Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 571
Filtrer
1.
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 ,
2.
Nature ; 631(8022): 876-883, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38987605

RÉSUMÉ

Advancements in precision oncology over the past decades have led to new therapeutic interventions, but the efficacy of such treatments is generally limited by an adaptive process that fosters drug resistance1. In addition to genetic mutations2, recent research has identified a role for non-genetic plasticity in transient drug tolerance3 and the acquisition of stable resistance4,5. However, the dynamics of cell-state transitions that occur in the adaptation to cancer therapies remain unknown and require a systems-level longitudinal framework. Here we demonstrate that resistance develops through trajectories of cell-state transitions accompanied by a progressive increase in cell fitness, which we denote as the 'resistance continuum'. This cellular adaptation involves a stepwise assembly of gene expression programmes and epigenetically reinforced cell states underpinned by phenotypic plasticity, adaptation to stress and metabolic reprogramming. Our results support the notion that epithelial-to-mesenchymal transition or stemness programmes-often considered a proxy for phenotypic plasticity-enable adaptation, rather than a full resistance mechanism. Through systematic genetic perturbations, we identify the acquisition of metabolic dependencies, exposing vulnerabilities that can potentially be exploited therapeutically. The concept of the resistance continuum highlights the dynamic nature of cellular adaptation and calls for complementary therapies directed at the mechanisms underlying adaptive cell-state transitions.


Sujet(s)
Adaptation physiologique , Plasticité cellulaire , Résistance aux médicaments antinéoplasiques , Tumeurs , Femelle , Humains , Souris , Adaptation physiologique/effets des médicaments et des substances chimiques , Adaptation physiologique/génétique , Lignée cellulaire tumorale , Plasticité cellulaire/effets des médicaments et des substances chimiques , Plasticité cellulaire/génétique , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/génétique , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Épigenèse génétique , Transition épithélio-mésenchymateuse/génétique , Régulation de l'expression des gènes tumoraux/génétique , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/anatomopathologie , Phénotype
3.
Biomaterials ; 311: 122708, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39047538

RÉSUMÉ

The resistance of glioblastoma multiforme (GBM) to standard chemotherapy is primarily attributed to the existence of tumor-associated macrophages (TAMs) in the GBM microenvironment, particularly the anti-inflammatory M2 phenotype. Targeted modulation of M2-TAMs is emerging as a promising strategy to enhance chemotherapeutic efficacy. However, combination TAM-targeted therapy with chemotherapy faces substantial challenges, notably in terms of delivery efficiency and targeting specificity. In this study, we designed a pH-responsive hierarchical brain-targeting micelleplex loaded with temozolomide (TMZ) and resiquimod (R848) for combination chemo-immunotherapy against GBM. This delivery system, termed PCPA&PPM@TR, features a primary Angiopep-2 decoration on the outer layer via a pH-cleavable linker and a secondary mannose analogue (MAN) on the middle layer. This pH-responsive hierarchical targeting strategy enables effective BBB permeability while simultaneous GBM- and TAMs-targeting delivery. GBM-targeted delivery of TMZ induces alkylation and triggers an anti-GBM immune response. Concurrently, TAM-targeted delivery of R848 reprograms their phenotype from M2 to pro-inflammatory M1, thereby diminishing GBM resistance to TMZ and amplifying the immune response. In vivo studies demonstrated that targeted modulation of TAMs using PCPA&PPM@TR significantly enhanced anti-GBM efficacy. In summary, this study proposes a promising brain-targeting delivery system for the targeted modulation of TAMs to combat GBM.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Glioblastome , Immunothérapie , Témozolomide , Macrophages associés aux tumeurs , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Glioblastome/thérapie , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/métabolisme , Témozolomide/pharmacologie , Témozolomide/usage thérapeutique , Animaux , Humains , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Immunothérapie/méthodes , Lignée cellulaire tumorale , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/thérapie , Souris , Imidazoles/pharmacologie , Imidazoles/composition chimique , Systèmes de délivrance de médicaments , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Micelles , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Concentration en ions d'hydrogène
4.
Nano Lett ; 24(29): 9104-9114, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39007505

RÉSUMÉ

Tumor-associated macrophages (TAMs), as the most prevalent immune cells in the tumor microenvironment, play a pivotal role in promoting tumor development through various signaling pathways. Herein, we have engineered a Se@ZIF-8 core-satellite nanoassembly to reprogram TAMs, thereby enhancing immunotherapy outcomes. When the nanoassembly reaches the tumor tissue, selenium nanoparticles and Zn2+ are released in response to the acidic tumor microenvironment, resulting in a collaborative effort to promote the production of reactive oxygen species (ROS). The generated ROS, in turn, activate the nuclear factor κB (NF-κB) signaling pathway, driving the repolarization of TAMs from M2-type to M1-type, effectively eliminating cancer cells. Moreover, the nanoassembly can induce the immunogenic death of cancer cells through excess ROS to expose calreticulin and boost macrophage phagocytosis. The Se@ZIF-8 core-satellite nanoassembly provides a potential paradigm for cancer immunotherapy by reversing the immunosuppressive microenvironment.


Sujet(s)
Immunothérapie , Espèces réactives de l'oxygène , Sélénium , Microenvironnement tumoral , Macrophages associés aux tumeurs , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Espèces réactives de l'oxygène/métabolisme , Souris , Animaux , Humains , Sélénium/composition chimique , Sélénium/pharmacologie , Tumeurs/thérapie , Tumeurs/immunologie , Facteur de transcription NF-kappa B/métabolisme , Nanoparticules/composition chimique , Nanoparticules/usage thérapeutique , Lignée cellulaire tumorale , Transduction du signal/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Phagocytose/effets des médicaments et des substances chimiques
5.
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 ,
6.
Cells ; 13(11)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38891029

RÉSUMÉ

Glioblastoma Multiforme (GBM) is an aggressive brain tumor with a high mortality rate. Direct reprogramming of glial cells to different cell lineages, such as induced neural stem cells (iNSCs) and induced neurons (iNeurons), provides genetic tools to manipulate a cell's fate as a potential therapy for neurological diseases. NeuroD1 (ND1) is a master transcriptional factor for neurogenesis and it promotes neuronal differentiation. In the present study, we tested the hypothesis that the expression of ND1 in GBM cells can force them to differentiate toward post-mitotic neurons and halt GBM tumor progression. In cultured human GBM cell lines, including LN229, U87, and U373 as temozolomide (TMZ)-sensitive and T98G as TMZ-resistant cells, the neuronal lineage conversion was induced by an adeno-associated virus (AAV) package carrying ND1. Twenty-one days after AAV-ND1 transduction, ND1-expressing cells displayed neuronal markers MAP2, TUJ1, and NeuN. The ND1-induced transdifferentiation was regulated by Wnt signaling and markedly enhanced under a hypoxic condition (2% O2 vs. 21% O2). ND1-expressing GBM cultures had fewer BrdU-positive proliferating cells compared to vector control cultures. Increased cell death was visualized by TUNEL staining, and reduced migrative activity was demonstrated in the wound-healing test after ND1 reprogramming in both TMZ-sensitive and -resistant GBM cells. In a striking contrast to cancer cells, converted cells expressed the anti-tumor gene p53. In an orthotopical GBM mouse model, AAV-ND1-reprogrammed U373 cells were transplanted into the fornix of the cyclosporine-immunocompromised C57BL/6 mouse brain. Compared to control GBM cell-formed tumors, cells from ND1-reprogrammed cultures formed smaller tumors and expressed neuronal markers such as TUJ1 in the brain. Thus, reprogramming using a single-factor ND1 overcame drug resistance, converting malignant cells of heterogeneous GBM cells to normal neuron-like cells in vitro and in vivo. These novel observations warrant further research using patient-derived GBM cells and patient-derived xenograft (PDX) models as a potentially effective treatment for a deadly brain cancer and likely other astrocytoma tumors.


Sujet(s)
Reprogrammation cellulaire , Glioblastome , Neurones , Glioblastome/anatomopathologie , Glioblastome/génétique , Glioblastome/métabolisme , Humains , Animaux , Lignée cellulaire tumorale , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Souris , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Témozolomide/pharmacologie , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique
7.
Stem Cells ; 42(8): 706-719, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38825983

RÉSUMÉ

The transformation from a fibroblast mesenchymal cell state to an epithelial-like state is critical for induced pluripotent stem cell (iPSC) reprogramming. In this report, we describe studies with PFI-3, a small-molecule inhibitor that specifically targets the bromodomains of SMARCA2/4 and PBRM1 subunits of SWI/SNF complex, as an enhancer of iPSC reprogramming efficiency. Our findings reveal that PFI-3 induces cellular plasticity in multiple human dermal fibroblasts, leading to a mesenchymal-epithelial transition during iPSC formation. This transition is characterized by the upregulation of E-cadherin expression, a key protein involved in epithelial cell adhesion. Additionally, we identified COL11A1 as a reprogramming barrier and demonstrated COL11A1 knockdown increased reprogramming efficiency. Notably, we found that PFI-3 significantly reduced the expression of numerous extracellular matrix (ECM) genes, particularly those involved in collagen assembly. Our research provides key insights into the early stages of iPSC reprogramming, highlighting the crucial role of ECM changes and cellular plasticity in this process.


Sujet(s)
Plasticité cellulaire , Reprogrammation cellulaire , Matrice extracellulaire , Cellules souches pluripotentes induites , Facteurs de transcription , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Reprogrammation cellulaire/génétique , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Matrice extracellulaire/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Plasticité cellulaire/génétique , Plasticité cellulaire/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/cytologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/génétique , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques
8.
Comput Biol Med ; 177: 108661, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38810477

RÉSUMÉ

With the use of specific genetic factors and recent developments in cellular reprogramming, it is now possible to generate lineage-committed cells or induced pluripotent stem cells (iPSCs) from readily available and common somatic cell types. However, there are still significant doubts regarding the safety and effectiveness of the current genetic methods for reprogramming cells, as well as the conventional culture methods for maintaining stem cells. Small molecules that target specific epigenetic processes, signaling pathways, and other cellular processes can be used as a complementary approach to manipulate cell fate to achieve a desired objective. It has been discovered that a growing number of small molecules can support lineage differentiation, maintain stem cell self-renewal potential, and facilitate reprogramming by either increasing the efficiency of reprogramming or acting as a genetic reprogramming factor substitute. However, ongoing challenges include improving reprogramming efficiency, ensuring the safety of small molecules, and addressing issues with incomplete epigenetic resetting. Small molecule iPSCs have significant clinical applications in regenerative medicine and personalized therapies. This review emphasizes the versatility and potential safety benefits of small molecules in overcoming challenges associated with the iPSCs reprogramming process.


Sujet(s)
Reprogrammation cellulaire , Cellules souches pluripotentes induites , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Cellules souches pluripotentes induites/métabolisme , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Animaux , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/composition chimique , Épigenèse génétique , Différenciation cellulaire/effets des médicaments et des substances chimiques
9.
Cells ; 13(9)2024 Apr 29.
Article de Anglais | MEDLINE | ID: mdl-38727298

RÉSUMÉ

The antipsychotic drug clozapine demonstrates superior efficacy in treatment-resistant schizophrenia, but its intracellular mode of action is not completely understood. Here, we analysed the effects of clozapine (2.5-20 µM) on metabolic fluxes, cell respiration, and intracellular ATP in human HL60 cells. Some results were confirmed in leukocytes of clozapine-treated patients. Neuroreceptor inhibition under clozapine reduced Akt activation with decreased glucose uptake, thereby inducing ER stress and the unfolded protein response (UPR). Metabolic profiling by liquid-chromatography/mass-spectrometry revealed downregulation of glycolysis and the pentose phosphate pathway, thereby saving glucose to keep the electron transport chain working. Mitochondrial respiration was dampened by upregulation of the F0F1-ATPase inhibitory factor 1 (IF1) leading to 30-40% lower oxygen consumption in HL60 cells. Blocking IF1 expression by cotreatment with epigallocatechin-3-gallate (EGCG) increased apoptosis of HL60 cells. Upregulation of the mitochondrial citrate carrier shifted excess citrate to the cytosol for use in lipogenesis and for storage as triacylglycerol in lipid droplets (LDs). Accordingly, clozapine-treated HL60 cells and leukocytes from clozapine-treated patients contain more LDs than untreated cells. Since mitochondrial disturbances are described in the pathophysiology of schizophrenia, clozapine-induced mitohormesis is an excellent way to escape energy deficits and improve cell survival.


Sujet(s)
Clozapine , Mitochondries , Humains , Clozapine/pharmacologie , Clozapine/analogues et dérivés , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Cellules HL-60 , Neuroleptiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Adénosine triphosphate/métabolisme , Schizophrénie/traitement médicamenteux , Schizophrénie/métabolisme , Schizophrénie/anatomopathologie , Leucocytes/effets des médicaments et des substances chimiques , Leucocytes/métabolisme , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/effets des médicaments et des substances chimiques ,
10.
Theranostics ; 14(7): 2856-2880, 2024.
Article de Anglais | MEDLINE | ID: mdl-38773968

RÉSUMÉ

Cell metabolism reprogramming to sustain energy production, while reducing oxygen and energy consuming processes is crucially important for the adaptation to hypoxia/ischemia. Adaptive metabolic rewiring is controlled by hypoxia-inducible factors (HIFs). Accumulating experimental evidence indicates that timely activation of HIF in brain-resident cells improves the outcome from acute ischemic stroke. However, the underlying molecular mechanisms are still incompletely understood. Thus, we investigated whether HIF-dependent metabolic reprogramming affects the vulnerability of brain-resident cells towards ischemic stress. Methods: We used genetic and pharmacological approaches to activate HIF in the murine brain in vivo and in primary neurons and astrocytes in vitro. Numerous metabolomic approaches and molecular biological techniques were applied to elucidate potential HIF-dependent effects on the central carbon metabolism of brain cells. In animal and cell models of ischemic stroke, we analysed whether HIF-dependent metabolic reprogramming influences the susceptibility to ischemic injury. Results: Neuron-specific gene ablation of prolyl-4-hydroxylase domain 2 (PHD2) protein, negatively regulating the protein stability of HIF-α in an oxygen dependent manner, reduced brain injury and functional impairment of mice after acute stroke in a HIF-dependent manner. Accordingly, PHD2 deficient neurons showed an improved tolerance towards ischemic stress in vitro, which was accompanied by enhanced HIF-1-mediated glycolytic lactate production through pyruvate dehydrogenase kinase-mediated inhibition of the pyruvate dehydrogenase. Systemic treatment of mice with roxadustat, a low-molecular weight pan-PHD inhibitor, not only increased the abundance of numerous metabolites of the central carbon and amino acid metabolism in murine brain, but also ameliorated cerebral tissue damage and sensorimotor dysfunction after acute ischemic stroke. In neurons and astrocytes roxadustat provoked a HIF-1-dependent glucose metabolism reprogramming including elevation of glucose uptake, glycogen synthesis, glycolytic capacity, lactate production and lactate release, which enhanced the ischemic tolerance of astrocytes, but not neurons. We found that strong activation of HIF-1 in neurons by non-selective inhibition of all PHD isoenzymes caused a HIF-1-dependent upregulation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 redirecting glucose-6-phosphate from pentose phosphate pathway (PPP) to the glycolysis pathway. This was accompanied by a reduction of NADPH production in the PPP, which further decreased the low intrinsic antioxidant reserve of neurons, making them more susceptible to ischemic stress. Nonetheless, in organotypic hippocampal cultures with preserved neuronal-glial interactions roxadustat decreased the neuronal susceptibility to ischemic stress, which was largely prevented by restricting glycolytic energy production through lactate transport blockade. Conclusion: Collectively, our results indicate that HIF-1-mediated metabolic reprogramming alleviates the intrinsic vulnerability of brain-resident cells to ischemic stress.


Sujet(s)
Astrocytes , Carbone , Sous-unité alpha du facteur-1 induit par l'hypoxie , Hypoxia-inducible factor-proline dioxygenases , Accident vasculaire cérébral ischémique , Neurones , Animaux , Femelle , Mâle , Souris , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Encéphalopathie ischémique/métabolisme , Carbone/métabolisme , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Glycolyse/effets des médicaments et des substances chimiques , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Hypoxia-inducible factor-proline dioxygenases/métabolisme , Accident vasculaire cérébral ischémique/métabolisme , Souris de lignée C57BL , Neurones/métabolisme , Procollagen-Proline Dioxygenase/métabolisme , Procollagen-Proline Dioxygenase/génétique
11.
Adv Mater ; 36(28): e2311845, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38720198

RÉSUMÉ

Sweat gland (SwG) regeneration is crucial for the functional rehabilitation of burn patients. In vivo chemical reprogramming that harnessing the patient's own cells in damaged tissue is of substantial interest to regenerate organs endogenously by pharmacological manipulation, which could compensate for tissue loss in devastating diseases and injuries, for example, burns. However, achieving in vivo chemical reprogramming is challenging due to the low reprogramming efficiency and an unfavorable tissue environment. Herein, this work has developed a functionalized proteinaceous nanoformulation delivery system containing prefabricated epidermal growth factor structure for on-demand delivery of a cocktail of seven SwG reprogramming components to the dermal site. Such a chemical reprogramming system can efficiently induce the conversion of epidermal keratinocytes into SwG myoepithelial cells, resulting in successful in situ regeneration of functional SwGs. Notably, in vivo chemical reprogramming of SwGs is achieved for the first time with an impressive efficiency of 30.6%, surpassing previously reported efficiencies. Overall, this proteinaceous nanoformulation provides a platform for coordinating the target delivery of multiple pharmacological agents and facilitating in vivo SwG reprogramming by chemicals. This advancement greatly improves the clinical accessibility of in vivo reprogramming and offers a non-surgical, non-viral, and cell-free strategy for in situ SwG regeneration.


Sujet(s)
Reprogrammation cellulaire , Animaux , Humains , Souris , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Facteur de croissance épidermique/composition chimique , Facteur de croissance épidermique/métabolisme , Facteur de croissance épidermique/pharmacologie , Régénération/effets des médicaments et des substances chimiques , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/métabolisme , Kératinocytes/cytologie , Nanoparticules/composition chimique
12.
Cell Rep Med ; 5(6): 101576, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38776909

RÉSUMÉ

Chemotherapy remains the first-line treatment for advanced esophageal cancer. However, durable benefits are achieved by only a limited subset of individuals due to the elusive chemoresistance. Here, we utilize patient-derived xenografts (PDXs) from esophageal squamous-cell carcinoma to investigate chemoresistance mechanisms in preclinical settings. We observe that activated cancer-associated fibroblasts (CAFs) are enriched in the tumor microenvironment of PDXs resistant to chemotherapy. Mechanistically, we reveal that cancer-cell-derived S100A8 triggers the intracellular RhoA-ROCK-MLC2-MRTF-A pathway by binding to the CD147 receptor of CAFs, inducing CAF polarization and leading to chemoresistance. Therapeutically, we demonstrate that blocking the S100A8-CD147 pathway can improve chemotherapy efficiency. Prognostically, we found the S100A8 levels in peripheral blood can serve as an indicator of chemotherapy responsiveness. Collectively, our study offers a comprehensive understanding of the molecular mechanisms underlying chemoresistance in esophageal cancer and highlights the potential value of S100A8 in the clinical management of esophageal cancer.


Sujet(s)
Calgranuline A , Fibroblastes associés au cancer , Résistance aux médicaments antinéoplasiques , Tumeurs de l'oesophage , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Humains , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/traitement médicamenteux , Tumeurs de l'oesophage/métabolisme , Tumeurs de l'oesophage/génétique , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Calgranuline A/métabolisme , Calgranuline A/génétique , Animaux , Souris , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Antigènes CD147/métabolisme , Antigènes CD147/génétique , Protéine G RhoA/métabolisme , Protéine G RhoA/génétique , Carcinome épidermoïde de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/traitement médicamenteux , Carcinome épidermoïde de l'oesophage/métabolisme , Carcinome épidermoïde de l'oesophage/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Femelle
13.
FEBS Open Bio ; 14(6): 983-1000, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38693086

RÉSUMÉ

Myocardial infarction results in extensive cardiomyocyte apoptosis, leading to the formation of noncontractile scar tissue. Given the limited regenerative capacity of adult mammalian cardiomyocytes, direct reprogramming of cardiac fibroblasts (CFs) into cardiomyocytes represents a promising therapeutic strategy for myocardial repair, and small molecule drugs might offer a more attractive alternative to gene editing approaches in terms of safety and clinical feasibility. This study aimed to reprogram rat CFs into cardiomyocytes using a small molecular chemical mixture comprising CHIR99021, Valproic acid, Dorsomorphin, SB431542, and Forskolin. Immunofluorescence analysis revealed a significant increase in the expression of cardiomyocyte-specific markers, including cardiac troponin T (cTnT), Connexin 43 (Cx43), α-actinin, and Tbx5. Changes in intracellular calcium ion levels and Ca2+ signal transfer between adjacent cells were monitored using a calcium ion fluorescence probe. mRNA sequencing analysis demonstrated the upregulation of genes associated with cardiac morphogenesis, myocardial differentiation, and muscle fiber contraction during CF differentiation induced by the small-molecule compounds. Conversely, the expression of fibroblast-related genes was downregulated. These findings suggest that chemical-induced cell fate conversion of rat CFs into cardiomyocyte-like cells is feasible, offering a potential therapeutic solution for myocardial injury.


Sujet(s)
Différenciation cellulaire , Reprogrammation cellulaire , Fibroblastes , Myocytes cardiaques , Animaux , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/cytologie , Rats , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/génétique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Bibliothèques de petites molécules/pharmacologie , Rat Sprague-Dawley , Calcium/métabolisme
14.
Front Immunol ; 15: 1375461, 2024.
Article de Anglais | MEDLINE | ID: mdl-38711514

RÉSUMÉ

Excess dietary fructose consumption has been long proposed as a culprit for the world-wide increase of incidence in metabolic disorders and cancer within the past decades. Understanding that cancer cells can gradually accumulate metabolic mutations in the tumor microenvironment, where glucose is often depleted, this raises the possibility that fructose can be utilized by cancer cells as an alternative source of carbon. Indeed, recent research has increasingly identified various mechanisms that show how cancer cells can metabolize fructose to support their proliferating and migrating needs. In light of this growing interest, this review will summarize the recent advances in understanding how fructose can metabolically reprogram different types of cancer cells, as well as how these metabolic adaptations can positively support cancer cells development and malignancy.


Sujet(s)
Fructose , Tumeurs , Microenvironnement tumoral , Humains , Fructose/métabolisme , Fructose/effets indésirables , Tumeurs/métabolisme , Tumeurs/étiologie , Animaux , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Métabolisme énergétique/effets des médicaments et des substances chimiques ,
15.
J Neuroinflammation ; 21(1): 137, 2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38802820

RÉSUMÉ

Hyperglycemia has been shown to modulate the immune response of peripheral immune cells and organs, but the impact of hyperglycemia on neuroinflammation within the brain remains elusive. In the present study, we provide evidences that streptozotocin (STZ)-induced hyperglycemic condition in mice drives a phenotypic switch of brain astrocytes to a proinflammatory state, and increases brain vulnerability to mild peripheral inflammation. In particular, we found that hyperglycemia led to a significant increase in the astrocyte proliferation as determined by flow cytometric and immunohistochemical analyses of mouse brain. The increased astrocyte proliferation by hyperglycemia was reduced by Glut1 inhibitor BAY-876. Transcriptomic analysis of isolated astrocytes from Aldh1l1CreERT2;tdTomato mice revealed that peripheral STZ injection induced astrocyte reprogramming into proliferative, and proinflammatory phenotype. Additionally, STZ-induced hyperglycemic condition significantly enhanced the infiltration of circulating myeloid cells into the brain and the disruption of blood-brain barrier in response to mild lipopolysaccharide (LPS) administration. Systemic hyperglycemia did not alter the intensity and sensitivity of peripheral inflammation in mice to LPS challenge, but increased the inflammatory potential of brain microglia. In line with findings from mouse experiments, a high-glucose environment intensified the LPS-triggered production of proinflammatory molecules in primary astrocyte cultures. Furthermore, hyperglycemic mice exhibited a significant impairment in cognitive function after mild LPS administration compared to normoglycemic mice as determined by novel object recognition and Y-maze tasks. Taken together, these results demonstrate that hyperglycemia directly induces astrocyte reprogramming towards a proliferative and proinflammatory phenotype, which potentiates mild LPS-triggered inflammation within brain parenchymal regions.


Sujet(s)
Astrocytes , Encéphale , Hyperglycémie , Lipopolysaccharides , Souris de lignée C57BL , Maladies neuro-inflammatoires , Animaux , Hyperglycémie/induit chimiquement , Hyperglycémie/anatomopathologie , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Souris , Lipopolysaccharides/toxicité , Lipopolysaccharides/pharmacologie , Encéphale/anatomopathologie , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/anatomopathologie , Maladies neuro-inflammatoires/induit chimiquement , Mâle , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/physiologie , Souris transgéniques , Cellules cultivées
16.
Cell Rep ; 43(5): 114176, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38691454

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) carries a dismal prognosis due to therapeutic resistance. We show that PDAC cells undergo global epigenetic reprogramming to acquire chemoresistance, a process that is driven at least in part by protein arginine methyltransferase 1 (PRMT1). Genetic or pharmacological PRMT1 inhibition impairs adaptive epigenetic reprogramming and delays acquired resistance to gemcitabine and other common chemo drugs. Mechanistically, gemcitabine treatment induces translocation of PRMT1 into the nucleus, where its enzymatic activity limits the assembly of chromatin-bound MAFF/BACH1 transcriptional complexes. Cut&Tag chromatin profiling of H3K27Ac, MAFF, and BACH1 suggests a pivotal role for MAFF/BACH1 in global epigenetic response to gemcitabine, which is confirmed by genetically silencing MAFF. PRMT1 and MAFF/BACH1 signature genes identified by Cut&Tag analysis distinguish gemcitabine-resistant from gemcitabine-sensitive patient-derived xenografts of PDAC, supporting the PRMT1-MAFF/BACH1 epigenetic regulatory axis as a potential therapeutic avenue for improving the efficacy and durability of chemotherapies in patients of PDAC.


Sujet(s)
Désoxycytidine , Résistance aux médicaments antinéoplasiques , Épigenèse génétique , , Tumeurs du pancréas , Protein-arginine N-methyltransferases , Protéines de répression , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Humains , Résistance aux médicaments antinéoplasiques/génétique , Tumeurs du pancréas/génétique , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/anatomopathologie , Désoxycytidine/analogues et dérivés , Désoxycytidine/pharmacologie , Désoxycytidine/usage thérapeutique , Protéines de répression/métabolisme , Protéines de répression/génétique , Lignée cellulaire tumorale , Animaux , Souris , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/anatomopathologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/génétique
17.
Acta Biomater ; 181: 308-316, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38570107

RÉSUMÉ

Acute lung injury (ALI) represents a critical respiratory condition typified by rapid-onset lung inflammation, contributing to elevated morbidity and mortality rates. Central to ALI pathogenesis lies macrophage dysfunction, characterized by an overabundance of pro-inflammatory cytokines and a shift in metabolic activity towards glycolysis. This study emphasizes the crucial function of glucose metabolism in immune cell function under inflammatory conditions and identifies hexokinase 2 (HK2) as a key regulator of macrophage metabolism and inflammation. Given the limitations of HK2 inhibitors, we propose the CRISPR/Cas9 system for precise HK2 downregulation. We developed an aerosolized core-shell liposomal nanoplatform (CSNs) complexed with CaP for efficient drug loading, targeting lung macrophages. Various CSNs were synthesized to encapsulate an mRNA based CRISPR/Cas9 system (mCas9/gHK2), and their gene editing efficiency and HK2 knockout were examined at both gene and protein levels in vitro and in vivo. The CSN-mCas9/gHK2 treatment demonstrated a significant reduction in glycolysis and inflammation in macrophages. In an LPS-induced ALI mouse model, inhaled CSN-mCas9/gHK2 mitigated the proinflammatory tumor microenvironment and reprogrammed glucose metabolism in the lung, suggesting a promising strategy for ALI prevention and treatment. This study highlights the potential of combining CRISPR/Cas9 gene editing with inhalation delivery systems for effective, localized pulmonary disease treatment, underscoring the importance of targeted gene modulation and metabolic reprogramming in managing ALI. STATEMENT OF SIGNIFICANCE: This study investigates an inhalable CRISPR/Cas9 gene editing system targeting pulmonary macrophages, with the aim of modulating glucose metabolism to alleviate Acute Lung Injury (ALI). The research highlights the role of immune cell metabolism in inflammation, as evidenced by changes in macrophage glucose metabolism and a notable reduction in pulmonary edema and inflammation. Additionally, observed alterations in macrophage polarization and cytokine levels in bronchoalveolar lavage fluid suggest potential therapeutic implications. These findings not only offer insights into possible ALI treatments but also contribute to the understanding of immune cell metabolism in inflammatory diseases, which could be relevant for various inflammatory and metabolic disorders.


Sujet(s)
Lésion pulmonaire aigüe , Systèmes CRISPR-Cas , Hexokinase , Lésion pulmonaire aigüe/anatomopathologie , Lésion pulmonaire aigüe/thérapie , Animaux , Souris , Hexokinase/génétique , Hexokinase/métabolisme , Souris de lignée C57BL , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Administration par inhalation , Liposomes/composition chimique , Cellules RAW 264.7 , Mâle , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Édition de gène , Glycolyse/effets des médicaments et des substances chimiques
18.
Cell Rep ; 43(4): 114054, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38578832

RÉSUMÉ

Cell fate conversion is associated with extensive post-translational modifications (PTMs) and architectural changes of sub-organelles, yet how these events are interconnected remains unknown. We report here the identification of a phosphorylation code in 14-3-3 binding motifs (PC14-3-3) that greatly stimulates induced cardiomyocyte (iCM) formation from fibroblasts. PC14-3-3 is identified in pivotal functional proteins for iCM reprogramming, including transcription factors and chromatin modifiers. Akt1 kinase and protein phosphatase 2A are the key writer and key eraser of the PC14-3-3 code, respectively. PC14-3-3 activation induces iCM formation with the presence of only Tbx5. In contrast, PC14-3-3 inhibition by mutagenesis or inhibitor-mediated code removal abolishes reprogramming. We discover that key PC14-3-3-embedded factors, such as histone deacetylase 4 (Hdac4), Mef2c, and Foxo1, form Hdac4-organized inhibitory nuclear condensates. PC14-3-3 activation disrupts Hdac4 condensates to promote cardiac gene expression. Our study suggests that sub-organelle dynamics regulated by a PTM code could be a general mechanism for stimulating cell reprogramming.


Sujet(s)
Protéines 14-3-3 , Reprogrammation cellulaire , Histone deacetylases , Myocytes cardiaques , Protéines 14-3-3/métabolisme , Histone deacetylases/métabolisme , Phosphorylation , Animaux , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Souris , Humains , Fibroblastes/métabolisme , Facteurs de transcription MEF2/métabolisme , Motifs d'acides aminés , Liaison aux protéines
19.
Immunopharmacol Immunotoxicol ; 46(3): 330-340, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38478467

RÉSUMÉ

Rheumatoid arthritis (RA) is a persistent autoimmune condition characterized by ongoing inflammation primarily affecting the synovial joint. This inflammation typically arises from an increase in immune cells such as neutrophils, macrophages, and T cells (TC). TC is recognized as a major player in RA pathogenesis. The involvement of HLA-DRB1 and PTPN-2 among RA patients confirms the TC involvement in RA. Metabolism of TC is maintained by various other factors like cytokines, mitochondrial proteins & other metabolites. Different TC subtypes utilize different metabolic pathways like glycolysis, oxidative phosphorylation and fatty acid oxidation for their activation from naive TC (T0). Although all subsets of TC are not deleterious for synovium, some subsets of TC are involved in joint repair using their anti-inflammatory properties. Hence artificially reprogramming of TC subset by interfering with their metabolic status poised a hope in future to design new molecules against RA.


Sujet(s)
Polyarthrite rhumatoïde , Humains , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Animaux , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lymphocytes T/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/immunologie
20.
Cell Prolif ; 57(7): e13612, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38348888

RÉSUMÉ

Ageing and cell senescence of mesenchymal stem cells (MSCs) limited their immunomodulation properties and therapeutic application. We previously reported that nucleosome assembly protein 1-like 2 (Nap1l2) contributes to MSCs senescence and osteogenic differentiation. Here, we sought to evaluate whether Nap1l2 impairs the immunomodulatory properties of MSCs and find a way to rescue the deficient properties. We demonstrated that metformin could rescue the impaired migration properties and T cell regulation properties of OE-Nap1l2 BMSCs. Moreover, metformin could improve the impaired therapeutic efficacy of OE-Nap1l2 BMSCs in the treatment of colitis and experimental autoimmune encephalomyelitis in mice. Mechanistically, metformin was capable of upregulating the activation of AMPK, synthesis of l-arginine and expression of inducible nitric oxide synthase in OE-Nap1l2 BMSCs, leading to an increasing level of nitric oxide. This study indicated that Nap1l2 negatively regulated the immunomodulatory properties of BMSCs and that the impaired functions could be rescued by metformin pretreatment via metabolic reprogramming. This strategy might serve as a practical therapeutic option to rescue impaired MSCs functions for further application.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Immunomodulation , Cellules souches mésenchymateuses , Metformine , Souris de lignée C57BL , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/cytologie , Animaux , Metformine/pharmacologie , Souris , Immunomodulation/effets des médicaments et des substances chimiques , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Encéphalomyélite auto-immune expérimentale/métabolisme , Encéphalomyélite auto-immune expérimentale/immunologie , Colite/traitement médicamenteux , Colite/métabolisme , Nitric oxide synthase type II/métabolisme , Monoxyde d'azote/métabolisme , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Cellules cultivées , Mouvement cellulaire/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/cytologie ,
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE