Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 10 de 10
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
PLoS Biol ; 22(8): e3002776, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39163475

RÉSUMÉ

The ultraviolet (UV) radiation triggers a pigmentation response in human skin, wherein, melanocytes rapidly activate divergent maturation and proliferation programs. Using single-cell sequencing, we demonstrate that these 2 programs are segregated in distinct subpopulations in melanocytes of human and zebrafish skin. The coexistence of these 2 cell states in cultured melanocytes suggests possible cell autonomy. Luria-Delbrück fluctuation test reveals that the initial establishment of these states is stochastic. Tracking of pigmenting cells ascertains that the stochastically acquired state is faithfully propagated in the progeny. A systemic approach combining single-cell multi-omics (RNA+ATAC) coupled to enhancer mapping with H3K27 acetylation successfully identified state-specific transcriptional networks. This comprehensive analysis led to the construction of a gene regulatory network (GRN) that under the influence of noise, establishes a bistable system of pigmentation and proliferation at the population level. This GRN recapitulates melanocyte behaviour in response to external cues that reinforce either of the states. Our work highlights that inherent stochasticity within melanocytes establishes dedicated states, and the mature state is sustained by selective enhancers mark through histone acetylation. While the initial cue triggers a proliferation response, the continued signal activates and maintains the pigmenting subpopulation via epigenetic imprinting. Thereby our study provides the basis of coexistence of distinct populations which ensures effective pigmentation response while preserving the self-renewal capacity.

2.
ACS Org Inorg Au ; 4(3): 319-328, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38855338

RÉSUMÉ

As the SIRTi analogue series (HL1-HL6) show potent antitumor activity in vitro, we synthesized their corresponding zinc(II) complexes (ZnL1-ZnL6) and investigated their potential as anticancer agents. The Zn(II) complexes showed substantially greater cytotoxicity than HL1-HL6 alone in several cancer cell-types. Notably, distinct structure-activity relationships confirmed the significance of tert-butyl (ZnL2) pharmacophore inclusion in their activity. ZnL2 complexes were found to transmetalate with copper ions inside cells, causing the formation of redox-active copper complexes that induced reactive oxygen species (ROS) production, mitochondrial membrane depolarization, ATP decay, and cell death. This is the first study to exhibit Zn(II) complexes that mediate their activity via transmetalation with copper ions to undergo paraptosis cell death pathway. To further confirm if the SIRT1/2 inhibitory property of SIRTi analogues is conserved, a docking simulation study is performed. The binding affinity and specific interactions of the Cu(II) complex obtained after transmetalation with ZnL2 were found to be higher for SIRT2 (K i = 0.06 µM) compared to SIRT1 (K i = 0.25 µM). Thus, the concurrent regulation of several biological targets using a single drug has been shown to have synergistic therapeutic effects, which are crucial for the effective treatment of cancer.

3.
Commun Biol ; 6(1): 1292, 2023 12 21.
Article de Anglais | MEDLINE | ID: mdl-38129585

RÉSUMÉ

Intra-tumor heterogeneity contributes to treatment failure and poor survival in urothelial bladder carcinoma (UBC). Analyzing transcriptome from a UBC cohort, we report that intra-tumor transcriptomic heterogeneity indicates co-existence of tumor cells in epithelial and mesenchymal-like transcriptional states and bi-directional transition between them occurs within and between tumor subclones. We model spontaneous and reversible transition between these partially heritable states in cell lines and characterize their population dynamics. SMAD3, KLF4 and PPARG emerge as key regulatory markers of the transcriptional dynamics. Nutrient limitation, as in the core of large tumors, and radiation treatment perturb the dynamics, initially selecting for a transiently resistant phenotype and then reconstituting heterogeneity and growth potential, driving adaptive evolution. Dominance of transcriptional states with low PPARG expression indicates an aggressive phenotype in UBC patients. We propose that phenotypic plasticity and dynamic, non-genetic intra-tumor heterogeneity modulate both the trajectory of disease progression and adaptive treatment response in UBC.


Sujet(s)
Carcinome transitionnel , Tumeurs de la vessie urinaire , Humains , Vessie urinaire , Récepteur PPAR gamma , Tumeurs de la vessie urinaire/thérapie , Carcinome transitionnel/anatomopathologie , Évolution de la maladie
4.
Biophys J ; 122(19): 3909-3923, 2023 Oct 03.
Article de Anglais | MEDLINE | ID: mdl-37598292

RÉSUMÉ

In the epithelium, cell density and cell proliferation are closely connected to each other through contact inhibition of proliferation (CIP). Depending on cell density, CIP proceeds through three distinct stages: the free-growing stage at low density, the pre-epithelial transition stage at medium density, and the post-epithelial transition stage at high density. Previous studies have elucidated how cell morphology, motion, and mechanics vary in these stages. However, it remains unknown whether cellular metabolism also has a density-dependent behavior. By measuring the mitochondrial membrane potential at different cell densities, here we reveal a heterogeneous landscape of metabolism in the epithelium, which appears qualitatively distinct in three stages of CIP and did not follow the trend of other CIP-associated parameters, which increases or decreases monotonically with increasing cell density. Importantly, epithelial cells established a collective metabolic heterogeneity exclusively in the pre-epithelial transition stage, where the multicellular clusters of high- and low-potential cells emerged. However, in the post-epithelial transition stage, the metabolic potential field became relatively homogeneous. Next, to study the underlying dynamics, we constructed a system biology model, which predicted the role of cell proliferation in metabolic potential toward establishing collective heterogeneity. Further experiments indeed revealed that the metabolic pattern spatially correlated with the proliferation capacity of cells, as measured by the nuclear localization of a pro-proliferation protein, YAP. Finally, experiments perturbing the actomyosin contractility revealed that, while metabolic heterogeneity was maintained in the absence of actomyosin contractility, its ab initio emergence depended on the latter. Taken together, our results revealed a density-dependent collective heterogeneity in the metabolic field of a pre-epithelial transition-stage epithelial monolayer, which may have significant implications for epithelial form and function.


Sujet(s)
Actomyosine , Inhibition de contact , Actomyosine/métabolisme , Cellules épithéliales/métabolisme , Épithélium/métabolisme , Prolifération cellulaire
5.
iScience ; 26(7): 106964, 2023 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-37426354

RÉSUMÉ

The Epithelial-to-Mesenchymal Transition (EMT) is a hallmark of cancer metastasis and morbidity. EMT is a non-binary process, and cells can be stably arrested en route to EMT in an intermediate hybrid state associated with enhanced tumor aggressiveness and worse patient outcomes. Understanding EMT progression in detail will provide fundamental insights into the mechanisms underlying metastasis. Despite increasingly available single-cell RNA sequencing (scRNA-seq) data that enable in-depth analyses of EMT at the single-cell resolution, current inferential approaches are limited to bulk microarray data. There is thus a great need for computational frameworks to systematically infer and predict the timing and distribution of EMT-related states at single-cell resolution. Here, we develop a computational framework for reliable inference and prediction of EMT-related trajectories from scRNA-seq data. Our model can be utilized across a variety of applications to predict the timing and distribution of EMT from single-cell sequencing data.

6.
Drugs ; 78(16): 1717-1740, 2018 Nov.
Article de Anglais | MEDLINE | ID: mdl-30392114

RÉSUMÉ

Chronic obstructive pulmonary disease (COPD) and lung cancer are major lung diseases affecting millions worldwide. Both diseases have links to cigarette smoking and exert a considerable societal burden. People suffering from COPD are at higher risk of developing lung cancer than those without, and are more susceptible to poor outcomes after diagnosis and treatment. Lung cancer and COPD are closely associated, possibly sharing common traits such as an underlying genetic predisposition, epithelial and endothelial cell plasticity, dysfunctional inflammatory mechanisms including the deposition of excessive extracellular matrix, angiogenesis, susceptibility to DNA damage and cellular mutagenesis. In fact, COPD could be the driving factor for lung cancer, providing a conducive environment that propagates its evolution. In the early stages of smoking, body defences provide a combative immune/oxidative response and DNA repair mechanisms are likely to subdue these changes to a certain extent; however, in patients with COPD with lung cancer the consequences could be devastating, potentially contributing to slower postoperative recovery after lung resection and increased resistance to radiotherapy and chemotherapy. Vital to the development of new-targeted therapies is an in-depth understanding of various molecular mechanisms that are associated with both pathologies. In this comprehensive review, we provide a detailed overview of possible underlying factors that link COPD and lung cancer, and current therapeutic advances from both human and preclinical animal models that can effectively mitigate this unholy relationship.


Sujet(s)
Tumeurs du poumon , Broncho-pneumopathie chronique obstructive , Animaux , Association thérapeutique , Humains , Tumeurs du poumon/physiopathologie , Tumeurs du poumon/thérapie , Thérapie moléculaire ciblée , Stress oxydatif , Broncho-pneumopathie chronique obstructive/étiologie , Broncho-pneumopathie chronique obstructive/physiopathologie , Broncho-pneumopathie chronique obstructive/thérapie , Fumer/effets indésirables
7.
J R Soc Interface ; 14(136)2017 11.
Article de Anglais | MEDLINE | ID: mdl-29187638

RÉSUMÉ

Epithelial-mesenchymal transition (EMT) plays key roles during embryonic development, wound healing and cancer metastasis. Cells in a partial EMT or hybrid epithelial/mesenchymal (E/M) phenotype exhibit collective cell migration, forming clusters of circulating tumour cells-the primary drivers of metastasis. Activation of cell-cell signalling pathways such as Notch fosters a partial or complete EMT, yet the mechanisms enabling cluster formation remain poorly understood. Using an integrated computational-experimental approach, we examine the role of Numb-an inhibitor of Notch intercellular signalling-in mediating EMT and clusters formation. We show via an mathematical model that Numb inhibits a full EMT by stabilizing a hybrid E/M phenotype. Consistent with this observation, knockdown of Numb in stable hybrid E/M cells H1975 results in a full EMT, thereby showing that Numb acts as a brake for a full EMT and thus behaves as a 'phenotypic stability factor' by modulating Notch-driven EMT. By generalizing the mathematical model to a multi-cell level, Numb is predicted to alter the balance of hybrid E/M versus mesenchymal cells in clusters, potentially resulting in a higher tumour-initiation ability. Finally, Numb correlates with a worse survival in multiple independent lung and ovarian cancer datasets, hence confirming its relationship with increased cancer aggressiveness.


Sujet(s)
Transition épithélio-mésenchymateuse , Protéines membranaires/physiologie , Protéines de tissu nerveux/physiologie , Récepteurs Notch/métabolisme , Communication cellulaire , Lignée cellulaire , Mouvement cellulaire , Biologie informatique , Humains , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Modèles théoriques , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme
8.
Cancer Res ; 77(16): 4414-4425, 2017 08 15.
Article de Anglais | MEDLINE | ID: mdl-28646020

RÉSUMÉ

Despite favorable responses to initial therapy, small-cell lung cancer (SCLC) relapse occurs within a year and exhibits resistance to multiple drugs. Because of limited accessibility of patient tissues for research purposes, SCLC patient-derived xenografts (PDX) have provided the best opportunity to address this limitation. Here, we sought to identify novel mechanisms involved in SCLC chemoresistance. Through in-depth proteomic profiling, we identified MCAM as a markedly upregulated surface receptor in chemoresistant SCLC cell lines and in chemoresistant PDX compared with matched treatment-naïve tumors. MCAM depletion in chemoresistant cells reduced cell proliferation and reduced the IC50 inhibitory concentration of chemotherapeutic drugs in vitro This MCAM-mediated sensitization to chemotherapy occurred via SOX2-dependent upregulation of mitochondrial 37S ribosomal protein 1/ATP-binding cassette subfamily C member 1 (MRP1/ABCC1) and the PI3/AKT pathway. Metabolomic profiling revealed that MCAM modulated lactate production in chemoresistant cells that exhibit a distinct metabolic phenotype characterized by low oxidative phosphorylation. Our results suggest that MCAM may serve as a novel therapeutic target to overcome chemoresistance in SCLC. Cancer Res; 77(16); 4414-25. ©2017 AACR.


Sujet(s)
Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Facteurs de transcription SOX-B1/métabolisme , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Carcinome pulmonaire à petites cellules/métabolisme , Animaux , Antigènes CD146/génétique , Antigènes CD146/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/physiologie , Résistance aux médicaments antinéoplasiques , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Souris , Souris de lignée BALB C , Souris nude , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Facteurs de transcription SOX-B1/génétique , Transduction du signal , Carcinome pulmonaire à petites cellules/génétique , Carcinome pulmonaire à petites cellules/anatomopathologie
9.
J Vis Exp ; (122)2017 04 07.
Article de Anglais | MEDLINE | ID: mdl-28448023

RÉSUMÉ

Phenotypic plasticity refers to a phenomenon in which cells transiently gain traits of another lineage. During carcinoma progression, phenotypic plasticity drives invasion, dissemination and metastasis. Indeed, while most of the studies of phenotypic plasticity have been in the context of epithelial-derived carcinomas, it turns out sarcomas, which are mesenchymal in origin, also exhibit phenotypic plasticity, with a subset of sarcomas undergoing a phenomenon that resembles a mesenchymal-epithelial transition (MET). Here, we developed a method comprising the miR-200 family and grainyhead-like 2 (GRHL2) to mimic this MET-like phenomenon observed in sarcoma patient samples.We sequentially express GRHL2 and the miR-200 family using cell transduction and transfection, respectively, to better understand the molecular underpinnings of these phenotypic transitions in sarcoma cells. Sarcoma cells expressing miR-200s and GRHL2 demonstrated enhanced epithelial characteristics in cell morphology and alteration of epithelial and mesenchymal biomarkers. Future studies using these methods can be used to better understand the phenotypic consequences of MET-like processes on sarcoma cells, such as migration, invasion, metastatic propensity, and therapy resistance.


Sujet(s)
Protéines de liaison à l'ADN/métabolisme , Transition épithélio-mésenchymateuse/physiologie , microARN/métabolisme , Sarcomes/anatomopathologie , Facteurs de transcription/métabolisme , Numération cellulaire , Lignée cellulaire tumorale , Cellules épithéliales/anatomopathologie , Régulation de l'expression des gènes , Humains , Transfection
10.
Mol Cell Biol ; 36(19): 2503-13, 2016 10 01.
Article de Anglais | MEDLINE | ID: mdl-27402864

RÉSUMÉ

Phenotypic plasticity involves a process in which cells transiently acquire phenotypic traits of another lineage. Two commonly studied types of phenotypic plasticity are epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET). In carcinomas, EMT drives invasion and metastatic dissemination, while MET is proposed to play a role in metastatic colonization. Phenotypic plasticity in sarcomas is not well studied; however, there is evidence that a subset of sarcomas undergo an MET-like phenomenon. While the exact mechanisms by which these transitions occur remain largely unknown, it is likely that some of the same master regulators that drive EMT and MET in carcinomas also act in sarcomas. In this study, we combined mathematical models with bench experiments to identify a core regulatory circuit that controls MET in sarcomas. This circuit comprises the microRNA 200 (miR-200) family, ZEB1, and GRHL2. Interestingly, combined expression of miR-200s and GRHL2 further upregulates epithelial genes to induce MET. This effect is phenocopied by downregulation of either ZEB1 or the ZEB1 cofactor, BRG1. In addition, an MET gene expression signature is prognostic for improved overall survival in sarcoma patients. Together, our results suggest that a miR-200, ZEB1, GRHL2 gene regulatory network may drive sarcoma cells to a more epithelial-like state and that this likely has prognostic relevance.


Sujet(s)
Protéines de liaison à l'ADN/génétique , Transition épithélio-mésenchymateuse , microARN/génétique , Sarcomes/anatomopathologie , Facteurs de transcription/génétique , Facteur de transcription Zeb1/génétique , Régulation négative , Régulation de l'expression des gènes tumoraux , Humains , Modèles théoriques , Invasion tumorale , Métastase tumorale , Pronostic , Sarcomes/génétique , Analyse de survie , Régulation positive
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE