Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 34
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Nat Commun ; 15(1): 5767, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982045

RÉSUMÉ

Multiple myeloma (MM) is a hematologic malignancy characterized by uncontrolled proliferation of plasma cells in the bone marrow. MM patients with aggressive progression have poor survival, emphasizing the urgent need for identifying new therapeutic targets. Here, we show that the leukocyte immunoglobulin-like receptor B1 (LILRB1), a transmembrane receptor conducting negative immune response, is a top-ranked gene associated with poor prognosis in MM patients. LILRB1 deficiency inhibits MM progression in vivo by enhancing the ferroptosis of MM cells. Mechanistic studies reveal that LILRB1 forms a complex with the low-density lipoprotein receptor (LDLR) and LDLR adapter protein 1 (LDLRAP1) to facilitate LDL/cholesterol uptake. Loss of LILRB1 impairs cholesterol uptake but activates the de novo cholesterol synthesis pathway to maintain cellular cholesterol homeostasis, leading to the decrease of anti-ferroptotic metabolite squalene. Our study uncovers the function of LILRB1 in regulating cholesterol metabolism and protecting MM cells from ferroptosis, implicating LILRB1 as a promising therapeutic target for MM patients.


Sujet(s)
Cholestérol , Ferroptose , Homéostasie , Récepteur B1 de type immunoglobuline des leucocytes , Myélome multiple , Récepteurs aux lipoprotéines LDL , Humains , Myélome multiple/métabolisme , Myélome multiple/anatomopathologie , Myélome multiple/génétique , Récepteur B1 de type immunoglobuline des leucocytes/métabolisme , Ferroptose/génétique , Cholestérol/métabolisme , Récepteurs aux lipoprotéines LDL/métabolisme , Récepteurs aux lipoprotéines LDL/génétique , Animaux , Lignée cellulaire tumorale , Souris , Antigènes CD
2.
Stem Cell Res ; 78: 103467, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38861774

RÉSUMÉ

Dilated cardiomyopathy (DCM) is one of the main causes of sudden cardiac death and heart failure and is the leading indication for cardiac transplantation worldwide. Mutations in dozens of cardiac genes have been connected to the development of DCM including the Troponin T2 gene (TNNT2). Here, we generated a human induced pluripotent stem cells (hiPSCs) from a DCM patient with a familial history that carries a missense mutation in TNNT2. The hiPSCs show typical morphology of pluripotent stem cells, expression of pluripotency markers, normal karyotype, and in vitro capacity to differentiate into all three germ layers.


Sujet(s)
Cardiomyopathie dilatée , Cellules souches pluripotentes induites , Troponine T , Humains , Cardiomyopathie dilatée/anatomopathologie , Cellules souches pluripotentes induites/métabolisme , Troponine T/métabolisme , Troponine T/génétique , Différenciation cellulaire , Lignée cellulaire , Mâle , Caryotype
3.
Signal Transduct Target Ther ; 9(1): 86, 2024 Apr 08.
Article de Anglais | MEDLINE | ID: mdl-38584163

RÉSUMÉ

During spaceflight, the cardiovascular system undergoes remarkable adaptation to microgravity and faces the risk of cardiac remodeling. Therefore, the effects and mechanisms of microgravity on cardiac morphology, physiology, metabolism, and cellular biology need to be further investigated. Since China started constructing the China Space Station (CSS) in 2021, we have taken advantage of the Shenzhou-13 capsule to send human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) to the Tianhe core module of the CSS. In this study, hPSC-CMs subjected to space microgravity showed decreased beating rate and abnormal intracellular calcium cycling. Metabolomic and transcriptomic analyses revealed a battery of metabolic remodeling of hPSC-CMs in spaceflight, especially thiamine metabolism. The microgravity condition blocked the thiamine intake in hPSC-CMs. The decline of thiamine utilization under microgravity or by its antagonistic analog amprolium affected the process of the tricarboxylic acid cycle. It decreased ATP production, which led to cytoskeletal remodeling and calcium homeostasis imbalance in hPSC-CMs. More importantly, in vitro and in vivo studies suggest that thiamine supplementation could reverse the adaptive changes induced by simulated microgravity. This study represents the first astrobiological study on the China Space Station and lays a solid foundation for further aerospace biomedical research. These data indicate that intervention of thiamine-modified metabolic reprogramming in human cardiomyocytes during spaceflight might be a feasible countermeasure against microgravity.


Sujet(s)
Cellules souches pluripotentes , Impesanteur , Humains , , Myocytes cardiaques/métabolisme , Calcium/métabolisme , Différenciation cellulaire , Cellules souches pluripotentes/métabolisme
4.
Stem Cell Res ; 76: 103370, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38428347

RÉSUMÉ

Restrictive cardiomyopathy (RCM) is a rare cardiomyopathy characterized by diastolic dysfunction, which affects cardiac systolic function. We successfully established human induced pluripotent stem cells (hiPSCs) from peripheral blood mononuclear cells of 24-year-old male with restrictive cardiomyopathy (RCM). The patient-derived hiPSCs carried heterozygous mutation of CRYAB gene (c.326A > G, p.D109G), which was consistent with clinical whole exon sequencing results. We confirmed the pluripotency, multipotential differentiation and karyotype of hiPSCs. The hiPSCs will be useful for studying the pathogenesis of RCM caused by CRYAB (c.326A > G) mutation.


Sujet(s)
Cardiomyopathies , Cardiomyopathie restrictive , Cellules souches pluripotentes induites , Humains , Mâle , Jeune adulte , Cardiomyopathies/génétique , Cardiomyopathie restrictive/génétique , Agranulocytes , Mutation/génétique
5.
Genes Dis ; 11(3): 101057, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38292200

RÉSUMÉ

Mitochondrial diseases are a heterogeneous group of inherited disorders characterized by mitochondrial dysfunction, and these diseases are often severe or even fatal. Mitochondrial diseases are often caused by mitochondrial DNA mutations. Currently, there is no curative treatment for patients with pathogenic mitochondrial DNA mutations. With the rapid development of traditional gene editing technologies, such as zinc finger nucleases and transcription activator-like effector nucleases methods, there has been a search for a mitochondrial gene editing technology that can edit mutated mitochondrial DNA; however, there are still some problems hindering the application of these methods. The discovery of the DddA-derived cytosine base editor has provided hope for mitochondrial gene editing. In this paper, we will review the progress in the research on several mitochondrial gene editing technologies with the hope that this review will be useful for further research on mitochondrial gene editing technologies to optimize the treatment of mitochondrial diseases in the future.

6.
Nat Biotechnol ; 42(1): 99-108, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37012448

RÉSUMÉ

RNA velocity provides an approach for inferring cellular state transitions from single-cell RNA sequencing (scRNA-seq) data. Conventional RNA velocity models infer universal kinetics from all cells in an scRNA-seq experiment, resulting in unpredictable performance in experiments with multi-stage and/or multi-lineage transition of cell states where the assumption of the same kinetic rates for all cells no longer holds. Here we present cellDancer, a scalable deep neural network that locally infers velocity for each cell from its neighbors and then relays a series of local velocities to provide single-cell resolution inference of velocity kinetics. In the simulation benchmark, cellDancer shows robust performance in multiple kinetic regimes, high dropout ratio datasets and sparse datasets. We show that cellDancer overcomes the limitations of existing RNA velocity models in modeling erythroid maturation and hippocampus development. Moreover, cellDancer provides cell-specific predictions of transcription, splicing and degradation rates, which we identify as potential indicators of cell fate in the mouse pancreas.


Sujet(s)
ARN , Analyse sur cellule unique , Animaux , Souris , ARN/génétique , Analyse de séquence d'ARN/méthodes , Analyse sur cellule unique/méthodes , Simulation numérique , , Analyse de profil d'expression de gènes/méthodes
7.
Zool Res ; 43(6): 1011-1022, 2022 Nov 18.
Article de Anglais | MEDLINE | ID: mdl-36266925

RÉSUMÉ

The evolutionary and functional features of RNA editing are well studied in mammals, cephalopods, and insects, but not in birds. Here, we integrated transcriptomic and whole-genomic analyses to exhaustively characterize the expansive repertoire of adenosine-to-inosine (A-to-I) RNA editing sites (RESs) in the chicken. In addition, we investigated the evolutionary status of the chicken editome as a potential mechanism of domestication. We detected the lowest editing level in the liver of chickens, compared to muscles in humans, and found higher editing activity and specificity in the brain than in non-neural tissues, consistent with the brain's functional complexity. To a certain extent, specific editing activity may account for the specific functions of tissues. Our results also revealed that sequences critical to RES secondary structures remained conserved within avian evolution. Furthermore, the RNA editome was shaped by purifying selection during chicken domestication and most RESs may have served as a selection pool for a few functional RESs involved in chicken domestication, including evolution of nervous and immune systems. Regulation of RNA editing in chickens by adenosine deaminase acting on RNA (ADAR) enzymes may be affected by non-ADAR factors whose expression levels changed widely after ADAR knockdown. Collectively, we provide comprehensive lists of candidate RESs and non-ADAR-editing regulators in the chicken, thus contributing to our current understanding of the functions and evolution of RNA editing in animals.


Sujet(s)
Adenosine deaminase , Poulets , Édition des ARN , Animaux , Humains , Adénosine/génétique , Adenosine deaminase/génétique , Adenosine deaminase/métabolisme , Poulets/génétique , Génomique , Inosine/génétique , ARN/génétique , Transcriptome
8.
JCI Insight ; 7(19)2022 10 10.
Article de Anglais | MEDLINE | ID: mdl-36040812

RÉSUMÉ

The proteasome inhibitors (PIs) bortezomib and carfilzomib, which target proteasome 20S subunit beta 5 (PSMB5) in cells, are widely used in multiple myeloma (MM) treatment. In this study, we demonstrated the role of interferon-stimulated 20 kDa exonuclease-like 2 (ISG20L2) in MM PI resistance. Gain- and loss-of-function studies showed that ISG20L2 suppressed MM cell sensitivity to PIs in vitro and in vivo. Patients with ISG20L2lo MM had a better response to PIs and a longer overall survival than patients with ISG20L2hi MM. Biotinylated bortezomib pull-down assays showed that ISG20L2 competed with PSMB5 in binding to bortezomib. The surface plasmon resonance assay confirmed the direct binding of bortezomib to ISG20L2. In ISG20L2hi MM cells, ISG20L2 attenuated the binding of bortezomib to PSMB5, resulting in lower inhibition of proteasome activity and therefore less bortezomib-induced cell death. Overall, we identified a potentially novel mechanism by which ISG20L2 conferred bortezomib resistance on MM. The expression of ISG20L2 correlated with MM PI responses and patient treatment outcomes.


Sujet(s)
Myélome multiple , Inhibiteurs du protéasome , Acides boroniques/pharmacologie , Bortézomib/pharmacologie , Bortézomib/usage thérapeutique , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques , Exonucleases , Humains , Interférons , Myélome multiple/traitement médicamenteux , Proteasome endopeptidase complex/métabolisme , Inhibiteurs du protéasome/pharmacologie , Pyrazines
9.
J Hematol Oncol ; 15(1): 55, 2022 05 07.
Article de Anglais | MEDLINE | ID: mdl-35526043

RÉSUMÉ

Lysophosphatidic acid (LPA) is a naturally occurring phospholipid that regulates cell proliferation, survival, and migration. However, its role on human multiple myeloma (MM) cells is largely unknown. In this study, we show that LPA, which is highly elevated in MM patients, plays an important role in protecting human MM cells against proteasome inhibitor (PI)-induced apoptosis. LPA bound to its receptor LPAR2 activated its downstream MEK1/2-ERK1/2 signaling pathway and enhanced oxidative phosphorylation (OXPHOS) in mitochondria in MM cells. Increased OXPHOS activity produced more NAD+ and ATP, reduced proteasome activity, and enhanced protein folding and refolding in endoplasmic reticulum (ER), leading to induction of MM resistance to PIs. Importantly, inhibiting LPAR2 activity or knocking out LPAR2 in MM cells significantly enhanced MM sensitivity to PI-induced apoptosis in vitro and in vivo. Interestingly, primary MM cells from LPA-high patients were more resistant to PI-induced apoptosis than MM cells from LPA-low patients. Thus, our study indicates that LPA-LPAR2-mediated signaling pathways play an important role in MM sensitivity to PIs and targeting LPA or LPAR2 may potentially be used to (re)sensitize patients to PI-based therapy.


Sujet(s)
Myélome multiple , Inhibiteurs du protéasome , Apoptose , Humains , Lysophospholipides/métabolisme , Lysophospholipides/pharmacologie , Myélome multiple/traitement médicamenteux , Myélome multiple/métabolisme
10.
J Clin Invest ; 132(7)2022 04 01.
Article de Anglais | MEDLINE | ID: mdl-35192544

RÉSUMÉ

CD8+ T cell longevity regulated by metabolic activity plays important roles in cancer immunotherapy. Although in vitro-polarized, transferred IL-9-secreting CD8+ Tc9 (cytotoxic T lymphocyte subset 9) cells exert greater persistence and antitumor efficacy than Tc1 cells, the underlying mechanism remains unclear. Here, we show that tumor-infiltrating Tc9 cells display significantly lower lipid peroxidation than Tc1 cells in several mouse models, which is strongly correlated with their persistence. Using RNA-sequence and functional validation, we found that Tc9 cells exhibited unique lipid metabolic programs. Tc9 cell-derived IL-9 activated STAT3, upregulated fatty acid oxidation and mitochondrial activity, and rendered Tc9 cells with reduced lipid peroxidation and resistance to tumor- or ROS-induced ferroptosis in the tumor microenvironment. IL-9 signaling deficiency, inhibiting STAT3, or fatty acid oxidation increased lipid peroxidation and ferroptosis of Tc9 cells, resulting in impaired longevity and antitumor ability. Similarly, human Tc9 cells also exhibited lower lipid peroxidation than Tc1 cells and tumor-infiltrating CD8+ T cells expressed lower IL9 and higher lipid peroxidation- and ferroptosis-related genes than circulating CD8+ T cells in patients with melanoma. This study indicates that lipid peroxidation regulates Tc9 cell longevity and antitumor effects via the IL-9/STAT3/fatty acid oxidation pathway and regulating T cell lipid peroxidation can be used to enhance T cell-based immunotherapy in human cancer.


Sujet(s)
Lymphocytes T CD8+ , Interleukine-9 , Animaux , Lymphocytes T CD8+/métabolisme , Acides gras/métabolisme , Humains , Immunothérapie/méthodes , Interleukine-9/génétique , Peroxydation lipidique , Souris , Facteur de transcription STAT-3/génétique , Facteur de transcription STAT-3/métabolisme
11.
J Clin Invest ; 132(4)2022 02 15.
Article de Anglais | MEDLINE | ID: mdl-35166240

RÉSUMÉ

The chromosomal t(4;14) (p16;q32) translocation drives high expression of histone methyltransferase nuclear SET domain-containing 2 (NSD2) and plays vital roles in multiple myeloma (MM) evolution and progression. However, the mechanisms of NSD2-driven epigenomic alterations in chemoresistance to proteasome inhibitors (PIs) are not fully understood. Using a CRISPR/Cas9 sgRNA library in a bone marrow-bearing MM model, we found that hepatoma-derived growth factor 2 (HRP2) was a suppressor of chemoresistance to PIs and that its downregulation correlated with a poor response and worse outcomes in the clinic. We observed suppression of HRP2 in bortezomib-resistant MM cells, and knockdown of HRP2 induced a marked tolerance to PIs. Moreover, knockdown of HRP2 augmented H3K27me3 levels, consequentially intensifying transcriptome alterations promoting cell survival and restriction of ER stress. Mechanistically, HRP2 recognized H3K36me2 and recruited the histone demethylase MYC-induced nuclear antigen (MINA) to remove H3K27me3. Tazemetostat, a highly selective epigenetic inhibitor that reduces H3K27me3 levels, synergistically sensitized the anti-MM effects of bortezomib both in vitro and in vivo. Collectively, these results provide a better understanding of the origin of chemoresistance in patients with MM with the t(4;14) translocation and a rationale for managing patients with MM who have different genomic backgrounds.


Sujet(s)
Reprogrammation cellulaire , Chromosomes humains de la paire 14/génétique , Chromosomes humains de la paire 4/génétique , Dioxygenases , Épigenèse génétique/effets des médicaments et des substances chimiques , Histone Demethylases , Myélome multiple , Protéines tumorales , Protéines nucléaires , Inhibiteurs du protéasome/pharmacologie , Translocation génétique , Lignée cellulaire tumorale , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/génétique , Dioxygenases/génétique , Dioxygenases/métabolisme , Épigénomique , Histone Demethylases/génétique , Histone Demethylases/métabolisme , Humains , Myélome multiple/traitement médicamenteux , Myélome multiple/génétique , Myélome multiple/métabolisme , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme
12.
Acta Pharmacol Sin ; 43(1): 240-250, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-33686244

RÉSUMÉ

Cardiovascular safety assessment is vital for drug development, yet human cardiovascular cell models are lacking. In vitro mass-generated human pluripotent stem cell (hPSC)-derived cardiovascular cells are a suitable cell model for preclinical cardiovascular safety evaluations. In this study, we established a preclinical toxicology model using same-origin hPSC-differentiated cardiomyocytes (hPSC-CMs) and endothelial cells (hPSC-ECs). For validation of this cell model, alirocumab, a human antibody against proprotein convertase subtilisin kexin type 9 (PCSK9), was selected as an emerging safe lipid-lowering drug; atorvastatin, a common statin (the most effective type of lipid-lowering drug), was used as a drug with reported side effects at high concentrations, while doxorubicin was chosen as a positive cardiotoxic drug. The cytotoxicity of these drugs was assessed using CCK8, ATP, and lactate dehydrogenase release assays at 24, 48, and 72 h. The influences of these drugs on cardiomyocyte electrophysiology were detected using the patch-clamp technique, while their effects on endothelial function were determined by tube formation and Dil-acetylated low-density lipoprotein (Dil-Ac-LDL) uptake assays. We showed that alirocumab did not affect the cell viability or cardiomyocyte electrophysiology in agreement with the clinical results. Atorvastatin (5-50 µM) dose-dependently decreased cardiovascular cell viability over time, and at a high concentration (50 µM, ~100 times the normal peak serum concentration in clinic), it affected the action potentials of hPSC-CMs and damaged tube formation and Dil-Ac-LDL uptake of hPSC-ECs. The results demonstrate that the established same-origin hPSC-derived cardiovascular cell model can be used to evaluate lipid-lowering drug safety in cardiovascular cells and allow highly accurate preclinical assessment of potential drugs.


Sujet(s)
Anticholestérolémiants/pharmacologie , Atorvastatine/pharmacologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Myocytes cardiaques/effets des médicaments et des substances chimiques , Anticholestérolémiants/composition chimique , Atorvastatine/composition chimique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Humains , Modèles moléculaires , Structure moléculaire , Relation structure-activité
13.
Cardiovasc Res ; 118(3): 859-871, 2022 02 21.
Article de Anglais | MEDLINE | ID: mdl-33956078

RÉSUMÉ

AIMS: Congenital heart disease (CHD) frequently occurs in newborns due to abnormal formation of the heart or major blood vessels. Mutations in the GATA4 gene, which encodes GATA binding protein 4, are responsible for atrial septal defect (ASD), a common CHD. This study aims to gain insights into the molecular mechanisms of CHD using human-induced pluripotent stem cells (iPSCs) from a family cohort with ASD. METHODS AND RESULTS: Patient-specific iPSCs possess the same genetic information as the donor and can differentiate into various cell types from all three germ layers in vitro, thus presenting a promising approach for disease modelling and molecular mechanism research. Here, we generated a patient-specific iPSC line (iPSC-G4T280M) from a family cohort carrying a hereditary ASD mutation in GATA4 gene (T280M), as well as a human embryonic stem cell line (ESC-G4T280M) carrying the isogenic T280M mutation using the CRISPR/Cas9 genome editing method. The GATA4-mutant iPSCs and ESCs were then differentiated into cardiomyocytes (CMs) to model GATA4 mutation-associated ASD. We observed an obvious defect in cell proliferation in cardiomyocytes derived from both GATA4T280M-mutant iPSCs (iPSC-G4T280M-CMs) and ESCs (ESC-G4T280M-CMs), while the impaired proliferation ability of iPSC-G4T280M-CMs could be restored by gene correction. Integrated analysis of RNA-Seq and ChIP-Seq data indicated that FGF16 is a direct target of wild-type GATA4. However, the T280M mutation obstructed GATA4 occupancy at the FGF16 promoter region, leading to impaired activation of FGF16 transcription. Overexpression of FGF16 in GATA4-mutant cardiomyocytes rescued the cell proliferation defect. The direct relationship between GATA4T280M and ASD was demonstrated in a human iPSC model for the first time. CONCLUSIONS: In summary, our study revealed the molecular mechanism of the GATA4T280M mutation in ASD. Understanding the roles of the GATA4-FGF16 axis in iPSC-CMs will shed light on heart development and provide novel insights for the treatment of ASD and other CHD disorders.


Sujet(s)
Facteurs de croissance fibroblastique , Communications interauriculaires , Cellules souches pluripotentes induites , Lignée cellulaire , Cellules souches embryonnaires , Facteurs de croissance fibroblastique/génétique , Facteur de transcription GATA-4/génétique , Communications interauriculaires/génétique , Communications interauriculaires/métabolisme , Humains , Cellules souches pluripotentes induites/métabolisme , Nouveau-né , Mutation , Myocytes cardiaques/métabolisme
14.
Methods Mol Biol ; 2549: 299-305, 2022.
Article de Anglais | MEDLINE | ID: mdl-34611814

RÉSUMÉ

In the past decades, human induced pluripotent stem cells (iPSCs) have been generated by the ectopic expression of "Yamanaka factors" in multiple somatic cells. However, the procedure to get access to donor cells is hard or invasive in most cases. Hereon, we depict a stepwise method developed in our laboratory for the generation of iPSCs from renal epithelial cells present in urine, which is noninvasive, nonintegrating, and universal. The resulting urinary iPSCs (UiPSCs) exhibit pluripotent characteristics resemble embryonic stem cells (ESCs) and thus urine may be a favorable source for generating iPSCs.


Sujet(s)
Cellules souches pluripotentes induites , Cellules souches embryonnaires/métabolisme , Cellules épithéliales , Humains , Cellules souches pluripotentes induites/métabolisme
15.
Blood ; 139(1): 59-72, 2022 01 06.
Article de Anglais | MEDLINE | ID: mdl-34411225

RÉSUMÉ

Proteasome inhibitors (PIs) such as bortezomib (Btz) and carfilzomib (Cfz) are highly efficacious for patients with multiple myeloma (MM). However, relapses are frequent, and acquired resistance to PI treatment emerges in most patients. Here, we performed a high-throughput screen of 1855 Food and Drug Administration (FDA)-approved drugs and identified all-trans retinoic acid (ATRA), which alone has no antimyeloma effect, as a potent drug that enhanced MM sensitivity to Cfz-induced cytotoxicity and resensitized Cfz-resistant MM cells to Cfz in vitro. ATRA activated retinoic acid receptor (RAR)γ and interferon-ß response pathway, leading to upregulated expression of IRF1. IRF1 in turn initiated the transcription of OAS1, which synthesized 2-5A upon binding to double-stranded RNA (dsRNA) induced by Cfz and resulted in cellular RNA degradation by RNase L and cell death. Similar to ATRA, BMS961, a selective RARγ agonist, could also (re)sensitize MM cells to Cfz in vitro, and both ATRA and BMS961 significantly enhanced the therapeutic effects of Cfz in established MM in vivo. In support of these findings, analyses of large datasets of patients' gene profiling showed a strong and positive correlation between RARγ and OAS1 expression and patient's response to PI treatment. Thus, this study highlights the potential for RARγ agonists to sensitize and overcome MM resistance to Cfz treatment in patients.


Sujet(s)
Antinéoplasiques/pharmacologie , Immunité innée/effets des médicaments et des substances chimiques , Myélome multiple/traitement médicamenteux , Oligopeptides/pharmacologie , Récepteurs à l'acide rétinoïque/agonistes , 2',5'-Oligoadenylate synthetase/immunologie , Lignée cellulaire tumorale , Endoribonucleases/immunologie , Humains , Récepteurs à l'acide rétinoïque/immunologie , Cellules cancéreuses en culture ,
16.
Blood Adv ; 5(23): 5269-5282, 2021 12 14.
Article de Anglais | MEDLINE | ID: mdl-34592762

RÉSUMÉ

Multiple myeloma, a plasma cell malignancy in the bone marrow, remains largely incurable with currently available therapeutics. In this study, we discovered that the activated leukocyte cell adhesion molecule (ALCAM) interacted with epidermal growth factor receptor (EGFR), and regulated myelomagenesis. ALCAM was a negative regulator of myeloma clonogenicity. ALCAM expression was positively correlated with patients' survival. ALCAM-knockdown myeloma cells displayed enhanced colony formation in the presence of bone marrow stromal cells (BMSCs). BMSCs supported myeloma colony formation by secreted epidermal growth factor (EGF), which bound with its receptor (EGFR) on myeloma cells and activated Mek/Erk cell signaling, PI3K/Akt cell signaling, and hedgehog pathway. ALCAM could also bind with EGFR, block EGF from binding to EGFR, and abolish EGFR-initiated cell signaling. Hence, our study identifies ALCAM as a novel negative regulator of myeloma pathogenesis.


Sujet(s)
Molécule d'adhérence cellulaire des leucocytes activés , Protéines Hedgehog , Antigènes CD , Molécules d'adhérence cellulaire neuronale , Récepteurs ErbB/génétique , Protéines foetales , Humains , Phosphatidylinositol 3-kinases , Transduction du signal
17.
Front Cell Dev Biol ; 9: 672039, 2021.
Article de Anglais | MEDLINE | ID: mdl-34079803

RÉSUMÉ

In the past decades, researchers discovered the contribution of genetic defects to the pathogenesis of primary cardiomyopathy and tried to explain the pathogenesis of these diseases by establishing a variety of disease models. Although human heart tissues and primary cardiomyocytes have advantages in modeling human heart diseases, they are difficult to obtain and culture in vitro. Defects developed in genetically modified animal models are notably different from human diseases at the molecular level. The advent of human induced pluripotent stem cells (hiPSCs) provides an unprecedented opportunity to further investigate the pathogenic mechanisms of inherited cardiomyopathies in vitro using patient-specific hiPSC-derived cardiomyocytes. In this review, we will make a summary of recent advances in in vitro inherited cardiomyopathy modeling using hiPSCs.

18.
Stem Cell Res ; 53: 102393, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-34088018

RÉSUMÉ

Heterozygous T280M mutation in the GATA4 gene, encoding GATA binding protein 4, was recently identified in patients with congenital heart disease (CHD). Here, a human embryonic stem cell line carrying a heterozygous GATA4T280M mutation was generated by using the CRISPR/Cas9 and piggyBac technologies. This GATA4T280M cell line remains normal morphology, pluripotency and karyotype, and could differentiate into all three germ layers both in vivo and in vitro. Thus, the GATA4T280M cell line will be a useful platform to model GATA4-associated CHD through the induction of cardiomyocyte differentiation in vitro.


Sujet(s)
Cellules souches embryonnaires humaines , Lignée cellulaire , Cellules souches embryonnaires , Facteur de transcription GATA-4/génétique , Hétérozygote , Humains , Mutation
19.
JBMR Plus ; 5(4): e10478, 2021 Apr.
Article de Anglais | MEDLINE | ID: mdl-33869994

RÉSUMÉ

Osteoporosis and its associated fractures are highly prevalent in older women. Recent studies have shown that gut microbiota play important roles in regulating bone metabolism. A previous randomized controlled trial (RCT) found that supplementation with Lactobacillus reuteri ATCC PTA 6475 (L.reuteri) led to substantially reduced bone loss in older women with low BMD. However, the total metabolic effects of L. reuteri supplementation on older women are still not clear. In this study, a post hoc analysis (not predefined) of serum metabolomic profiles of older women from the previous RCT was performed to investigate the metabolic dynamics over 1 year and to evaluate the effects of L. reuteri supplementation on human metabolism. Distinct segregation of the L. reuteri and placebo groups in response to the treatment was revealed by partial least squares-discriminant analysis. Although no individual metabolite was differentially and significantly associated with treatment after correction for multiple testing, 97 metabolites responded differentially at any one time point between L. reuteri and placebo groups (variable importance in projection score >1 and p value <0.05). These metabolites were involved in multiple processes, including amino acid, peptide, and lipid metabolism. Butyrylcarnitine was particularly increased at all investigated time points in the L. reuteri group compared with placebo, indicating that the effects of L. reuteri on bone loss are mediated through butyrate signaling. Furthermore, the metabolomic profiles in a case (low BMD) and control population (high BMD) of elderly women were analyzed to confirm the associations between BMD and the identified metabolites regulated by L. reuteri supplementation. The amino acids, especially branched-chain amino acids, showed association with L. reuteri treatment and with low BMD in older women, and may serve as potential therapeutic targets. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

20.
Cell Metab ; 33(5): 1001-1012.e5, 2021 05 04.
Article de Anglais | MEDLINE | ID: mdl-33691090

RÉSUMÉ

Understanding the mechanisms underlying how T cells become dysfunctional in a tumor microenvironment (TME) will greatly benefit cancer immunotherapy. We found that increased CD36 expression in tumor-infiltrating CD8+ T cells, which was induced by TME cholesterol, was associated with tumor progression and poor survival in human and murine cancers. Genetic ablation of Cd36 in effector CD8+ T cells exhibited increased cytotoxic cytokine production and enhanced tumor eradication. CD36 mediated uptake of fatty acids by tumor-infiltrating CD8+ T cells in TME, induced lipid peroxidation and ferroptosis, and led to reduced cytotoxic cytokine production and impaired antitumor ability. Blocking CD36 or inhibiting ferroptosis in CD8+ T cells effectively restored their antitumor activity and, more importantly, possessed greater antitumor efficacy in combination with anti-PD-1 antibodies. This study reveals a new mechanism of CD36 regulating the function of CD8+ effector T cells and therapeutic potential of targeting CD36 or inhibiting ferroptosis to restore T cell function.


Sujet(s)
Antigènes CD36/métabolisme , Lymphocytes T CD8+/immunologie , Ferroptose , Animaux , Anticorps monoclonaux humanisés/pharmacologie , Anticorps monoclonaux humanisés/usage thérapeutique , Antigènes CD36/antagonistes et inhibiteurs , Antigènes CD36/génétique , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Cytokines/métabolisme , Acides gras/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Humains , Immunothérapie , Peroxydation lipidique , Mélanome expérimental/traitement médicamenteux , Mélanome expérimental/thérapie , Souris , Souris de lignée C57BL , Souris knockout , Myélome multiple/traitement médicamenteux , Myélome multiple/mortalité , Myélome multiple/thérapie , Espèces réactives de l'oxygène/métabolisme , Taux de survie , Microenvironnement tumoral
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...