown accelerated erythropoietin-driven maturation, leading to a significant reduction (~45%) in the number of red blood cells (RBCs), without affecting cell viability. The phenotype induced by RINF-silencing was TGFß-dependent and mediated by SMAD7, a TGFß- signaling inhibitor. RINF upregulates SMAD7 expression by direct binding to its promoter and we found a close correlation between RINF and SMAD7 mRNA levels both in CD34+ cells isolated from bone marrow of healthy donors and MDS patients with del(5q). Importantly, RINF knockdown attenuated SMAD7 expression in primary cells and ectopic SMAD7 expression was sufficient to prevent the RINF knockdowndependent erythroid phenotype. Finally, RINF silencing affects 5'-hydroxymethylation of human erythroblasts, in agreement with its recently described role as a Tet2- anchoring platform in mouse. Altogether, our data bring insight into how the epigenetic factor RINF, as a transcriptional regulator of SMAD7, may fine-tune cell sensitivity to TGFß superfamily cytokines and thus play an important role in both normal and pathological erythropoiesis.


Subject(s)
DNA-Binding Proteins , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Smad7 Protein , Transcription Factors , Adult , Animals , Cell Cycle , Epigenesis, Genetic , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Myelodysplastic Syndromes/genetics , RNA, Messenger , Smad7 Protein/genetics
3.
Exp Hematol ; 99: 12-20.e3, 2021 07.
Article in English | MEDLINE | ID: mdl-34077792

ABSTRACT

Red blood cell production, or erythropoiesis, is a proliferative process that requires tight regulation. Erythropoietin (Epo) is a glycoprotein cytokine that plays a major role in erythropoiesis by triggering erythroid progenitors/precursors of varying sensitivity. The concentration of Epo in bone marrow is hypothesized to be suboptimal, and the survival of erythroid cells has been suggested to depend on Epo sensitivity. However, the key factors that control Epo sensitivity remain unknown. Two types of transferrin receptors (TfRs), TfR1 and TfR2, are known to play a role in iron uptake in erythroid cells. Here, we hypothesized that TfRs may additionally modulate Epo sensitivity during erythropoiesis by modulating Epo receptor (EpoR) signaling. Using an Epo-sensitive UT-7 (UT7/Epo) erythroid cell and human erythroid progenitor cell models, we report that iron-loaded transferrin, that is, holo-transferrin (holo-Tf), synergizes with suboptimal Epo levels to improve erythroid cell survival, proliferation, and differentiation. This is accomplished via the major signaling pathways of erythropoiesis, which include signal transducer and activator of transcription 5 (STAT5), mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK), and phosphoinositide-3-kinase (PI3K)/AKT. Furthermore, we found that this cooperation is improved by, but does not require, the internalization of TfR1. Interestingly, we observed that loss of TfR2 stabilizes EpoR levels and abolishes the beneficial effects of holo-Tf. Overall, these data reveal novel signaling properties of TfRs, which involve the regulation of erythropoiesis through EpoR signaling.


Subject(s)
Antigens, CD/metabolism , Cell Proliferation/drug effects , Erythroblasts/metabolism , Erythropoietin/pharmacology , Iron/pharmacology , MAP Kinase Signaling System/drug effects , Receptors, Transferrin/metabolism , Transferrin/pharmacology , Cell Line , Cell Survival/drug effects , Erythropoietin/metabolism , Humans , Iron/metabolism , Transferrin/metabolism
4.
Am J Hematol ; 96(4): 480-492, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33476437

ABSTRACT

Efficient erythropoiesis relies on the expression of the transferrin receptor type 2 (TFR2). In erythroid precursors, TFR2 facilitates the export of the erythropoietin receptor (EPOR) to cell surface, which ensures the survival and proliferation of erythroblasts. Although TFR2 has a crucial role in erythropoiesis regulation, its mechanism of action remains to be clarified. To understand its role better, we aimed at identifying its protein partners by mass-spectrometry after immunoprecipitation in erythroid cells. Here we report the kinase MRCKα (myotonic dystrophy kinase-related CDC42-binding kinase α) as a new partner of both TFR2 and EPOR in erythroblasts. We show that MRCKα is co-expressed with TFR2, and TFR1 during terminal differentiation and regulates the internalization of the two types of transferrin receptors. The knockdown of MRCKα by shRNA in human primary erythroblasts leads to a decreased cell surface expression of both TFR1 and TFR2, an increased cell-surface expression of EPOR, and a delayed differentiation. Additionally, knockout of Mrckα in the murine MEDEP cells also leads to a striking delay in erythropoiesis, showcasing the importance of this kinase in both species. Our data highlight the importance of MRCKα in the regulation of erythropoiesis.


Subject(s)
Erythropoiesis/physiology , Myotonin-Protein Kinase/physiology , Animals , CRISPR-Cas Systems , Cells, Cultured , Endocytosis , Erythroblasts/cytology , Erythroblasts/metabolism , Gene Knockout Techniques , Humans , Iron/metabolism , Mice , Myotonin-Protein Kinase/isolation & purification , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Receptors, Erythropoietin/metabolism , Receptors, Transferrin/metabolism , cdc42 GTP-Binding Protein/metabolism
6.
Sci Rep ; 10(1): 8184, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32424321

ABSTRACT

The corallivorous Crown-of-Thorns Starfish (CoTS, Acanthaster spp.) has been linked with the widespread loss of scleractinian coral cover on Indo-Pacific reefs during periodic population outbreaks. Here, we re-examine CoTS consumption by coral reef fish species by using new DNA technologies to detect Pacific Crown-of-Thorns Starfish (Acanthaster cf. solaris) in fish faecal and gut content samples. CoTS DNA was detected in samples from 18 different coral reef fish species collected on reefs at various stages of CoTS outbreaks in the Great Barrier Reef Marine Park, nine of which had not been previously reported to feed on CoTS. A comprehensive set of negative and positive control samples confirmed that our collection, processing and analysis procedures were robust, although food web transfer of CoTS DNA cannot be ruled out for some fish species. Our results, combined with the (i) presence of CoTS spines in some samples, (ii) reported predation on CoTS gametes, larvae and settled individuals, and (iii) known diet information for fish species examined, strongly indicate that direct fish predation on CoTS may well be more common than is currently appreciated. We provide recommendations for specific management approaches to enhance predation on CoTS by coral reef fishes, and to support the mitigation of CoTS outbreaks and reverse declines in hard coral cover.


Subject(s)
DNA Barcoding, Taxonomic , Feces , Intestines , Starfish/classification , Starfish/genetics , Animals , Coral Reefs , Predatory Behavior
7.
Blood Adv ; 4(7): 1464-1477, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32282884

ABSTRACT

Murine-based cellular models have provided and continue to provide many useful insights into the fundamental mechanisms of erythropoiesis, as well as insights into the pathophysiology of inherited and acquired red cell disorders. Although detailed information on many aspects of these cell models is available, comprehensive proteomic data are lacking. This is a critical knowledge gap, as proteins are effectors of most biologic processes. To address this critical unmet need, proteomes of the murine cell lines Friend erythroleukemia (MEL), GATA1 erythroid (G1ER), and embryonic stem cell-derived erythroid progenitor (MEDEP) and proteomes of cultured murine marrow-derived erythroblasts at different stages of terminal erythroid differentiation were analyzed. The proteomes of MEDEP cells and primary murine erythroid cells were most similar, whereas those of MEL and G1ER cells were more distantly related. We demonstrated that the overall cellular content of histones does not decrease during terminal differentiation, despite strong chromatin condensation. Comparison of murine and human proteomes throughout terminal erythroid differentiation revealed that many noted transcriptomic changes were significantly dampened at the proteome level, especially at the end of the terminal differentiation process. Analysis of the early events associated with induction of terminal differentiation in MEDEP cells revealed divergent alterations in associated transcriptomes and proteomes. These proteomic data are powerful and valuable tools for the study of fundamental mechanisms of normal and disordered erythropoiesis and will be of broad interest to a wide range of investigators for making the appropriate choice of various cell lines to study inherited and acquired diseases of the erythrocyte.


Subject(s)
Leukemia, Erythroblastic, Acute , Proteomics , Animals , Erythroblasts , Erythroid Cells , Erythropoiesis , Humans , Mice
8.
Sci Transl Med ; 11(500)2019 07 10.
Article in English | MEDLINE | ID: mdl-31292266

ABSTRACT

Myelodysplastic syndromes (MDS) with ring sideroblasts are hematopoietic stem cell disorders with erythroid dysplasia and mutations in the SF3B1 splicing factor gene. Patients with MDS with SF3B1 mutations often accumulate excessive tissue iron, even in the absence of transfusions, but the mechanisms that are responsible for their parenchymal iron overload are unknown. Body iron content, tissue distribution, and the supply of iron for erythropoiesis are controlled by the hormone hepcidin, which is regulated by erythroblasts through secretion of the erythroid hormone erythroferrone (ERFE). Here, we identified an alternative ERFE transcript in patients with MDS with the SF3B1 mutation. Induction of this ERFE transcript in primary SF3B1-mutated bone marrow erythroblasts generated a variant protein that maintained the capacity to suppress hepcidin transcription. Plasma concentrations of ERFE were higher in patients with MDS with an SF3B1 gene mutation than in patients with SF3B1 wild-type MDS. Thus, hepcidin suppression by a variant ERFE is likely responsible for the increased iron loading in patients with SF3B1-mutated MDS, suggesting that ERFE could be targeted to prevent iron-mediated toxicity. The expression of the variant ERFE transcript that was restricted to SF3B1-mutated erythroblasts decreased in lenalidomide-responsive anemic patients, identifying variant ERFE as a specific biomarker of clonal erythropoiesis.


Subject(s)
Homeostasis , Iron/metabolism , Mutation/genetics , Myelodysplastic Syndromes/genetics , Peptide Hormones/genetics , Phosphoproteins/genetics , RNA Splicing Factors/genetics , Alternative Splicing/drug effects , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Blood Transfusion , Cell Line , Cell Lineage/drug effects , Cell Survival/drug effects , Clone Cells , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Hepcidins/metabolism , Homeostasis/drug effects , Humans , Lenalidomide/pharmacology , Mice , Myelodysplastic Syndromes/blood , Peptide Hormones/blood , Peptide Hormones/chemistry , Peptide Hormones/metabolism , Protein Biosynthesis/drug effects , RNA Splice Sites/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
9.
Curr Opin Hematol ; 24(3): 191-197, 2017 May.
Article in English | MEDLINE | ID: mdl-28072603

ABSTRACT

PURPOSE OF REVIEW: Myelodysplastic syndromes (MDS) are heterogeneous diseases of the hematopoietic stem cell in the elderly. Anemia is the main symptom that mostly correlates with dysplastic erythropoiesis in the bone marrow. We will review the recent advances in understanding the diverse mechanisms of dyserythropoiesis. RECENT FINDINGS: Dyserythropoiesis defined as 10% dysplastic erythroid cells in the bone marrow is found in more than 80% of early MDS. Immature erythroblasts accumulate at the expense of mature erythroblasts due to differentiation arrest and apoptosis. In early MDS with dyserythropoiesis, caspase-dependent cleavage of the erythroid transcription factor GATA-1 occurring in basophilic erythroblasts accounts for impairment of final maturation. Depending on initiating genetic alteration, specific mechanisms contribute to erythroid defect. In MDS with 5q deletion, the haploinsufficiency of ribosomal protein gene, RPS14, opposes the transition of immature to mature erythroblasts by inducing a p53-dependent ribosome stress, cell cycle arrest and apoptosis. Recent work identifies the activation of a p53-S100A8/9 innate immune pathway that both intrinsically and extrinsically contributes to defective erythropoiesis. In MDS with ring sideroblasts, a paradigm of dyserythropoiesis, a unique mutation in SF3B1 splicing factor gene induces a multiplicity of alterations at RNA level that deeply modifies the patterns of gene expression. SUMMARY: Insights in the pathophysiology of MDS with dyserythropoiesis may guide the choice of the appropriate therapy, for instance lenalidomide in MDS with del(5q). A better understanding of the mechanisms of dyserthropoiesis is required to treat anemia in non-del(5q) MDS, especially in case of resistance to first-line therapy by erythropoiesis-stimulating agents.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow/metabolism , Erythropoiesis , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/metabolism , Anemia, Macrocytic/genetics , Anemia, Macrocytic/metabolism , Anemia, Macrocytic/pathology , Anemia, Sideroblastic/etiology , Anemia, Sideroblastic/metabolism , Anemia, Sideroblastic/pathology , Animals , Bone Marrow/pathology , Bone Marrow Cells/pathology , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Chromosomes, Human, Pair 5/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , Erythroid Cells/pathology , Erythropoiesis/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Expression Regulation , Humans , Immunity, Innate , Mitochondria/genetics , Mitochondria/immunology , Mitochondria/metabolism , Myelodysplastic Syndromes/diagnosis , RNA Splicing , Signal Transduction
10.
Blood ; 129(4): 484-496, 2017 01 26.
Article in English | MEDLINE | ID: mdl-27856460

ABSTRACT

Myelodysplastic syndromes (MDSs) are hematopoietic stem cell disorders in which recurrent mutations define clonal hematopoiesis. The origin of the phenotypic diversity of non-del(5q) MDS remains unclear. Here, we investigated the clonal architecture of the CD34+CD38- hematopoietic stem/progenitor cell (HSPC) compartment and interrogated dominant clones for MDS-initiating cells. We found that clones mainly accumulate mutations in a linear succession with retention of a dominant subclone. The clone detected in the long-term culture-initiating cell compartment that reconstitutes short-term human hematopoiesis in xenotransplantation models is usually the dominant clone, which gives rise to the myeloid and to a lesser extent to the lymphoid lineage. The pattern of mutations may differ between common myeloid progenitors (CMPs), granulomonocytic progenitors (GMPs), and megakaryocytic-erythroid progenitors (MEPs). Rare STAG2 mutations can amplify at the level of GMPs, from which it may drive the transformation to acute myeloid leukemia. We report that major truncating BCOR gene mutation affecting HSPC and CMP was beneath the threshold of detection in GMP or MEP. Consistently, BCOR knock-down (KD) in normal CD34+ progenitors modifies their granulocytic and erythroid differentiation. Clonal architecture of the HSPC compartment and mutations selected during differentiation contribute to the phenotypic heterogeneity of MDS. Defining the hierarchy of driver mutations provides insights into the process of transformation and may guide the search for novel therapeutic strategies.


Subject(s)
Chromosomes, Human, Pair 5 , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/genetics , Lymphocytes/metabolism , Mutation , Myelodysplastic Syndromes/genetics , Myeloid Cells/metabolism , ADP-ribosyl Cyclase 1/deficiency , ADP-ribosyl Cyclase 1/genetics , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Cell Cycle Proteins , Cell Differentiation , Cell Lineage/genetics , Clone Cells , Disease Progression , Female , Gene Expression , Gene Knockdown Techniques , Hematopoietic Stem Cells/pathology , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Lymphocytes/pathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred NOD , Myelodysplastic Syndromes/complications , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Myeloid Cells/pathology , Phenotype , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transplantation, Heterologous
11.
Genes Dev ; 29(24): 2603-16, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26680303

ABSTRACT

Tight coordination of cell proliferation and differentiation is central to red blood cell formation. Erythropoietin controls the proliferation and survival of red blood cell precursors, while variations in GATA-1/FOG-1 complex composition and concentrations drive their maturation. However, clear evidence of cross-talk between molecular pathways is lacking. Here, we show that erythropoietin activates AKT, which phosphorylates GATA-1 at Ser310, thereby increasing GATA-1 affinity for FOG-1. In turn, FOG-1 displaces pRb/E2F-2 from GATA-1, ultimately releasing free, proproliferative E2F-2. Mice bearing a Gata-1(S310A) mutation suffer from fatal anemia when a compensatory pathway for E2F-2 production involving insulin-like growth factor-1 (IGF-1) signaling is simultaneously abolished. In the context of the GATA-1(V205G) mutation resulting in lethal anemia, we show that the Ser310 cannot be phosphorylated and that constitutive phosphorylation at this position restores partial erythroid differentiation. This study sheds light on the GATA-1 pathways that synchronize cell proliferation and differentiation for tissue homeostasis.


Subject(s)
Cell Differentiation/genetics , Erythroid Cells/cytology , Erythropoiesis/physiology , Erythropoietin/metabolism , Insulin-Like Growth Factor I/metabolism , Signal Transduction , Anemia, Hemolytic/genetics , Animals , Cell Proliferation/genetics , Enzyme Activation/genetics , Erythropoiesis/genetics , Erythropoietin/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Knock-In Techniques , Mice , Mutation , Nuclear Proteins/metabolism , Oncogene Protein v-akt/metabolism , Phosphorylation , Protein Binding/genetics , Transcription Factors/metabolism
12.
Biochem Biophys Res Commun ; 429(1-2): 1-5, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23137537

ABSTRACT

Malignant transformation is a multistep process requiring oncogenic activation, promoting cellular proliferation, frequently coupled to inhibition of terminal differentiation. Consequently, forcing the reengagement of terminal differentiation of transformed cells coupled or not with an inhibition of their proliferation is a putative therapeutic approach to counteracting tumorigenicity. UT7 is a human leukemic cell line able to grow in the presence of IL3, GM-CSF and Epo. This cell line has been widely used to study Epo-R/Epo signaling pathways but is a poor model for erythroid differentiation. We used the BET bromodomain inhibition drug JQ1 to target gene expression, including that of c-Myc. We have shown that only 2 days of JQ1 treatment was required to transitory inhibit Epo-induced UT7 proliferation and to restore terminal erythroid differentiation. This study highlights the importance of a cellular erythroid cycle break mediated by c-Myc inhibition before initiation of the erythropoiesis program and describes a new model for BET bromodomain inhibitor drug application.


Subject(s)
Azepines/pharmacology , Erythropoiesis/drug effects , Erythropoietin/pharmacology , Leukemia, Erythroblastic, Acute/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Triazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Humans , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc/metabolism
13.
Oecologia ; 170(2): 567-73, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22447198

ABSTRACT

The dynamic nature of coral reefs offers a rare opportunity to examine the response of ecosystems to disruption due to climate change. In 1998, the Great Barrier Reef experienced widespread coral bleaching and mortality. As a result, cryptobenthic fish assemblages underwent a dramatic phase-shift. Thirteen years, and up to 96 fish generations later, the cryptobenthic fish assemblage has not returned to its pre-bleach configuration. This is despite coral abundances returning to, or exceeding, pre-bleach values. The post-bleach fish assemblage exhibits no evidence of recovery. If these short-lived fish species are a model for their longer-lived counterparts, they suggest that (1) the full effects of the 1998 bleaching event on long-lived fish populations have yet to be seen, (2) it may take decades, or more, before recovery or regeneration of these long-lived species will begin, and (3) fish assemblages may not recover to their previous composition despite the return of corals.


Subject(s)
Climate Change , Coral Reefs , Fishes/growth & development , Animals , Biodiversity , Conservation of Natural Resources , Population Dynamics
14.
Photochem Photobiol Sci ; 7(3): 328-36, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18389150

ABSTRACT

Twenty-nine analogs of indirubin, an isomer of indigo, have been synthesized to optimize its promising kinase inhibitory scaffold. These compounds being also pigmented, have been tested for their photoreactivity. Absorption maxima were between 485 nm and 560 nm. Addition of fetal calf serum induced fluorescence and time dependent absorption modifications. Appropriate illumination induced Reactive Oxygen Species (ROS) production for nineteen compounds out of twenty-nine. The relationship between fluorescence and ROS production is discussed. Six compounds showed an important toxicity on F98 cells, a murine glioma cell line. Three of these were found to be also phototoxic, as four other non-toxic compounds. All but one phototoxic compounds were detected as ROS producers by in vitro tests. Photoreactivity assessment is important to anticipate adverse reactions for compounds that might be clinically developed. The experimental assay was found to be the only way to evaluate the photoreactivity of this family of compounds since no predictive criteria on structures could be found. Combining the vascular tumor growth inhibition induced by kinase inhibitors with the massive local blood flow arrest following photodynamic treatment may be an efficient anti-cancer strategy. These data could orientate further syntheses of either non-photoreactive compounds or compounds displaying both kinase inhibitory activity and strong phototoxicity.


Subject(s)
Light , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/radiation effects , Protein Kinases/drug effects , Animals , Cell Death/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Fluorescence , Humans , Indoles/chemistry , Indoles/pharmacology , Indoles/radiation effects , Molecular Structure , Photochemistry , Protein Kinase Inhibitors/pharmacology , Rats , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/radiation effects , Sensitivity and Specificity , Spectrometry, Fluorescence/methods , Spectrophotometry, Ultraviolet/methods , Stereoisomerism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...