Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.900
Filtrer
1.
Cancer Immunol Immunother ; 73(9): 171, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38954021

RÉSUMÉ

In the intricate landscape of the tumor microenvironment, tumor-associated macrophages (TAMs) emerge as a ubiquitous cellular component that profoundly affects the oncogenic process. The microenvironment of hepatocellular carcinoma (HCC) is characterized by a pronounced infiltration of TAMs, underscoring their pivotal role in modulating the trajectory of the disease. Amidst the evolving therapeutic paradigms for HCC, the strategic reprogramming of metabolic pathways presents a promising avenue for intervention, garnering escalating interest within the scientific community. Previous investigations have predominantly focused on elucidating the mechanisms of metabolic reprogramming in cancer cells without paying sufficient attention to understanding how TAM metabolic reprogramming, particularly lipid metabolism, affects the progression of HCC. In this review article, we intend to elucidate how TAMs exert their regulatory effects via diverse pathways such as E2F1-E2F2-CPT2, LKB1-AMPK, and mTORC1-SREBP, and discuss correlations of TAMs with these processes and the characteristics of relevant pathways in HCC progression by consolidating various studies on TAM lipid uptake, storage, synthesis, and catabolism. It is our hope that our summary could delineate the impact of specific mechanisms underlying TAM lipid metabolic reprogramming on HCC progression and provide useful information for future research on HCC and the development of new treatment strategies.


Sujet(s)
Carcinome hépatocellulaire , Métabolisme lipidique , Tumeurs du foie , Microenvironnement tumoral , Macrophages associés aux tumeurs , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/immunologie , Humains , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/immunologie , Microenvironnement tumoral/immunologie , Animaux , Reprogrammation cellulaire , Transduction du signal ,
2.
Theranostics ; 14(10): 3793-3809, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994031

RÉSUMÉ

Rationale: CD8+ T cells undergo a series of metabolic reprogramming processes during their activation and proliferation, including increased glycolysis, decreased aerobic oxidation of sugars, increased amino acid metabolism and increased protein synthesis. However, it is still unclear what factors regulate these metabolic reprogramming processes in CD8+ T cells in the tumor immune microenvironment. Methods: T cell chromobox protein 4 (CBX4) knock-out mice models were used to determine the role of CBX4 in CD8+ T cells on the tumor immune microenvironment and tumor progression. Flow cytometry, Cut-Tag qPCR, Chip-seq, immunoprecipitation, metabolite detection, lentivirus infection and adoptive T cells transfer were performed to explore the underlying mechanisms of CBX4 knock-out in promoting CD8+ T cell activation and inhibiting tumor growth. Results: We found that CBX4 expression was induced in tumor-infiltrating CD8+ T cells and inhibited CD8+ T cell function by regulating glucose metabolism in tumor tissue. Mechanistically, CBX4 increases the expression of the metabolism-associated molecule aldolase B (Aldob) through sumoylation of trans-acting transcription factor 1 (SP1) and Krüppel-like factor 3 (KLF3). In addition, Aldob inhibits glycolysis and ATP synthesis in T cells by reducing the phosphorylation of the serine/threonine protein kinase (Akt) and ultimately suppresses CD8+ T cell function. Significantly, knocking out CBX4 may improve the efficacy of anti-PD-1 therapy by enhancing the function of CD8+ T cells in the tumor microenvironment. Conclusion: CBX4 is involved in CD8+ T cell metabolic reprogramming and functional persistence in tumor tissues, and serves as an inhibitor in CD8+ T cells' glycolysis and effector function.


Sujet(s)
Lymphocytes T CD8+ , Glycolyse , Souris knockout , Microenvironnement tumoral , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Souris , Microenvironnement tumoral/immunologie , Lignée cellulaire tumorale , Souris de lignée C57BL , Fructose bisphosphate aldolase/métabolisme , Fructose bisphosphate aldolase/génétique , Protéines du groupe Polycomb/métabolisme , Protéines du groupe Polycomb/génétique , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Humains , Reprogrammation cellulaire
3.
Nat Neurosci ; 27(7): 1260-1273, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38956165

RÉSUMÉ

Direct neuronal reprogramming is a promising approach to regenerate neurons from local glial cells. However, mechanisms of epigenome remodeling and co-factors facilitating this process are unclear. In this study, we combined single-cell multiomics with genome-wide profiling of three-dimensional nuclear architecture and DNA methylation in mouse astrocyte-to-neuron reprogramming mediated by Neurogenin2 (Ngn2) and its phosphorylation-resistant form (PmutNgn2), respectively. We show that Ngn2 drives multilayered chromatin remodeling at dynamic enhancer-gene interaction sites. PmutNgn2 leads to higher reprogramming efficiency and enhances epigenetic remodeling associated with neuronal maturation. However, the differences in binding sites or downstream gene activation cannot fully explain this effect. Instead, we identified Yy1, a transcriptional co-factor recruited by direct interaction with Ngn2 to its target sites. Upon deletion of Yy1, activation of neuronal enhancers, genes and ultimately reprogramming are impaired without affecting Ngn2 binding. Thus, our work highlights the key role of interactors of proneural factors in direct neuronal reprogramming.


Sujet(s)
Astrocytes , Facteurs de transcription à motif basique hélice-boucle-hélice , Reprogrammation cellulaire , Protéines de tissu nerveux , Neurones , Facteur de transcription YY1 , Animaux , Facteur de transcription YY1/métabolisme , Facteur de transcription YY1/génétique , Astrocytes/métabolisme , Souris , Reprogrammation cellulaire/physiologie , Neurones/métabolisme , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Épigénome , Assemblage et désassemblage de la chromatine , Épigenèse génétique , Cellules cultivées
4.
Cancer Res ; 84(14): 2297-2312, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39005053

RÉSUMÉ

Metabolic reprogramming is a hallmark of cancer and is crucial for cancer progression, making it an attractive therapeutic target. Understanding the role of metabolic reprogramming in cancer initiation could help identify prevention strategies. To address this, we investigated metabolism during acinar-to-ductal metaplasia (ADM), the first step of pancreatic carcinogenesis. Glycolytic markers were elevated in ADM lesions compared with normal tissue from human samples. Comprehensive metabolic assessment in three mouse models with pancreas-specific activation of KRAS, PI3K, or MEK1 using Seahorse measurements, nuclear magnetic resonance metabolome analysis, mass spectrometry, isotope tracing, and RNA sequencing analysis revealed a switch from oxidative phosphorylation to glycolysis in ADM. Blocking the metabolic switch attenuated ADM formation. Furthermore, mitochondrial metabolism was required for de novo synthesis of serine and glutathione (GSH) but not for ATP production. MYC mediated the increase in GSH intermediates in ADM, and inhibition of GSH synthesis suppressed ADM development. This study thus identifies metabolic changes and vulnerabilities in the early stages of pancreatic carcinogenesis. Significance: Metabolic reprogramming from oxidative phosphorylation to glycolysis mediated by MYC plays a crucial role in the development of pancreatic cancer, revealing a mechanism driving tumorigenesis and potential therapeutic targets. See related commentary by Storz, p. 2225.


Sujet(s)
Métaplasie , Tumeurs du pancréas , Animaux , Humains , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Souris , Métaplasie/métabolisme , Métaplasie/anatomopathologie , Glycolyse , Carcinogenèse/métabolisme , Cellules acineuses/métabolisme , Cellules acineuses/anatomopathologie , Phosphorylation oxydative , Glutathion/métabolisme , Reprogrammation cellulaire , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Mâle , Mitochondries/métabolisme , Mitochondries/anatomopathologie ,
5.
PeerJ ; 12: e17657, 2024.
Article de Anglais | MEDLINE | ID: mdl-39011384

RÉSUMÉ

Background: Our previous studies have successfully reported the reprogramming of fibroblasts into induced mammary epithelial cells (iMECs). However, the regulatory relationships and functional roles of MicroRNAs (miRNAs) in the progression of fibroblasts achieving the cell fate of iMECs are insufficiently understood. Methods: First, we performed pre-and post-induction miRNAs sequencing analysis by using high-throughput sequencing. Following that, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment studies were used to determine the primary roles of the significantly distinct miRNAs and targeted genes. Finally, the effect of miR-222-3p on iMECs fate reprogramming in vitro by transfecting. Results: As a result goat ear fibroblasts (GEFs) reprogramming into iMECs activates a regulatory program, involving 79 differentially expressed miRNAs. Besides, the programming process involved changes in multiple signaling pathways such as adherens junction, TGF-ß signaling pathway, GnRH secretion and the prolactin signaling pathway, etc. Furthermore, it was discovered that the expression of miR-222-3p downregulation by miR-222-3p inhibitor significantly increase the reprogramming efficiency and promoted lipid accumulation of iMECs.


Sujet(s)
Reprogrammation cellulaire , Cellules épithéliales , Fibroblastes , Capra , microARN , microARN/génétique , microARN/métabolisme , Animaux , Fibroblastes/métabolisme , Cellules épithéliales/métabolisme , Femelle , Reprogrammation cellulaire/génétique , Glandes mammaires animales/cytologie , Glandes mammaires animales/métabolisme , Transduction du signal , Cellules cultivées , Régulation négative
6.
Cell Rep Methods ; 4(7): 100819, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38986613

RÉSUMÉ

Cell reprogramming, which guides the conversion between cell states, is a promising technology for tissue repair and regeneration, with the ultimate goal of accelerating recovery from diseases or injuries. To accomplish this, regulators must be identified and manipulated to control cell fate. We propose Fatecode, a computational method that predicts cell fate regulators based only on single-cell RNA sequencing (scRNA-seq) data. Fatecode learns a latent representation of the scRNA-seq data using a deep learning-based classification-supervised autoencoder and then performs in silico perturbation experiments on the latent representation to predict genes that, when perturbed, would alter the original cell type distribution to increase or decrease the population size of a cell type of interest. We assessed Fatecode's performance using simulations from a mechanistic gene-regulatory network model and scRNA-seq data mapping blood and brain development of different organisms. Our results suggest that Fatecode can detect known cell fate regulators from single-cell transcriptomics datasets.


Sujet(s)
Analyse sur cellule unique , Analyse sur cellule unique/méthodes , Humains , Animaux , Réseaux de régulation génique , Biologie informatique/méthodes , Différenciation cellulaire/génétique , Analyse de séquence d'ARN/méthodes , Transcriptome , Apprentissage profond , Lignage cellulaire/génétique , Souris , Reprogrammation cellulaire/génétique , RNA-Seq/méthodes
7.
Theranostics ; 14(9): 3439-3469, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948053

RÉSUMÉ

Rationale: Synergic reprogramming of metabolic dominates neuroblastoma (NB) progression. It is of great clinical implications to develop an individualized risk prognostication approach with stratification-guided therapeutic options for NB based on elucidating molecular mechanisms of metabolic reprogramming. Methods: With a machine learning-based multi-step program, the synergic mechanisms of metabolic reprogramming-driven malignant progression of NB were elucidated at single-cell and metabolite flux dimensions. Subsequently, a promising metabolic reprogramming-associated prognostic signature (MPS) and individualized therapeutic approaches based on MPS-stratification were developed and further validated independently using pre-clinical models. Results: MPS-identified MPS-I NB showed significantly higher activity of metabolic reprogramming than MPS-II counterparts. MPS demonstrated improved accuracy compared to current clinical characteristics [AUC: 0.915 vs. 0.657 (MYCN), 0.713 (INSS-stage), and 0.808 (INRG-stratification)] in predicting prognosis. AZD7762 and etoposide were identified as potent therapeutics against MPS-I and II NB, respectively. Subsequent biological tests revealed AZD7762 substantially inhibited growth, migration, and invasion of MPS-I NB cells, more effectively than that of MPS-II cells. Conversely, etoposide had better therapeutic effects on MPS-II NB cells. More encouragingly, AZD7762 and etoposide significantly inhibited in-vivo subcutaneous tumorigenesis, proliferation, and pulmonary metastasis in MPS-I and MPS-II samples, respectively; thereby prolonging survival of tumor-bearing mice. Mechanistically, AZD7762 and etoposide-induced apoptosis of the MPS-I and MPS-II cells, respectively, through mitochondria-dependent pathways; and MPS-I NB resisted etoposide-induced apoptosis by addiction of glutamate metabolism and acetyl coenzyme A. MPS-I NB progression was fueled by multiple metabolic reprogramming-driven factors including multidrug resistance, immunosuppressive and tumor-promoting inflammatory microenvironments. Immunologically, MPS-I NB suppressed immune cells via MIF and THBS signaling pathways. Metabolically, the malignant proliferation of MPS-I NB cells was remarkably supported by reprogrammed glutamate metabolism, tricarboxylic acid cycle, urea cycle, etc. Furthermore, MPS-I NB cells manifested a distinct tumor-promoting developmental lineage and self-communication patterns, as evidenced by enhanced oncogenic signaling pathways activated with development and self-communications. Conclusions: This study provides deep insights into the molecular mechanisms underlying metabolic reprogramming-mediated malignant progression of NB. It also sheds light on developing targeted medications guided by the novel precise risk prognostication approaches, which could contribute to a significantly improved therapeutic strategy for NB.


Sujet(s)
Évolution de la maladie , Étoposide , Neuroblastome , Microenvironnement tumoral , Neuroblastome/traitement médicamenteux , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Humains , Animaux , Souris , Lignée cellulaire tumorale , Étoposide/pharmacologie , Étoposide/usage thérapeutique , Pronostic , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Thérapie moléculaire ciblée/méthodes , Apprentissage machine , Apoptose/effets des médicaments et des substances chimiques ,
8.
Sci Adv ; 10(28): eadn2091, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996013

RÉSUMÉ

Generation of neurons through direct reprogramming has emerged as a promising therapeutic approach for treating neurodegenerative diseases. In this study, we present an efficient method for reprogramming retinal glial cells into neurons. By suppressing Notch signaling by disrupting either Rbpj or Notch1/2, we induced mature Müller glial cells to reprogram into bipolar- and amacrine-like neurons. We demonstrate that Rbpj directly activates both Notch effector genes and genes specific to mature Müller glia while indirectly repressing expression of neurogenic basic helix-loop-helix (bHLH) factors. Combined loss of function of Rbpj and Nfia/b/x resulted in conversion of nearly all Müller glia to neurons. Last, inducing Müller glial proliferation by overexpression of dominant-active Yap promotes neurogenesis in both Rbpj- and Nfia/b/x/Rbpj-deficient Müller glia. These findings demonstrate that Notch signaling and NFI factors act in parallel to inhibit neurogenic competence in mammalian Müller glia and help clarify potential strategies for regenerative therapies aimed at treating retinal dystrophies.


Sujet(s)
Reprogrammation cellulaire , Cellules épendymogliales , Facteurs nucléaires-I , Névroglie , Neurones , Récepteurs Notch , Rétine , Transduction du signal , Animaux , Facteurs nucléaires-I/métabolisme , Facteurs nucléaires-I/génétique , Souris , Rétine/métabolisme , Rétine/cytologie , Cellules épendymogliales/métabolisme , Cellules épendymogliales/cytologie , Névroglie/métabolisme , Récepteurs Notch/métabolisme , Neurones/métabolisme , Neurones/cytologie , Facteur de transcription CBF-1/métabolisme , Facteur de transcription CBF-1/génétique , Neurogenèse , Protéines de signalisation YAP/métabolisme , Prolifération cellulaire
9.
Cells ; 13(12)2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38920635

RÉSUMÉ

Prostate cancer (PCa) remains a leading cause of mortality among American men, with metastatic and recurrent disease posing significant therapeutic challenges due to a limited comprehension of the underlying biological processes governing disease initiation, dormancy, and progression. The conventional use of PCa cell lines has proven inadequate in elucidating the intricate molecular mechanisms driving PCa carcinogenesis, hindering the development of effective treatments. To address this gap, patient-derived primary cell cultures have been developed and play a pivotal role in unraveling the pathophysiological intricacies unique to PCa in each individual, offering valuable insights for translational research. This review explores the applications of the conditional reprogramming (CR) cell culture approach, showcasing its capability to rapidly and effectively cultivate patient-derived normal and tumor cells. The CR strategy facilitates the acquisition of stem cell properties by primary cells, precisely recapitulating the human pathophysiology of PCa. This nuanced understanding enables the identification of novel therapeutics. Specifically, our discussion encompasses the utility of CR cells in elucidating PCa initiation and progression, unraveling the molecular pathogenesis of metastatic PCa, addressing health disparities, and advancing personalized medicine. Coupled with the tumor organoid approach and patient-derived xenografts (PDXs), CR cells present a promising avenue for comprehending cancer biology, exploring new treatment modalities, and advancing precision medicine in the context of PCa. These approaches have been used for two NCI initiatives (PDMR: patient-derived model repositories; HCMI: human cancer models initiatives).


Sujet(s)
Reprogrammation cellulaire , Tumeurs de la prostate , Humains , Tumeurs de la prostate/anatomopathologie , Mâle , Reprogrammation cellulaire/génétique , Animaux
10.
Cells ; 13(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38891074

RÉSUMÉ

Glioblastoma (GBM) is the most common yet uniformly fatal adult brain cancer. Intra-tumoral molecular and cellular heterogeneities are major contributory factors to therapeutic refractoriness and futility in GBM. Molecular heterogeneity is represented through molecular subtype clusters whereby the proneural (PN) subtype is associated with significantly increased long-term survival compared to the highly resistant mesenchymal (MES) subtype. Furthermore, it is universally recognized that a small subset of GBM cells known as GBM stem cells (GSCs) serve as reservoirs for tumor recurrence and progression. The clonal evolution of GSC molecular subtypes in response to therapy drives intra-tumoral heterogeneity and remains a critical determinant of GBM outcomes. In particular, the intra-tumoral MES reprogramming of GSCs using current GBM therapies has emerged as a leading hypothesis for therapeutic refractoriness. Preventing the intra-tumoral divergent evolution of GBM toward the MES subtype via new treatments would dramatically improve long-term survival for GBM patients and have a significant impact on GBM outcomes. In this review, we examine the challenges of the role of MES reprogramming in the malignant clonal evolution of glioblastoma and provide future perspectives for addressing the unmet therapeutic need to overcome resistance in GBM.


Sujet(s)
Tumeurs du cerveau , Reprogrammation cellulaire , Évolution clonale , Glioblastome , Humains , Glioblastome/anatomopathologie , Glioblastome/génétique , Évolution clonale/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Reprogrammation cellulaire/génétique , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/métabolisme , Animaux , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie
11.
Mol Med ; 30(1): 96, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38914953

RÉSUMÉ

Lupus nephritis (LN) is a severe and common manifestation of systemic lupus erythematosus (SLE) that is frequently identified with a poor prognosis. Macrophages play an important role in its pathogenesis. Different macrophage subtypes have different effects on lupus-affected kidneys. Based on their origin, macrophages can be divided into monocyte-derived macrophages (MoMacs) and tissue-resident macrophages (TrMacs). During nephritis, TrMacs develop a hybrid pro-inflammatory and anti-inflammatory functional phenotype, as they do not secrete arginase or nitric oxide (NO) when stimulated by cytokines. The infiltration of these mixed-phenotype macrophages is related to the continuous damage caused by immune complexes and exposure to circulating inflammatory mediators, which is an indication of the failure to resolve inflammation. On the other hand, MoMacs differentiate into M1 or M2 cells under cytokine stimulation. M1 macrophages are pro-inflammatory and secrete pro-inflammatory cytokines, while the M2 main phenotype is essentially anti-inflammatory and promotes tissue repair. Conversely, MoMacs undergo differentiation into M1 or M2 cells in response to cytokine stimulation. M1 macrophages are considered pro-inflammatory cells and secrete pro-inflammatory mediators, whereas the M2 main phenotype is primarily anti-inflammatory and promotes tissue repair. Moreover, based on cytokine expression, M2 macrophages can be further divided into M2a, M2b, and M2c phenotypes. M2a and M2c have anti-inflammatory effects and participate in tissue repair, while M2b cells have immunoregulatory and pro-inflammatory properties. Further, memory macrophages also have a role in the advancement of LN. Studies have demonstrated that the polarization of macrophages is controlled by multiple metabolic pathways, such as glycolysis, the pentose phosphate pathway, fatty acid oxidation, sphingolipid metabolism, the tricarboxylic acid cycle, and arginine metabolism. The changes in these metabolic pathways can be regulated by substances such as fish oil, polyenylphosphatidylcholine, taurine, fumaric acid, metformin, and salbutamol, which inhibit M1 polarization of macrophages and promote M2 polarization, thereby alleviating LN.


Sujet(s)
Glomérulonéphrite lupique , Macrophages , Humains , Glomérulonéphrite lupique/métabolisme , Glomérulonéphrite lupique/thérapie , Glomérulonéphrite lupique/immunologie , Macrophages/métabolisme , Macrophages/immunologie , Animaux , Activation des macrophages , Cytokines/métabolisme , Différenciation cellulaire , Prise en charge de la maladie , Reprogrammation cellulaire ,
12.
Sheng Li Xue Bao ; 76(3): 457-474, 2024 Jun 25.
Article de Chinois | MEDLINE | ID: mdl-38939940

RÉSUMÉ

Abdominal aortic aneurysm (AAA) is a life-threatening disease that remains undetected until it acutely ruptures. Due to lack of effective pharmaceutic therapies, it is urgent to explore new prevention and treatment strategies. Metabolic reprogramming is a cellular process through which cells change their metabolic patterns to meet material and energy requirements, including glucose metabolism, lipid metabolism and amino acid metabolism. Recently, the regulatory role of metabolic reprogramming in cardiovascular diseases, especially AAA, has attracted significant attention. This review article focuses on the research progress regarding the effects of metabolic reprogramming of vascular smooth muscle cells (VSMCs) and macrophages on the occurrence and development of AAA, especially their roles in major pathological processes such as VSMCs apoptosis and phenotype transformation, extracellular matrix remodeling, oxidative stress, and inflammatory response. The aim is to provide new clues for the mechanism research and clinical treatment of AAA from the perspective of metabolism.


Sujet(s)
Anévrysme de l'aorte abdominale , Muscles lisses vasculaires , Anévrysme de l'aorte abdominale/métabolisme , Humains , Muscles lisses vasculaires/métabolisme , Animaux , Myocytes du muscle lisse/métabolisme , Macrophages/métabolisme , Stress oxydatif , Apoptose , Métabolisme lipidique , Reprogrammation cellulaire ,
13.
Sci Rep ; 14(1): 13146, 2024 06 07.
Article de Anglais | MEDLINE | ID: mdl-38849434

RÉSUMÉ

Multiple sclerosis (MS) is an autoimmune demyelinating disease affecting the central nervous system (CNS). T helper (Th) 17 cells are involved in the pathogenesis of MS and its animal model of experimental autoimmune encephalomyelitis (EAE) by infiltrating the CNS and producing effector molecules that engage resident glial cells. Among these glial cells, astrocytes have a central role in coordinating inflammatory processes by responding to cytokines and chemokines released by Th17 cells. In this study, we examined the impact of pathogenic Th17 cells on astrocytes in vitro and in vivo. We identified that Th17 cells reprogram astrocytes by driving transcriptomic changes partly through a Janus Kinase (JAK)1-dependent mechanism, which included increased chemokines, interferon-inducible genes, and cytokine receptors. In vivo, we observed a region-specific heterogeneity in the expression of cell surface cytokine receptors on astrocytes, including those for IFN-γ, IL-1, TNF-α, IL-17, TGFß, and IL-10. Additionally, these receptors were dynamically regulated during EAE induced by adoptive transfer of myelin-reactive Th17 cells. This study overall provides evidence of Th17 cell reprogramming of astrocytes, which may drive changes in the astrocytic responsiveness to cytokines during autoimmune neuroinflammation.


Sujet(s)
Astrocytes , Encéphalomyélite auto-immune expérimentale , Janus kinase 1 , Glycoprotéine MOG , Récepteurs aux cytokines , Cellules Th17 , Encéphalomyélite auto-immune expérimentale/métabolisme , Encéphalomyélite auto-immune expérimentale/immunologie , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Animaux , Astrocytes/métabolisme , Cellules Th17/immunologie , Cellules Th17/métabolisme , Souris , Récepteurs aux cytokines/métabolisme , Récepteurs aux cytokines/génétique , Janus kinase 1/métabolisme , Souris de lignée C57BL , Cytokines/métabolisme , Reprogrammation cellulaire , Femelle , Cellules cultivées
14.
Front Immunol ; 15: 1395786, 2024.
Article de Anglais | MEDLINE | ID: mdl-38835758

RÉSUMÉ

It is commonly known that different macrophage phenotypes play specific roles in different pathophysiological processes. In recent years, many studies have linked the phenotypes of macrophages to their characteristics in different metabolic pathways, suggesting that macrophages can perform different functions through metabolic reprogramming. It is now gradually recognized that lactate, previously overlooked as a byproduct of glycolytic metabolism, acts as a signaling molecule in regulating multiple biological processes, including immunological responses and metabolism. Recently, lactate has been found to mediate epigenetic changes in macrophages through a newfound lactylation modification, thereby regulating their phenotypic transformation. This novel finding highlights the significant role of lactate metabolism in macrophage function. In this review, we summarize the features of relevant metabolic reprogramming in macrophages and the role of lactate metabolism therein. We also review the progress of research on the regulation of macrophage metabolic reprogramming by lactylation through epigenetic mechanisms.


Sujet(s)
Reprogrammation cellulaire , Épigenèse génétique , Acide lactique , Macrophages , Macrophages/métabolisme , Macrophages/immunologie , Humains , Animaux , Acide lactique/métabolisme ,
15.
BMC Cancer ; 24(1): 698, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849760

RÉSUMÉ

BACKGROUND: Tumor-associated macrophages (TAMs) constitute a substantial part of human hepatocellular carcinoma (HCC). The present study was devised to explore TAM diversity and their roles in HCC progression. METHODS: Through the integration of multiple 10 × single-cell transcriptomic data derived from HCC samples and the use of consensus nonnegative matrix factorization (an unsupervised clustering algorithm), TAM molecular subtypes and expression programs were evaluated in detail. The roles played by these TAM subtypes in HCC were further probed through pseudotime, enrichment, and intercellular communication analyses. Lastly, vitro experiments were performed to validate the relationship between CD63, which is an inflammatory TAM expression program marker, and tumor cell lines. RESULTS: We found that the inflammatory expression program in TAMs had a more obvious interaction with HCC cells, and CD63, as a marker gene of the inflammatory expression program, was associated with poor prognosis of HCC patients. Both bulk RNA-seq and vitro experiments confirmed that higher TAM CD63 expression was associated with the growth of HCC cells as well as their epithelial-mesenchymal transition, metastasis, invasion, and the reprogramming of lipid metabolism. CONCLUSIONS: These analyses revealed that the TAM inflammatory expression program in HCC is closely associated with malignant tumor cells, with the hub gene CD63 thus representing an ideal target for therapeutic intervention in this cancer type.


Sujet(s)
Carcinome hépatocellulaire , Évolution de la maladie , Transition épithélio-mésenchymateuse , Tumeurs du foie , Antigène CD63 , Macrophages associés aux tumeurs , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Humains , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Transition épithélio-mésenchymateuse/génétique , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/anatomopathologie , Antigène CD63/métabolisme , Antigène CD63/génétique , Métabolisme lipidique/génétique , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Pronostic , Reprogrammation cellulaire/génétique
16.
Proc Natl Acad Sci U S A ; 121(26): e2320835121, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38900797

RÉSUMÉ

Upper aerodigestive squamous cell carcinoma (UASCC) is a common and aggressive malignancy with few effective therapeutic options. Here, we investigate amino acid metabolism in this cancer, surprisingly noting that UASCC exhibits the highest methionine level across all human cancers, driven by its transporter LAT1. We show that LAT1 is also expressed at the highest level in UASCC, transcriptionally activated by UASCC-specific promoter and enhancers, which are directly coregulated by SCC master regulators TP63/KLF5/SREBF1. Unexpectedly, unbiased bioinformatic screen identifies EZH2 as the most significant target downstream of the LAT1-methionine pathway, directly linking methionine metabolism to epigenomic reprogramming. Importantly, this cascade is indispensable for the survival and proliferation of UASCC patient-derived tumor organoids. In addition, LAT1 expression is closely associated with cellular sensitivity to inhibition of the LAT1-methionine-EZH2 axis. Notably, this unique LAT1-methionine-EZH2 cascade can be targeted effectively by either pharmacological approaches or dietary intervention in vivo. In summary, this work maps a unique mechanistic cross talk between epigenomic reprogramming with methionine metabolism, establishes its biological significance in the biology of UASCC, and identifies a unique tumor-specific vulnerability which can be exploited both pharmacologically and dietarily.


Sujet(s)
Carcinome épidermoïde , Régulation de l'expression des gènes tumoraux , Transporteur-1 d'acides aminés neutres à longue chaîne , Méthionine , Méthionine/métabolisme , Humains , Transporteur-1 d'acides aminés neutres à longue chaîne/métabolisme , Transporteur-1 d'acides aminés neutres à longue chaîne/génétique , Carcinome épidermoïde/génétique , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Protéine-2 homologue de l'activateur de Zeste/génétique , Lignée cellulaire tumorale , Épigenèse génétique , Épigénomique/méthodes , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/métabolisme , Tumeurs de la tête et du cou/anatomopathologie , Souris , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Animaux , Prolifération cellulaire , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Reprogrammation cellulaire/génétique
17.
Cells ; 13(11)2024 May 26.
Article de Anglais | MEDLINE | ID: mdl-38891050

RÉSUMÉ

Acute inflammation is a rapid and dynamic process involving the recruitment and activation of multiple cell types in a coordinated and precise manner. Here, we investigate the origin and transcriptional reprogramming of monocytes using a model of acute inflammation, zymosan-induced peritonitis. Monocyte trafficking and adoptive transfer experiments confirmed that monocytes undergo rapid phenotypic change as they exit the blood and give rise to monocyte-derived macrophages that persist during the resolution of inflammation. Single-cell transcriptomics revealed significant heterogeneity within the surface marker-defined CD11b+Ly6G-Ly6Chi monocyte populations within the blood and at the site of inflammation. We show that two major transcriptional reprogramming events occur during the initial six hours of Ly6Chi monocyte mobilisation, one in the blood priming monocytes for migration and a second at the site of inflammation. Pathway analysis revealed an important role for oxidative phosphorylation (OxPhos) during both these reprogramming events. Experimentally, we demonstrate that OxPhos via the intact mitochondrial electron transport chain is essential for murine and human monocyte chemotaxis. Moreover, OxPhos is needed for monocyte-to-macrophage differentiation and macrophage M(IL-4) polarisation. These new findings from transcriptional profiling open up the possibility that shifting monocyte metabolic capacity towards OxPhos could facilitate enhanced macrophage M2-like polarisation to aid inflammation resolution and tissue repair.


Sujet(s)
Antigènes Ly , Différenciation cellulaire , Inflammation , Macrophages , Monocytes , Phosphorylation oxydative , Monocytes/métabolisme , Animaux , Macrophages/métabolisme , Inflammation/anatomopathologie , Inflammation/métabolisme , Humains , Souris , Antigènes Ly/métabolisme , Chimiotaxie , Souris de lignée C57BL , Péritonite/métabolisme , Péritonite/induit chimiquement , Péritonite/anatomopathologie , Zymosan/pharmacologie , Mitochondries/métabolisme , Reprogrammation cellulaire
18.
Signal Transduct Target Ther ; 9(1): 151, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38910148

RÉSUMÉ

Pancreatic cancer is one of the deadly malignancies with a significant mortality rate and there are currently few therapeutic options for it. The tumor microenvironment (TME) in pancreatic cancer, distinguished by fibrosis and the existence of cancer-associated fibroblasts (CAFs), exerts a pivotal influence on both tumor advancement and resistance to therapy. Recent advancements in the field of engineered extracellular vesicles (EVs) offer novel avenues for targeted therapy in pancreatic cancer. This study aimed to develop engineered EVs for the targeted reprogramming of CAFs and modulating the TME in pancreatic cancer. EVs obtained from bone marrow mesenchymal stem cells (BMSCs) were loaded with miR-138-5p and the anti-fibrotic agent pirfenidone (PFD) and subjected to surface modification with integrin α5-targeting peptides (named IEVs-PFD/138) to reprogram CAFs and suppress their pro-tumorigenic effects. Integrin α5-targeting peptide modification enhanced the CAF-targeting ability of EVs. miR-138-5p directly inhibited the formation of the FERMT2-TGFBR1 complex, inhibiting TGF-ß signaling pathway activation. In addition, miR-138-5p inhibited proline-mediated collagen synthesis by directly targeting the FERMT2-PYCR1 complex. The combination of miR-138-5p and PFD in EVs synergistically promoted CAF reprogramming and suppressed the pro-cancer effects of CAFs. Preclinical experiments using the orthotopic stroma-rich and patient-derived xenograft mouse models yielded promising results. In particular, IEVs-PFD/138 effectively reprogrammed CAFs and remodeled TME, which resulted in decreased tumor pressure, enhanced gemcitabine perfusion, tumor hypoxia amelioration, and greater sensitivity of cancer cells to chemotherapy. Thus, the strategy developed in this study can improve chemotherapy outcomes. Utilizing IEVs-PFD/138 as a targeted therapeutic agent to modulate CAFs and the TME represents a promising therapeutic approach for pancreatic cancer.


Sujet(s)
Fibroblastes associés au cancer , Vésicules extracellulaires , microARN , Tumeurs du pancréas , Microenvironnement tumoral , Tumeurs du pancréas/génétique , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/métabolisme , Vésicules extracellulaires/génétique , Vésicules extracellulaires/métabolisme , Humains , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/anatomopathologie , Souris , microARN/génétique , Animaux , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/génétique , Reprogrammation cellulaire/génétique , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Cellules souches mésenchymateuses/métabolisme , Protéines tumorales/génétique , Protéines tumorales/métabolisme ,
20.
Nat Commun ; 15(1): 4946, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862540

RÉSUMÉ

Genomic aberrations are a critical impediment for the safe medical use of iPSCs and their origin and developmental mechanisms remain unknown. Here we find through WGS analysis of human and mouse iPSC lines that genomic mutations are de novo events and that, in addition to unmodified cytosine base prone to deamination, the DNA methylation sequence CpG represents a significant mutation-prone site. CGI and TSS regions show increased mutations in iPSCs and elevated mutations are observed in retrotransposons, especially in the AluY subfamily. Furthermore, increased cytosine to thymine mutations are observed in differentially methylated regions. These results indicate that in addition to deamination of cytosine, demethylation of methylated cytosine, which plays a central role in genome reprogramming, may act mutagenically during iPSC generation.


Sujet(s)
Ilots CpG , Cytosine , Méthylation de l'ADN , Cellules souches pluripotentes induites , Mutation ponctuelle , Cellules souches pluripotentes induites/métabolisme , Cytosine/métabolisme , Animaux , Humains , Souris , Reprogrammation cellulaire/génétique , Rétroéléments/génétique , Lignée cellulaire
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE