Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.670
Filter
1.
Nature ; 627(8005): 880-889, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38480884

ABSTRACT

The evolutionary processes that underlie the marked sensitivity of small cell lung cancer (SCLC) to chemotherapy and rapid relapse are unknown1-3. Here we determined tumour phylogenies at diagnosis and throughout chemotherapy and immunotherapy by multiregion sequencing of 160 tumours from 65 patients. Treatment-naive SCLC exhibited clonal homogeneity at distinct tumour sites, whereas first-line platinum-based chemotherapy led to a burst in genomic intratumour heterogeneity and spatial clonal diversity. We observed branched evolution and a shift to ancestral clones underlying tumour relapse. Effective radio- or immunotherapy induced a re-expansion of founder clones with acquired genomic damage from first-line chemotherapy. Whereas TP53 and RB1 alterations were exclusively part of the common ancestor, MYC family amplifications were frequently not constituents of the founder clone. At relapse, emerging subclonal mutations affected key genes associated with SCLC biology, and tumours harbouring clonal CREBBP/EP300 alterations underwent genome duplications. Gene-damaging TP53 alterations and co-alterations of TP53 missense mutations with TP73, CREBBP/EP300 or FMN2 were significantly associated with shorter disease relapse following chemotherapy. In summary, we uncover key processes of the genomic evolution of SCLC under therapy, identify the common ancestor as the source of clonal diversity at relapse and show central genomic patterns associated with sensitivity and resistance to chemotherapy.


Subject(s)
Evolution, Molecular , Immunotherapy , Lung Neoplasms , Platinum , Small Cell Lung Carcinoma , Animals , Female , Humans , Male , Mice , Middle Aged , Clone Cells/drug effects , Clone Cells/metabolism , Clone Cells/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Genes, myc/genetics , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Mutation , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Platinum/pharmacology , Platinum/therapeutic use , Recurrence , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/immunology , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/therapy
2.
Nature ; 620(7974): 651-659, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37468627

ABSTRACT

Even among genetically identical cancer cells, resistance to therapy frequently emerges from a small subset of those cells1-7. Molecular differences in rare individual cells in the initial population enable certain cells to become resistant to therapy7-9; however, comparatively little is known about the variability in the resistance outcomes. Here we develop and apply FateMap, a framework that combines DNA barcoding with single-cell RNA sequencing, to reveal the fates of hundreds of thousands of clones exposed to anti-cancer therapies. We show that resistant clones emerging from single-cell-derived cancer cells adopt molecularly, morphologically and functionally distinct resistant types. These resistant types are largely predetermined by molecular differences between cells before drug addition and not by extrinsic factors. Changes in the dose and type of drug can switch the resistant type of an initial cell, resulting in the generation and elimination of certain resistant types. Samples from patients show evidence for the existence of these resistant types in a clinical context. We observed diversity in resistant types across several single-cell-derived cancer cell lines and cell types treated with a variety of drugs. The diversity of resistant types as a result of the variability in intrinsic cell states may be a generic feature of responses to external cues.


Subject(s)
Antineoplastic Agents , Clone Cells , Drug Resistance, Neoplasm , Neoplasms , Humans , Clone Cells/drug effects , Clone Cells/metabolism , Clone Cells/pathology , DNA Barcoding, Taxonomic , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , RNA-Seq , Single-Cell Gene Expression Analysis , Tumor Cells, Cultured , Antineoplastic Agents/pharmacology
3.
Nature ; 615(7950): 127-133, 2023 03.
Article in English | MEDLINE | ID: mdl-36813966

ABSTRACT

Haematopoietic stem cells (HSCs) are a rare cell type that reconstitute the entire blood and immune systems after transplantation and can be used as a curative cell therapy for a variety of haematological diseases1,2. However, the low number of HSCs in the body makes both biological analyses and clinical application difficult, and the limited extent to which human HSCs can be expanded ex vivo remains a substantial barrier to the wider and safer therapeutic use of HSC transplantation3. Although various reagents have been tested in attempts to stimulate the expansion of human HSCs, cytokines have long been thought to be essential for supporting HSCs ex vivo4. Here we report the establishment of a culture system that allows the long-term ex vivo expansion of human HSCs, achieved through the complete replacement of exogenous cytokines and albumin with chemical agonists and a caprolactam-based polymer. A phosphoinositide 3-kinase activator, in combination with a thrombopoietin-receptor agonist and the pyrimidoindole derivative UM171, were sufficient to stimulate the expansion of umbilical cord blood HSCs that are capable of serial engraftment in xenotransplantation assays. Ex vivo HSC expansion was further supported by split-clone transplantation assays and single-cell RNA-sequencing analysis. Our chemically defined expansion culture system will help to advance clinical HSC therapies.


Subject(s)
Cell Culture Techniques , Cell Proliferation , Cytokines , Hematopoietic Stem Cells , Humans , Cell Proliferation/drug effects , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/metabolism , Fetal Blood/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Cell Culture Techniques/methods , Albumins , Caprolactam , Polymers , Receptors, Thrombopoietin , Transplantation, Heterologous , Single-Cell Gene Expression Analysis
5.
Nature ; 601(7891): 125-131, 2022 01.
Article in English | MEDLINE | ID: mdl-34880496

ABSTRACT

All cancers emerge after a period of clonal selection and subsequent clonal expansion. Although the evolutionary principles imparted by genetic intratumour heterogeneity are becoming increasingly clear1, little is known about the non-genetic mechanisms that contribute to intratumour heterogeneity and malignant clonal fitness2. Here, using single-cell profiling and lineage tracing (SPLINTR)-an expressed barcoding strategy-we trace isogenic clones in three clinically relevant mouse models of acute myeloid leukaemia. We find that malignant clonal dominance is a cell-intrinsic and heritable property that is facilitated by the repression of antigen presentation and increased expression of the secretory leukocyte peptidase inhibitor gene (Slpi), which we genetically validate as a regulator of acute myeloid leukaemia. Increased transcriptional heterogeneity is a feature that enables clonal fitness in diverse tissues and immune microenvironments and in the context of clonal competition between genetically distinct clones. Similar to haematopoietic stem cells3, leukaemia stem cells (LSCs) display heritable clone-intrinsic properties of high, and low clonal output that contribute to the overall tumour mass. We demonstrate that LSC clonal output dictates sensitivity to chemotherapy and, although high- and low-output clones adapt differently to therapeutic pressure, they coordinately emerge from minimal residual disease with increased expression of the LSC program. Together, these data provide fundamental insights into the non-genetic transcriptional processes that underpin malignant clonal fitness and may inform future therapeutic strategies.


Subject(s)
Cell Competition , Clone Cells/pathology , Leukemia, Myeloid, Acute/pathology , Single-Cell Analysis , Animals , Cell Competition/drug effects , Cell Line , Cell Lineage/drug effects , Clone Cells/drug effects , Clone Cells/metabolism , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Inbred C57BL , Secretory Leukocyte Peptidase Inhibitor/metabolism
6.
Biomolecules ; 11(12)2021 12 04.
Article in English | MEDLINE | ID: mdl-34944475

ABSTRACT

The BALB/c cell transformation assay (BALB-CTA) considers inter- and intra-tumor heterogeneities and affords the possibility of a direct comparison between untransformed and malignant cells. In the present study, we established monoclonal cell lines that originate from the BALB-CTA and mimic heterogeneous tumor cell populations, in order to investigate phenotype-specific effects of the anti-diabetic drug metformin and the short-chain fatty acid butyrate. Growth inhibitory effects were measured with a ViCell XR cell counter. The BALB/c tumor therapy model (BALB-TTM) was performed, and the extracellular glucose level was measured in the medium supernatant. Using a Seahorse Analyzer, the metabolic phenotypes of four selected clones were characterized, and effects on energy metabolism were investigated. Anti-carcinogenic effects and reduced glucose uptake after butyrate application were observed in the BALB-TTM. Metabolic characterization of the cell clones revealed three different phenotypes. Surprisingly, treatment with metformin or butyrate induced opposite metabolic shifts with similar patterns in all cell clones tested. In conclusion, the BALB-TTM is a relevant model for mechanistic cancer research, and the generation of monoclonal cell lines offers a novel possibility to investigate specific drug effects in a heterogeneous tumor cell population. The results indicate that induced alterations in energy metabolism seem to be independent of the original metabolic phenotype.


Subject(s)
Butyrates/pharmacology , Cell Transformation, Neoplastic/drug effects , Energy Metabolism/drug effects , Glucose/metabolism , Metformin/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/metabolism , Clone Cells/cytology , Clone Cells/drug effects , Culture Media/chemistry , Humans , Mice , Models, Biological , Phenotype
7.
Nat Commun ; 12(1): 4803, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34376657

ABSTRACT

Chemotherapies may increase mutagenesis of healthy cells and change the selective pressures in tissues, thus influencing their evolution. However, their contributions to the mutation burden and clonal expansions of healthy somatic tissues are not clear. Here, exploiting the mutational footprint of some chemotherapies, we explore their influence on the evolution of hematopoietic cells. Cells of Acute Myeloid Leukemia (AML) secondary to treatment with platinum-based drugs show the mutational footprint of these drugs, indicating that non-malignant blood cells receive chemotherapy mutations. No trace of the 5-fluorouracil (5FU) mutational signature is found in AMLs secondary to exposure to 5FU, suggesting that cells establishing the leukemia could be quiescent during treatment. Using the platinum-based mutational signature as a barcode, we determine that the clonal expansion originating the secondary AMLs begins after the start of the cytotoxic treatment. Its absence in clonal hematopoiesis cases is consistent with the start of the clonal expansion predating the exposure to platinum-based drugs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Hematopoiesis/drug effects , Leukemia, Myeloid/genetics , Mutagenesis/drug effects , Acute Disease , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Clonal Evolution/drug effects , Clonal Evolution/genetics , Clone Cells/drug effects , Clone Cells/metabolism , Clone Cells/pathology , Cohort Studies , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Hematopoiesis/genetics , Humans , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid/chemically induced , Mutation/drug effects , Neoplasms, Second Primary/chemically induced , Neoplasms, Second Primary/genetics , Platinum/administration & dosage , Platinum/adverse effects , Tumor Suppressor Protein p53/genetics
8.
Nature ; 596(7873): 576-582, 2021 08.
Article in English | MEDLINE | ID: mdl-34381210

ABSTRACT

Non-genetic mechanisms have recently emerged as important drivers of cancer therapy failure1, where some cancer cells can enter a reversible drug-tolerant persister state in response to treatment2. Although most cancer persisters remain arrested in the presence of the drug, a rare subset can re-enter the cell cycle under constitutive drug treatment. Little is known about the non-genetic mechanisms that enable cancer persisters to maintain proliferative capacity in the presence of drugs. To study this rare, transiently resistant, proliferative persister population, we developed Watermelon, a high-complexity expressed barcode lentiviral library for simultaneous tracing of each cell's clonal origin and proliferative and transcriptional states. Here we show that cycling and non-cycling persisters arise from different cell lineages with distinct transcriptional and metabolic programs. Upregulation of antioxidant gene programs and a metabolic shift to fatty acid oxidation are associated with persister proliferative capacity across multiple cancer types. Impeding oxidative stress or metabolic reprogramming alters the fraction of cycling persisters. In human tumours, programs associated with cycling persisters are induced in minimal residual disease in response to multiple targeted therapies. The Watermelon system enabled the identification of rare persister lineages that are preferentially poised to proliferate under drug pressure, thus exposing new vulnerabilities that can be targeted to delay or even prevent disease recurrence.


Subject(s)
Cell Cycle , Cell Lineage , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasms/drug therapy , Neoplasms/pathology , Antioxidants/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Clone Cells/drug effects , Clone Cells/metabolism , Clone Cells/pathology , DNA Barcoding, Taxonomic , Fatty Acids/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lentivirus/genetics , Neoplasm Recurrence, Local/genetics , Neoplasms/genetics , Neoplasms/metabolism , Oncogene Proteins/antagonists & inhibitors , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism , Transcription, Genetic/drug effects
9.
Leuk Res ; 110: 106663, 2021 11.
Article in English | MEDLINE | ID: mdl-34304129

ABSTRACT

In silico simulation of pre-clinical and clinical data may accelerate pre-clinical and clinical trial advances, leading to benefits for therapeutic outcomes, toxicity and cost savings. Combining this with clonal architecture data may permit truly personalized therapy. Chronic lymphocytic leukemia (CLL) exhibits clonal diversity, evolution and selection, spontaneously and under treatment pressure. We apply a dynamic simulation model to published CLL clonal architecture data to explore alternative therapeutic strategies, focusing on BTK inhibition. By deriving parameters of clonal growth and death behavior we model continuous vs time-limited ibrutinib therapy, and find that, despite persistence of disease, time to clinical progression may not differ. This is a testable hypothesis. We model IgVH-mutated CLL vs unmutated CLL by varying proliferation and find, based on the limited available data about clonal dynamics after such therapy, that there are differences predicted in response to anti-CD20 efficacy. These models can suggest potential clinical trials, and also indicate what additional data are needed to improve predictions. Ongoing work will expand modeling to agents such as venetoclax and to T cell therapies.


Subject(s)
Adenine/analogs & derivatives , Clone Cells/pathology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Mutation , Piperidines/therapeutic use , Rituximab/therapeutic use , Adenine/therapeutic use , Antineoplastic Agents, Immunological/standards , Antineoplastic Agents, Immunological/therapeutic use , Clonal Evolution , Clone Cells/drug effects , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Prognosis
10.
PLoS One ; 16(7): e0254805, 2021.
Article in English | MEDLINE | ID: mdl-34310625

ABSTRACT

The purpose of the study was to characterize the resistome, virulome, mobilome and Clustered Regularly Interspaced Short Palindromic Repeats-associated (CRISPR-Cas) system of extended-spectrum ß-lactamase producing Klebsiella pneumoniae (ESBL-KP) clinical isolates and to determine their phylogenetic relatedness. The isolates were from Algeria, isolated at the University Hospital Establishment of Oran, between 2011 and 2012. ESBL-KP isolates (n = 193) were screened for several antibiotic resistance genes (ARGs) using qPCR followed by Pulsed-Field Gel Electrophoresis (PFGE). Representative isolates were selected from PFGE clusters and subjected to whole-genome sequencing (WGS). Genomic characterization of the WGS data by studying prophages, CRISPR-Cas systems, Multi-Locus Sequence Typing (MLST), serotype, ARGs, virulence genes, plasmid replicons, and their pMLST. Phylogenetic and comparative genomic were done using core genome MLST and SNP-Based analysis. Generally, the ESBL-KP isolates were polyclonal. The whole genome sequences of nineteen isolates were taken of main PFGE clusters. Sixteen sequence types (ST) were found including high-risk clones ST14, ST23, ST37, and ST147. Serotypes K1 (n = 1), K2 (n = 2), K3 (n = 1), K31 (n = 1), K62 (n = 1), and K151 (n = 1) are associated with hyper-virulence. CRISPR-Cas system was found in 47.4%, typed I-E and I-E*. About ARGs, from 193 ESBL-KP, the majority of strains were multidrug-resistant, the CTX-M-1 enzyme was predominant (99%) and the prevalence of plasmid-mediated quinolone resistance (PMQR) genes was high with aac(6')-lb-cr (72.5%) and qnr's (65.8%). From 19 sequenced isolates we identified ESBL, AmpC, and carbapenemase genes: blaCTX-M-15 (n = 19), blaOXA-48 (n = 1), blaCMY-2 (n = 2), and blaCMY-16 (n = 2), as well as non-ESBL genes: qnrB1 (n = 12), qnrS1 (n = 1) and armA (n = 2). We found IncF, IncN, IncL/M, IncA/C2, and Col replicon types, at least once per isolate. This study is the first to report qnrS in ESBL-KP in Algeria. Our analysis shows the concerning co-existence of virulence and resistance genes and would support that genomic surveillance should be a high priority in the hospital environment.


Subject(s)
Bacterial Proteins/genetics , Klebsiella Infections/drug therapy , Klebsiella pneumoniae/enzymology , beta-Lactamases/genetics , Algeria , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Clone Cells/cytology , Clone Cells/drug effects , Drug Resistance, Multiple, Bacterial/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Genome, Bacterial/genetics , Hospitals, University , Humans , Klebsiella Infections/enzymology , Klebsiella Infections/microbiology , Klebsiella Infections/pathology , Klebsiella pneumoniae/pathogenicity , Microbial Sensitivity Tests , Multilocus Sequence Typing , Phylogeny , Plasmids/drug effects , Whole Genome Sequencing , beta-Lactamases/metabolism
11.
Front Immunol ; 12: 659625, 2021.
Article in English | MEDLINE | ID: mdl-33912187

ABSTRACT

Aberrant T-cell function is implicated in the pathogenesis of myelodysplastic syndrome (MDS). Monitoring the T-cell receptor (TCR) repertoire can provide insights into T-cell adaptive immunity. Previous studies found skewed TCR repertoires in MDS compared to healthy patients; however these studies that leverage mRNA-based spectratyping have limitations. Furthermore, evaluating the TCR repertoire in context of hypomethylating agents (HMAs) treatment can provide insights into the dynamics of T-cell mediated responses in MDS. We conducted immunosequencing of the CDR3 regions of TCRß chains in bone marrows of 11 MDS patients prior to treatment (n=11 bone marrows prior to treatment), and in at least 2 timepoints for each patient following treatment (n=26 bone marrow aspirates post-treatment) with (HMA), alongside analyzing bone marrows from 4 healthy donors as controls. TCR repertoires in MDS patients were more clonal and less diverse than healthy donors. However, unlike previous reports, we did not observe significant skewness in CDR3 length or spectratyping. The global metrics of TCR profiling including richness, clonality, overlaps were not significantly changed in responders or non-responders following treatment with HMAs. However, we found an emergence of novel clonotypes in MDS patients who responded to treatment, while non-responders had a higher frequency of contracted clonotypes following treatment. By applying GLIPH2 for antigen prediction, we found rare TCR specificity clusters shared by TCR clonotypes from different patients at pre- or following treatment. Our data show clear differences in TCR repertoires of MDS compared with healthy patients and that novel TCR clonotype emergence in response to HMA therapy was correlated with response. This suggests that response to HMA therapy may be partially driven by T-cell mediated immunity and that the immune-based therapies, which target the adaptive immune system, may play a significant role in select patients with MDS.


Subject(s)
Azacitidine/therapeutic use , DNA Methylation/drug effects , Decitabine/therapeutic use , Myelodysplastic Syndromes/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Aged , Aged, 80 and over , Clone Cells/drug effects , Clone Cells/immunology , Clone Cells/metabolism , Cohort Studies , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/immunology , Polymerase Chain Reaction/methods , Receptors, Antigen, T-Cell, alpha-beta/immunology , Sequence Analysis, DNA/methods , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome
12.
Ann Clin Microbiol Antimicrob ; 20(1): 19, 2021 Mar 20.
Article in English | MEDLINE | ID: mdl-33743736

ABSTRACT

BACKGROUND: Acinetobacter baumannii has emerged as one of the common multidrug resistance pathogens causing hospital-acquired infections. This study was conducted to elucidate the distribution of antimicrobial resistance genes in the bacterial population in Thailand. Multidrug-resistant A. baumannii (MDR A. baumannii) isolates were characterized phenotypically, and the molecular epidemiology of clinical isolates in 11 tertiary hospitals was investigated at a country-wide level. METHODS: A total of 135 nonrepetitive MDR A. baumannii isolates collected from tertiary care hospitals across 5 regions of Thailand were examined for antibiotic susceptibility, resistance genes, and sequence types. Multilocus sequence typing (MLST) was performed to characterize the spread of regional lineages. RESULTS: ST2 belonging to IC2 was the most dominant sequence type in Thailand (65.19%), and to a lesser extent, there was also evidence of the spread of ST164 (10.37%), ST129 (3.70%), ST16 (2.96%), ST98 (2.96%), ST25 (2.96%), ST215 (2.22%), ST338 (1.48%), and ST745 (1.48%). The novel sequence types ST1551, ST1552, ST1553, and ST1557 were also identified in this study. Among these, the blaoxa-23 gene was by far the most widespread in MDR A. baumannii, while the blaoxa-24/40 and blaoxa-58 genes appeared to be less dominant in this region. The results demonstrated that the predominant class D carbapenemase was blaOXA-23, followed by the class B carbapenemase blaNDM-like, while the mcr-1 gene was not observed in any isolate. Most of the MDR A. baumannii isolates were resistant to ceftazidime (99.23%), gentamicin (91.85%), amikacin (82.96%), and ciprofloxacin (97.78%), while all of them were resistant to carbapenems. The results suggested that colistin could still be effective against MDR A. baumannii in this region. CONCLUSION: This is the first molecular epidemiological analysis of MDR A. baumannii clinical isolates at the national level in Thailand to date. Studies on the clonal relatedness of MDR A. baumannii isolates could generate useful data to understand the local epidemiology and international comparisons of nosocomial outbreaks.


Subject(s)
Acinetobacter Infections/microbiology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/isolation & purification , Anti-Bacterial Agents/pharmacology , Clone Cells/drug effects , Drug Resistance, Multiple, Bacterial , Molecular Epidemiology , Acinetobacter baumannii/genetics , Bacterial Proteins , Carbapenems/pharmacology , Ciprofloxacin/pharmacology , Colistin/pharmacology , Cross Infection/microbiology , Drug Resistance, Multiple, Bacterial/genetics , Humans , Microbial Sensitivity Tests , Multilocus Sequence Typing , Thailand , beta-Lactamases
13.
Infect Disord Drug Targets ; 21(3): 459-463, 2021.
Article in English | MEDLINE | ID: mdl-32389116

ABSTRACT

BACKGROUND: Methicillin-resistant staphylococcus capitis (MRSC) NRCS-A clone (Multi- resistant and vancomycin-non susceptible) has been recently described as an emerging cause of nosocomial bacteremia, especially in neonatal intensive-care units (NICUs). OBJECTIVE: The objective of this study was to evaluate the antibiotic and antiseptic resistance patterns, biofilm-producing ability and the prevalence of SCCmec and ACME types among MRSC isolates as well as to check the possible presence of NRCS-A clone at Tehran's Children's Medical Center, Iran. METHODS: A total of 256 coagulase-negative Staphylococcal isolates were collected, of which 10 S. capitis isolates were obtained and tested for susceptibility against 13 antimicrobial and 3 antiseptic agents, as well as biofilm production. The presence of 15 distinct resistance genes, staphylococcal cassette chromosome mec (SCCmec), and arginine catabolic mobile elements (ACMEs) were tracked. RESULTS: Seven out of 10 S. capitis isolates were MRSC (MIC90 van=8µg/mL) and resistant to trimethoprim/sulfamethoxazole, produced biofilm, (3 as strong biofilm producers) and carried ACME types I and II. Despite the identification of mec and ccr complexes in some isolates, all the SCCmec cassettes were untypeable (UT). CONCLUSION: According to the studied features, only one isolate belonged to the NRSC-A clone. The results indicate that MRSC with high antibiotic resistance and unknown SCCmec might become a serious problem in the future for the treatment of patients, particularly children.


Subject(s)
Staphylococcal Infections , Staphylococcus capitis , Anti-Bacterial Agents/pharmacology , Child , Clone Cells/drug effects , Humans , Infant, Newborn , Iran/epidemiology , Methicillin Resistance/drug effects , Methicillin Resistance/genetics , Microbial Sensitivity Tests , Staphylococcal Infections/epidemiology , Staphylococcus capitis/drug effects
14.
Nutrients ; 12(12)2020 Nov 29.
Article in English | MEDLINE | ID: mdl-33260329

ABSTRACT

Isoquercitrin (IQ), a major flavonol present in Prunus mume fruit, has gained much attention in recent studies because of its superior bioavailability and physiological effects. In this study, the anti-cancer mechanism of IQ against human melanoma, particularly its effect on the mitochondria-mediated apoptosis, was investigated. Treatment with IQ at 25 µM concentration effectively inhibited the proliferation of SK-MEL-2 skin cancer cells while the same concentration did not exhibit cytotoxicity against human keratinocytes HaCaT. Morphological analysis and clonogenic assay also showed that IQ can alter the growth and long-term survival of SK-MEL-2 cells. IQ also induced apoptosis in the melanoma cells as manifested in the nuclear morphology changes, DNA fragmentation, increase in the apoptosis rate (17.69% at 25 µM) and accumulation of sub-G1 cell cycle phase population (19.55% at 25 µM). Western blot analysis revealed the involvement of the mitochondrial apoptosis signaling pathway in the anti-cancer property of IQ. Treatment with IQ resulted in the decrease in the levels of procaspase-8 and -9, and Bcl-2 protein, and an increase in the expression of cleaved PARP and Bax. Moreover, AIF and Endo G protein expression increased, indicating a caspase-independent mitochondrial-mediated apoptosis. The anti-proliferative activity of IQ against SK-MEL-2 can also be attributed to the downregulation of the PI3K/AktmTOR signaling pathway. These findings showed that IQ can be developed into a chemopreventive therapeutic agent against the melanoma cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Melanoma/pathology , Mitochondria/metabolism , Quercetin/analogs & derivatives , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Clone Cells/drug effects , Down-Regulation , Humans , Melanoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quercetin/pharmacology , Signal Transduction/drug effects , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism
15.
Genome Med ; 12(1): 104, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33239092

ABSTRACT

BACKGROUND: Multidrug-resistant (MDR) Mycobacterium tuberculosis complex strains not detected by commercial molecular drug susceptibility testing (mDST) assays due to the RpoB I491F resistance mutation are threatening the control of MDR tuberculosis (MDR-TB) in Eswatini. METHODS: We investigate the evolution and spread of MDR strains in Eswatini with a focus on bedaquiline (BDQ) and clofazimine (CFZ) resistance using whole-genome sequencing in two collections ((1) national drug resistance survey, 2009-2010; (2) MDR strains from the Nhlangano region, 2014-2017). RESULTS: MDR strains in collection 1 had a high cluster rate (95%, 117/123 MDR strains) with 55% grouped into the two largest clusters (gCL3, n = 28; gCL10, n = 40). All gCL10 isolates, which likely emerged around 1993 (95% highest posterior density 1987-1998), carried the mutation RpoB I491F that is missed by commercial mDST assays. In addition, 21 (53%) gCL10 isolates shared a Rv0678 M146T mutation that correlated with elevated minimum inhibitory concentrations (MICs) to BDQ and CFZ compared to wild type isolates. gCL10 isolates with the Rv0678 M146T mutation were also detected in collection 2. CONCLUSION: The high clustering rate suggests that transmission has been driving the MDR-TB epidemic in Eswatini for three decades. The presence of MDR strains in Eswatini that are not detected by commercial mDST assays and have elevated MICs to BDQ and CFZ potentially jeopardizes the successful implementation of new MDR-TB treatment guidelines. Measures to limit the spread of these outbreak isolates need to be implemented urgently.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , Diarylquinolines/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Tuberculosis, Multidrug-Resistant/genetics , Clone Cells/drug effects , Disease Outbreaks , Eswatini , Humans , Microbial Sensitivity Tests , Mutation , Tuberculosis, Multidrug-Resistant/microbiology
16.
Genome Res ; 30(12): 1846-1855, 2020 12.
Article in English | MEDLINE | ID: mdl-33203764

ABSTRACT

The levels and subcellular localizations of proteins regulate critical aspects of many cellular processes and can become targets of therapeutic intervention. However, high-throughput methods for the discovery of proteins that change localization either by shuttling between compartments, by binding larger complexes, or by localizing to distinct membraneless organelles are not available. Here we describe a scalable strategy to characterize effects on protein localizations and levels in response to different perturbations. We use CRISPR-Cas9-based intron tagging to generate cell pools expressing hundreds of GFP-fusion proteins from their endogenous promoters and monitor localization changes by time-lapse microscopy followed by clone identification using in situ sequencing. We show that this strategy can characterize cellular responses to drug treatment and thus identify nonclassical effects such as modulation of protein-protein interactions, condensate formation, and chemical degradation.


Subject(s)
Clone Cells/drug effects , Proteins/metabolism , Sequence Analysis, DNA/methods , Time-Lapse Imaging/methods , CRISPR-Cas Systems , Clone Cells/metabolism , Gene Editing , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Molecular Imaging , Pharmaceutical Preparations , Protein Transport/drug effects , Proteins/drug effects
17.
Exp Hematol ; 91: 46-54, 2020 11.
Article in English | MEDLINE | ID: mdl-32946982

ABSTRACT

Clonal heterogeneity fuels leukemia evolution, therapeutic resistance, and relapse. Upfront detection of therapy-resistant leukemia clones at diagnosis may allow adaptation of treatment and prevention of relapse, but this is hampered by a paucity of methods to identify and trace single leukemia-propagating cells and their clonal offspring. Here, we tested methods of cellular barcoding analysis, to trace the in vivo competitive dynamics of hundreds of patient-derived leukemia clones upon chemotherapy-mediated selective pressure. We transplanted Nod/Scid/Il2Rγ-/- (NSG) mice with barcoded patient-derived or SupB15 acute lymphoblastic leukemia (ALL) cells and assessed clonal responses to dexamethasone, methotrexate, and vincristine, longitudinally and across nine anatomic locations. We illustrate that chemotherapy reduces clonal diversity in a drug-dependent manner. At end-stage disease, methotrexate-treated patient-derived xenografts had significantly fewer clones compared with placebo-treated mice (100 ± 10 vs. 160 ± 15 clones, p = 0.0005), while clonal complexity in vincristine- and dexamethasone-treated xenografts was unaffected (115 ± 33 and 150 ± 7 clones, p = NS). Using tools developed to assess differential gene expression, we determined whether these clonal patterns resulted from random clonal drift or selection. We identified 5 clones that were reproducibly enriched in methotrexate-treated patient-derived xenografts, suggestive of pre-existent resistance. Finally, we found that chemotherapy-mediated selection resulted in a more asymmetric distribution of leukemia clones across anatomic sites. We found that cellular barcoding is a powerful method to trace the clonal dynamics of human patient-derived leukemia cells in response to chemotherapy. In the future, integration of cellular barcoding with single-cell sequencing technology may allow in-depth characterization of therapy-resistant leukemia clones and identify novel targets to prevent relapse.


Subject(s)
Clone Cells/drug effects , DNA Barcoding, Taxonomic , Drug Resistance, Neoplasm , Leukemia, B-Cell/pathology , Neoplastic Stem Cells/drug effects , Adolescent , Animals , DNA, Neoplasm/genetics , Dexamethasone/pharmacology , Heterografts , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Methotrexate/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Selection, Genetic , Single-Cell Analysis , Vincristine/pharmacology
18.
Nature ; 584(7820): 268-273, 2020 08.
Article in English | MEDLINE | ID: mdl-32728211

ABSTRACT

The ability of the skin to grow in response to stretching has been exploited in reconstructive surgery1. Although the response of epidermal cells to stretching has been studied in vitro2,3, it remains unclear how mechanical forces affect their behaviour in vivo. Here we develop a mouse model in which the consequences of stretching on skin epidermis can be studied at single-cell resolution. Using a multidisciplinary approach that combines clonal analysis with quantitative modelling and single-cell RNA sequencing, we show that stretching induces skin expansion by creating a transient bias in the renewal activity of epidermal stem cells, while a second subpopulation of basal progenitors remains committed to differentiation. Transcriptional and chromatin profiling identifies how cell states and gene-regulatory networks are modulated by stretching. Using pharmacological inhibitors and mouse mutants, we define the step-by-step mechanisms that control stretch-mediated tissue expansion at single-cell resolution in vivo.


Subject(s)
Mechanotransduction, Cellular/physiology , Single-Cell Analysis , Skin/cytology , Skin/growth & development , Adaptor Proteins, Signal Transducing/metabolism , Adherens Junctions/metabolism , Animals , Base Sequence , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Self Renewal/drug effects , Chromatin/drug effects , Chromatin/genetics , Chromatin Assembly and Disassembly/drug effects , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Regulatory Networks/drug effects , Hydrogels/administration & dosage , Hydrogels/pharmacology , Mechanotransduction, Cellular/drug effects , Mechanotransduction, Cellular/genetics , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , RNA, Messenger/genetics , RNA-Seq , Skin/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic/drug effects , YAP-Signaling Proteins
20.
Blood ; 136(1): 36-49, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32430502

ABSTRACT

Acquired aplastic anemia (AA) and paroxysmal nocturnal hemoglobinuria (PNH) are pathogenically related nonmalignant bone marrow failure disorders linked to T-cell-mediated autoimmunity; they are associated with an increased risk of secondary myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Approximately 15% to 20% of AA patients and 2% to 6% of PNH patients go on to develop secondary MDS/AML by 10 years of follow-up. Factors determining an individual patient's risk of malignant transformation remain poorly defined. Recent studies identified nearly ubiquitous clonal hematopoiesis (CH) in AA patients. Similarly, CH with additional, non-PIGA, somatic alterations occurs in the majority of patients with PNH. Factors associated with progression to secondary MDS/AML include longer duration of disease, increased telomere attrition, presence of adverse prognostic mutations, and multiple mutations, particularly when occurring early in the disease course and at a high allelic burden. Here, we will review the prevalence and characteristics of somatic alterations in AA and PNH and will explore their prognostic significance and mechanisms of clonal selection. We will then discuss the available data on post-AA and post-PNH progression to secondary MDS/AML and provide practical guidance for approaching patients with PNH and AA who have CH.


Subject(s)
Anemia, Aplastic/pathology , Hemoglobinuria, Paroxysmal/pathology , Leukemia, Myeloid, Acute/etiology , Myelodysplastic Syndromes/etiology , Age of Onset , Anemia, Aplastic/drug therapy , Anemia, Aplastic/genetics , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Benzoates/adverse effects , Benzoates/therapeutic use , Bone Marrow/pathology , Chromosome Aberrations , Chromosomes, Human, Pair 7/genetics , Clonal Evolution/drug effects , Clone Cells/drug effects , Clone Cells/pathology , Disease Progression , Granulocyte Colony-Stimulating Factor/adverse effects , Granulocyte Colony-Stimulating Factor/therapeutic use , Hemoglobinuria, Paroxysmal/drug therapy , Hemoglobinuria, Paroxysmal/genetics , Humans , Hydrazines/adverse effects , Hydrazines/therapeutic use , Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Models, Biological , Monosomy , Mutation , Myelodysplastic Syndromes/epidemiology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Oncogene Proteins, Fusion/genetics , Pyrazoles/adverse effects , Pyrazoles/therapeutic use , Selection, Genetic , Telomere Shortening
SELECTION OF CITATIONS
SEARCH DETAIL
...