Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 31.665
Filtrer
1.
Transpl Int ; 37: 12708, 2024.
Article de Anglais | MEDLINE | ID: mdl-39100755

RÉSUMÉ

Liver transplantation is the only curative option for many liver diseases that end up in liver failure, and cholangiopathy remains a challenging complication post-liver transplant, associated with significant morbidity and potential graft loss. The low availability of organs and high demand for transplantation motivate scientists to find novel interventions. Organoids, as three-dimensional cell cultures derived from adult cells or induced pluripotent cells, may help to address this problem. Different types of organoids have been described, from which cholangiocyte organoids offer a high level of versatility and plasticity for a deeper study of liver disease mechanisms. Cholangiocytes can be obtained from different segments of the biliary tree and have shown a remarkable capacity to adapt to new environments, presenting an effective system for studying cholangiopathies. Studies using cholangiocyte organoids show promising results for disease modeling, where organoids offer fundamental features to recapitulate the complexities of tissues in vitro and uncover fundamental pathological pathways to potentially reveal therapeutic strategies for personalized medicine. Organoids could hold the potential for regeneration of injured livers, representing tools of clinical impact in regenerative medicine when tissue damage is already present.


Sujet(s)
Transplantation hépatique , Organoïdes , Humains , Transplantation hépatique/effets indésirables , Animaux , Conduits biliaires/cytologie , Foie/cytologie , Foie/anatomopathologie , Cellules souches pluripotentes induites/cytologie , Médecine régénérative/méthodes , Maladies du foie/chirurgie , Maladies du foie/thérapie , Maladies du foie/anatomopathologie
2.
Stem Cell Res Ther ; 15(1): 244, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39113154

RÉSUMÉ

The liver is the most important metabolic organ in the body. While mouse models and cell lines have further deepened our understanding of liver biology and related diseases, they are flawed in replicating key aspects of human liver tissue, particularly its complex structure and metabolic functions. The organoid model represents a major breakthrough in cell biology that revolutionized biomedical research. Organoids are in vitro three-dimensional (3D) physiological structures that recapitulate the morphological and functional characteristics of tissues in vivo, and have significant advantages over traditional cell culture methods. In this review, we discuss the generation strategies and current advances in the field focusing on their application in regenerative medicine, drug discovery and modeling diseases.


Sujet(s)
Foie , Organoïdes , Organoïdes/métabolisme , Organoïdes/cytologie , Humains , Foie/cytologie , Foie/métabolisme , Animaux , Médecine régénérative/méthodes
3.
Int J Biol Sci ; 20(9): 3544-3556, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993564

RÉSUMÉ

Hepatic progenitor cells (HPCs) have a bidirectional potential to differentiate into hepatocytes and bile duct epithelial cells and constitute a second barrier to liver regeneration in the adult liver. They are usually located in the Hering duct in the portal vein region where various cells, extracellular matrix, cytokines, and communication signals together constitute the niche of HPCs in homeostasis to maintain cellular plasticity. In various types of liver injury, different cellular signaling streams crosstalk with each other and point to the inducible transcription factor set, including FoxA1/2/3, YB-1, Foxl1, Sox9, HNF4α, HNF1α, and HNF1ß. These transcription factors exert different functions by binding to specific target genes, and their products often interact with each other, with diverse cascades of regulation in different molecular events that are essential for homeostatic regulation, self-renewal, proliferation, and selective differentiation of HPCs. Furthermore, the tumor predisposition of adult HPCs is found to be significantly increased under transcriptional factor dysregulation in transcriptional analysis, and the altered initial commitment of the differentiation pathway of HPCs may be one of the sources of intrahepatic tumors. Related transcription factors such as HNF4α and HNF1 are expected to be future targets for tumor treatment.


Sujet(s)
Différenciation cellulaire , Humains , Animaux , Cellules souches/métabolisme , Cellules souches/cytologie , Foie/métabolisme , Foie/cytologie , Hépatocytes/métabolisme , Hépatocytes/cytologie , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Transcription génétique
4.
Cells ; 13(13)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38995000

RÉSUMÉ

Erythropoiesis occurs first in the yolk sac as a transit "primitive" form, then is gradually replaced by the "definitive" form in the fetal liver (FL) during fetal development and in the bone marrow (BM) postnatally. While it is well known that differences exist between primitive and definitive erythropoiesis, the similarities and differences between FL and BM definitive erythropoiesis have not been studied. Here we performed comprehensive comparisons of erythroid progenitors and precursors at all maturational stages sorted from E16.5 FL and adult BM. We found that FL cells at all maturational stages were larger than their BM counterparts. We further found that FL BFU-E cells divided at a faster rate and underwent more cell divisions than BM BFU-E. Transcriptome comparison revealed that genes with increased expression in FL BFU-Es were enriched in cell division. Interestingly, the expression levels of glucocorticoid receptor Nr3c1, Myc and Myc downstream target Ccna2 were significantly higher in FL BFU-Es, indicating the role of the Nr3c1-Myc-Ccna2 axis in the enhanced proliferation/cell division of FL BFU-E cells. At the CFU-E stage, the expression of genes associated with hemoglobin biosynthesis were much higher in FL CFU-Es, indicating more hemoglobin production. During terminal erythropoiesis, overall temporal patterns in gene expression were conserved between the FL and BM. While biological processes related to translation, the tricarboxylic acid cycle and hypoxia response were upregulated in FL erythroblasts, those related to antiviral signal pathway were upregulated in BM erythroblasts. Our findings uncovered previously unrecognized differences between FL and BM definitive erythropoiesis and provide novel insights into erythropoiesis.


Sujet(s)
Moelle osseuse , Érythropoïèse , Foetus , Foie , Transcriptome , Animaux , Érythropoïèse/génétique , Souris , Foie/métabolisme , Foie/embryologie , Foie/cytologie , Transcriptome/génétique , Foetus/métabolisme , Foetus/cytologie , Moelle osseuse/métabolisme , Souris de lignée C57BL , Régulation de l'expression des gènes au cours du développement , Femelle , Précurseurs érythroïdes/métabolisme , Précurseurs érythroïdes/cytologie
5.
Biomaterials ; 311: 122691, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38996673

RÉSUMÉ

Acoustic holography (AH), a promising approach for cell patterning, emerges as a powerful tool for constructing novel invitro 3D models that mimic organs and cancers features. However, understanding changes in cell function post-AH remains limited. Furthermore, replicating complex physiological and pathological processes solely with cell lines proves challenging. Here, we employed acoustical holographic lattice to assemble primary hepatocytes directly isolated from mice into a cell cluster matrix to construct a liver-shaped tissue sample. For the first time, we evaluated the liver functions of AH-patterned primary hepatocytes. The patterned model exhibited large numbers of self-assembled spheroids and superior multifarious core hepatocyte functions compared to cells in 2D and traditional 3D culture models. AH offers a robust protocol for long-term in vitro culture of primary cells, underscoring its potential for future applications in disease pathogenesis research, drug testing, and organ replacement therapy.


Sujet(s)
Hépatocytes , Holographie , Foie , Hépatocytes/cytologie , Hépatocytes/métabolisme , Animaux , Foie/cytologie , Holographie/méthodes , Souris , Acoustique , Cellules cultivées , Sphéroïdes de cellules/cytologie , Souris de lignée C57BL
6.
Nature ; 632(8024): 411-418, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39048831

RÉSUMÉ

It is estimated that only 0.02% of disseminated tumour cells are able to seed overt metastases1. While this suggests the presence of environmental constraints to metastatic seeding, the landscape of host factors controlling this process remains largely unclear. Here, combining transposon technology2 and fluorescence niche labelling3, we developed an in vivo CRISPR activation screen to systematically investigate the interactions between hepatocytes and metastatic cells. We identify plexin B2 as a critical host-derived regulator of liver colonization in colorectal and pancreatic cancer and melanoma syngeneic mouse models. We dissect a mechanism through which plexin B2 interacts with class IV semaphorins on tumour cells, leading to KLF4 upregulation and thereby promoting the acquisition of epithelial traits. Our results highlight the essential role of signals from the liver parenchyma for the seeding of disseminated tumour cells before the establishment of a growth-promoting niche. Our findings further suggest that epithelialization is required for the adaptation of CRC metastases to their new tissue environment. Blocking the plexin-B2-semaphorin axis abolishes metastatic colonization of the liver and therefore represents a therapeutic strategy for the prevention of hepatic metastases. Finally, our screening approach, which evaluates host-derived extrinsic signals rather than tumour-intrinsic factors for their ability to promote metastatic seeding, is broadly applicable and lays a framework for the screening of environmental constraints to metastasis in other organs and cancer types.


Sujet(s)
Systèmes CRISPR-Cas , Hépatocytes , Tumeurs du foie , Foie , Métastase tumorale , Protéines de tissu nerveux , Animaux , Femelle , Humains , Mâle , Souris , Lignée cellulaire tumorale , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Systèmes CRISPR-Cas/génétique , Modèles animaux de maladie humaine , Éléments transposables d'ADN , Fluorescence , Hépatocytes/métabolisme , Hépatocytes/cytologie , Hépatocytes/anatomopathologie , Facteur-4 de type Kruppel/métabolisme , Foie/cytologie , Foie/métabolisme , Foie/anatomopathologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/prévention et contrôle , Tumeurs du foie/secondaire , Mélanome/métabolisme , Mélanome/anatomopathologie , Métastase tumorale/traitement médicamenteux , Métastase tumorale/anatomopathologie , Métastase tumorale/prévention et contrôle , Protéines de tissu nerveux/antagonistes et inhibiteurs , Protéines de tissu nerveux/métabolisme , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Sémaphorines/antagonistes et inhibiteurs , Sémaphorines/métabolisme
7.
Proc Natl Acad Sci U S A ; 121(31): e2404193121, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39042698

RÉSUMÉ

Hematopoietic stem cells (HSCs) develop from hemogenic endothelial cells (HECs) in vivo during mouse embryogenesis. When cultured in vitro, cells from the embryo phenotypically defined as pre-HSC-I and pre-HSC-II have the potential to differentiate into HSCs. However, minimal factors required for HSC induction from HECs have not yet been determined. In this study, we demonstrated that stem cell factor (SCF) and thrombopoietin (TPO) induced engrafting HSCs from embryonic day (E) 11.5 pre-HSC-I in a serum-free and feeder-free culture condition. In contrast, E10.5 pre-HSC-I and HECs required an endothelial cell layer in addition to SCF and TPO to differentiate into HSCs. A single-cell RNA sequencing analysis of E10.5 to 11.5 dorsal aortae with surrounding tissues and fetal livers detected TPO expression confined in hepatoblasts, while SCF was expressed in various tissues, including endothelial cells and hepatoblasts. Our results suggest a transition of signal requirement during HSC development from HECs. The differentiation of E10.5 HECs to E11.5 pre-HSC-I in the aorta-gonad-mesonephros region depends on SCF and endothelial cell-derived factors. Subsequently, SCF and TPO drive the differentiation of E11.5 pre-HSC-I to pre-HSC-II/HSCs in the fetal liver. The culture system established in this study provides a beneficial tool for exploring the molecular mechanisms underlying the development of HSCs from HECs.


Sujet(s)
Différenciation cellulaire , Hémangioblastes , Cellules souches hématopoïétiques , Facteur de croissance des cellules souches , Thrombopoïétine , Animaux , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/cytologie , Souris , Thrombopoïétine/métabolisme , Facteur de croissance des cellules souches/métabolisme , Hémangioblastes/métabolisme , Hémangioblastes/cytologie , Cellules endothéliales/métabolisme , Cellules endothéliales/cytologie , Transduction du signal , Hématopoïèse/physiologie , Développement embryonnaire , Embryon de mammifère/métabolisme , Embryon de mammifère/cytologie , Foie/embryologie , Foie/métabolisme , Foie/cytologie
9.
Biomaterials ; 311: 122681, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38944968

RÉSUMÉ

Cell-laden bioprinting is a promising biofabrication strategy for regenerating bioactive transplants to address organ donor shortages. However, there has been little success in reproducing transplantable artificial organs with multiple distinctive cell types and physiologically relevant architecture. In this study, an omnidirectional printing embedded network (OPEN) is presented as a support medium for embedded 3D printing. The medium is state-of-the-art due to its one-step preparation, fast removal, and versatile ink compatibility. To test the feasibility of OPEN, exceptional primary mouse hepatocytes (PMHs) and endothelial cell line-C166, were used to print hepatospheroid-encapsulated-artificial livers (HEALs) with vein structures following predesigned anatomy-based printing paths in OPEN. PMHs self-organized into hepatocyte spheroids within the ink matrix, whereas the entire cross-linked structure remained intact for a minimum of ten days of cultivation. Cultivated HEALs maintained mature hepatic functions and marker gene expression at a higher level than conventional 2D and 3D conditions in vitro. HEALs with C166-laden vein structures promoted endogenous neovascularization in vivo compared with hepatospheroid-only liver prints within two weeks of transplantation. Collectively, the proposed platform enables the manufacture of bioactive tissues or organs resembling anatomical architecture, and has broad implications for liver function replacement in clinical applications.


Sujet(s)
Bio-impression , Veines hépatiques , Hépatocytes , Foie , Néovascularisation physiologique , Impression tridimensionnelle , Sphéroïdes de cellules , Animaux , Bio-impression/méthodes , Hépatocytes/cytologie , Souris , Sphéroïdes de cellules/cytologie , Foie/cytologie , Transplantation hépatique , Foie artificiel , Ingénierie tissulaire/méthodes , Structures d'échafaudage tissulaires/composition chimique , Lignée cellulaire , Souris de lignée C57BL , Mâle
10.
Biol Pharm Bull ; 47(7): 1241-1247, 2024.
Article de Anglais | MEDLINE | ID: mdl-38945897

RÉSUMÉ

Primary hepatocytes are valuable for studying liver diseases, drug-induced liver injury, and drug metabolism. However, when cultured in a two-dimensional (2D) environment, primary hepatocytes undergo rapid dedifferentiation via an epithelial-mesenchymal transition (EMT) and lose their liver-specific functions. On the other hand, a three-dimensional (3D) culture of primary hepatocyte organoids presents challenges for analyzing cellular functions and molecular behaviors due to strong cell-cell adhesion among heterogeneous cells. In this study, we developed a novel dispersion culture method of hepatocytes within a dome-shaped collagen matrix, overcoming conventional limitations. The expression levels of EMT-related genes were lower in rat primary hepatocytes cultured using this method for 4 d than in cells cultured using the 2D method. Furthermore, albumin production, a marker of liver function, declined sharply in rat primary hepatocytes cultured in two dimensions from 6.40 µg/mL/48 h on day 4 to 1.35 µg/mL/48 h on day 8, and declined gradually from 4.92 µg/mL/48 h on day 8 to 3.89 µg/mL/48 h on day 14 in rat primary hepatocytes cultured using our new method. These findings indicate that the newly developed culture method can suppress EMT and maintain liver functions for 14 d in rat primary hepatocytes, potentially expanding the utility of primary hepatocyte cultured by using conventional 3D methods.


Sujet(s)
Collagène , Transition épithélio-mésenchymateuse , Hépatocytes , Foie , Animaux , Hépatocytes/métabolisme , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/physiologie , Cellules cultivées , Collagène/métabolisme , Mâle , Foie/métabolisme , Foie/cytologie , Rats , Techniques de culture cellulaire/méthodes , Rat Sprague-Dawley , Albumines/métabolisme
11.
Biofabrication ; 16(3)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38865994

RÉSUMÉ

The lack of adequate humanin vitromodels that recapitulate the cellular composition and response of the human liver to injury hampers the development of anti-fibrotic drugs. The goal of this study was to develop a human spheroid culture model to study liver fibrosis by using induced pluripotent stem cell (iPSC)-derived liver cells. iPSCs were independently differentiated towards hepatoblasts (iHepatoblasts), hepatic stellate cells (iHSCs), endothelial cells (iECs) and macrophages (iMΦ), before assembly into free floating spheroids by culturing cells in 96-well U-bottom plates and orbital shaking for up to 21 days to allow further maturation. Through transcriptome analysis, we show further maturation of iECs and iMΦ, the differentiation of the iHepatoblasts towards hepatocyte-like cells (iHeps) and the inactivation of the iHSCs by the end of the 3D culture. Moreover, these cultures display a similar expression of cell-specific marker genes (CYP3A4, PDGFRß, CD31andCD68) and sensitivity to hepatotoxicity as spheroids made using freshly isolated primary human liver cells. Furthermore, we show the functionality of the iHeps and the iHSCs by mimicking liver fibrosis through iHep-induced iHSC activation, using acetaminophen. In conclusion, we have established a reproducible human iPSC-derived liver culture model that can be used to mimic fibrosisin vitroas a replacement of primary human liver derived 3D models. The model can be used to investigate pathways involved in fibrosis development and to identify new targets for chronic liver disease therapy.


Sujet(s)
Différenciation cellulaire , Techniques de coculture , Cellules souches pluripotentes induites , Cirrhose du foie , Foie , Sphéroïdes de cellules , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Sphéroïdes de cellules/anatomopathologie , Sphéroïdes de cellules/cytologie , Sphéroïdes de cellules/métabolisme , Foie/anatomopathologie , Foie/cytologie , Modèles biologiques , Hépatocytes/cytologie , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/cytologie , Cellules étoilées du foie/anatomopathologie , Cellules cultivées
12.
Biophys J ; 123(13): 1869-1881, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38835167

RÉSUMÉ

Cell mechanics are pivotal in regulating cellular activities, diseases progression, and cancer development. However, the understanding of how cellular viscoelastic properties vary in physiological and pathological stimuli remains scarce. Here, we develop a hybrid self-similar hierarchical theory-microrheology approach to accurately and efficiently characterize cellular viscoelasticity. Focusing on two key cell types associated with livers fibrosis-the capillarized liver sinusoidal endothelial cells and activated hepatic stellate cells-we uncover a universal two-stage power-law rheology characterized by two distinct exponents, αshort and αlong. The mechanical profiles derived from both exponents exhibit significant potential for discriminating among diverse cells. This finding suggests a potential common dynamic creep characteristic across biological systems, extending our earlier observations in soft tissues. Using a tailored hierarchical model for cellular mechanical structures, we discern significant variations in the viscoelastic properties and their distribution profiles across different cell types and states from the cytoplasm (elastic stiffness E1 and viscosity η), to a single cytoskeleton fiber (elastic stiffness E2), and then to the cell level (transverse expansion stiffness E3). Importantly, we construct a logistic-regression-based machine-learning model using the dynamic parameters that outperforms conventional cell-stiffness-based classifiers in assessing cell states, achieving an area under the curve of 97% vs. 78%. Our findings not only advance a robust framework for monitoring intricate cell dynamics but also highlight the crucial role of cellular viscoelasticity in discerning cell states across a spectrum of liver diseases and prognosis, offering new avenues for developing diagnostic and therapeutic strategies based on cellular viscoelasticity.


Sujet(s)
Élasticité , Viscosité , Phénomènes biomécaniques , Animaux , Cellules endothéliales/cytologie , Cellules endothéliales/métabolisme , Cellules étoilées du foie/cytologie , Cellules étoilées du foie/métabolisme , Rhéologie , Humains , Modèles biologiques , Foie/cytologie , Apprentissage machine
13.
Methods Mol Biol ; 2817: 97-113, 2024.
Article de Anglais | MEDLINE | ID: mdl-38907150

RÉSUMÉ

Spatially resolved mass spectrometry-based proteomics at single-cell resolution promises to provide insights into biological heterogeneity. We describe a protocol based on multiplexed data-independent acquisition (mDIA) with dimethyl labeling to enhance proteome depth, accuracy, and throughput while minimizing costs. It enables high-quality proteome analysis of single isolated hepatocytes and utilizes liver zonation for single-cell proteomics benchmarking. This adaptable, modular protocol will promote the use of single-cell proteomics in spatial biology.


Sujet(s)
Hépatocytes , Protéome , Protéomique , Analyse sur cellule unique , Hépatocytes/métabolisme , Hépatocytes/cytologie , Protéomique/méthodes , Analyse sur cellule unique/méthodes , Animaux , Protéome/analyse , Spectrométrie de masse/méthodes , Souris , Foie/métabolisme , Foie/cytologie
14.
Nature ; 630(8016): 412-420, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38839950

RÉSUMÉ

The processes that govern human haematopoietic stem cell (HSC) self-renewal and engraftment are poorly understood and challenging to recapitulate in culture to reliably expand functional HSCs1-3. Here we identify MYC target 1 (MYCT1; also known as MTLC) as a crucial human HSC regulator that moderates endocytosis and environmental sensing in HSCs. MYCT1 is selectively expressed in undifferentiated human haematopoietic stem and progenitor cells (HSPCs) and endothelial cells but becomes markedly downregulated during HSC culture. Lentivirus-mediated knockdown of MYCT1 prevented human fetal liver and cord blood (CB) HSPC expansion and engraftment. By contrast, restoring MYCT1 expression improved the expansion and engraftment of cultured CB HSPCs. Single-cell RNA sequencing of human CB HSPCs in which MYCT1 was knocked down or overexpressed revealed that MYCT1 governs important regulatory programmes and cellular properties essential for HSC stemness, such as ETS factor expression and low mitochondrial activity. MYCT1 is localized in the endosomal membrane in HSPCs and interacts with vesicle trafficking regulators and signalling machinery. MYCT1 loss in HSPCs led to excessive endocytosis and hyperactive signalling responses, whereas restoring MYCT1 expression balanced culture-induced endocytosis and dysregulated signalling. Moreover, sorting cultured CB HSPCs on the basis of lowest endocytosis rate identified HSPCs with preserved MYCT1 expression and MYCT1-regulated HSC stemness programmes. Our work identifies MYCT1-moderated endocytosis and environmental sensing as essential regulatory mechanisms required to preserve human HSC stemness. Our data also pinpoint silencing of MYCT1 as a cell-culture-induced vulnerability that compromises human HSC expansion.


Sujet(s)
Auto-renouvellement cellulaire , Cellules souches hématopoïétiques , Protéines nucléaires , Animaux , Femelle , Humains , Mâle , Souris , Cellules cultivées , Endocytose , Endosomes/métabolisme , Cellules endothéliales/cytologie , Cellules endothéliales/métabolisme , Sang foetal/cytologie , Techniques de knock-down de gènes , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Foie/cytologie , Foie/métabolisme , Foie/embryologie , Mitochondries/métabolisme , Protéines nucléaires/métabolisme , Transduction du signal , Protéines proto-oncogènes c-ets/génétique , Protéines proto-oncogènes c-ets/métabolisme , Analyse de l'expression du gène de la cellule unique
15.
Sci Rep ; 14(1): 13831, 2024 06 15.
Article de Anglais | MEDLINE | ID: mdl-38879647

RÉSUMÉ

Liver sinusoidal endothelial cells (LSECs) are highly specialized endothelial cells (ECs) that play an important role in liver development and regeneration. Additionally, it is involved in various pathological processes, including steatosis, inflammation, fibrosis and hepatocellular carcinoma. However, the rapid dedifferentiation of LSECs after culture greatly limits their use in vitro modeling for biomedical applications. In this study, we developed a highly efficient protocol to induce LSEC-like cells from human induced pluripotent stem cells (hiPSCs) in only 8 days. Using single-cell transcriptomic analysis, we identified several novel LSEC-specific markers, such as EPAS1, LIFR, and NID1, as well as several previously revealed markers, such as CLEC4M, CLEC1B, CRHBP and FCN3. These LSEC markers are specifically expressed in our LSEC-like cells. Furthermore, hiPSC-derived cells expressed LSEC-specific proteins and exhibited LSEC-related functions, such as the uptake of acetylated low density lipoprotein (ac-LDL) and immune complex endocytosis. Overall, this study confirmed that our novel protocol allowed hiPSCs to rapidly acquire an LSEC-like phenotype and function in vitro. The ability to generate LSECs efficiently and rapidly may help to more precisely mimic liver development and disease progression in a liver-specific multicellular microenvironment, offering new insights into the development of novel therapeutic strategies.


Sujet(s)
Différenciation cellulaire , Cellules endothéliales , Cellules souches pluripotentes induites , Foie , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Cellules endothéliales/métabolisme , Cellules endothéliales/cytologie , Foie/métabolisme , Foie/cytologie , Analyse sur cellule unique/méthodes , Cellules cultivées , Marqueurs biologiques/métabolisme , Lipoprotéines LDL/métabolisme , Analyse de profil d'expression de gènes
16.
Stem Cell Rev Rep ; 20(6): 1599-1617, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38769232

RÉSUMÉ

In animal experimental models the administration of stem cells into the spleen should ensure high effectiveness of their implantation in the liver due to a direct vascular connection between the two organs. The aim of this study was to update the methods of experimental intrasplenic cell transplantation using human amniotic epithelial cells (hAECs) which are promising cells in the treatment of liver diseases. BALB/c mice were administered intrasplenically with 0.5, 1, and 2 million hAECs by direct bolus injection (400 µl/min) and via a subcutaneous splenic port by fast (20 µl/min) and slow (10 µl/min) infusion. The port was prepared by translocating the spleen to the skin pocket. The spleen, liver, and lungs were collected at 3 h, 6 h, and 24 h after the administration of cells. The distribution of hAECs, histopathological changes in the organs, complete blood count, and biochemical markers of liver damage were assessed. It has been shown that the method of intrasplenic cell administration affects the degree of liver damage. The largest number of mice showing significant liver damage was observed after direct administration and the lowest after slow administration through a port. Liver damage increased with the number of administered cells, which, paradoxically, resulted in increased liver colonization efficiency. It was concluded that the administration of 1 × 106 hAECs by slow infusion via a subcutaneous splenic port reduces the incidence of complications at the expense of a slight decrease in the effectiveness of implantation of the transplanted cells in the liver.


Sujet(s)
Amnios , Cellules épithéliales , Maladies du foie , Souris de lignée BALB C , Rate , Animaux , Humains , Cellules épithéliales/cytologie , Amnios/cytologie , Maladies du foie/thérapie , Maladies du foie/anatomopathologie , Souris , Rate/cytologie , Femelle , Thérapie cellulaire et tissulaire/méthodes , Foie/cytologie
17.
Nature ; 630(8015): 158-165, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38693268

RÉSUMÉ

The liver has a unique ability to regenerate1,2; however, in the setting of acute liver failure (ALF), this regenerative capacity is often overwhelmed, leaving emergency liver transplantation as the only curative option3-5. Here, to advance understanding of human liver regeneration, we use paired single-nucleus RNA sequencing combined with spatial profiling of healthy and ALF explant human livers to generate a single-cell, pan-lineage atlas of human liver regeneration. We uncover a novel ANXA2+ migratory hepatocyte subpopulation, which emerges during human liver regeneration, and a corollary subpopulation in a mouse model of acetaminophen (APAP)-induced liver regeneration. Interrogation of necrotic wound closure and hepatocyte proliferation across multiple timepoints following APAP-induced liver injury in mice demonstrates that wound closure precedes hepatocyte proliferation. Four-dimensional intravital imaging of APAP-induced mouse liver injury identifies motile hepatocytes at the edge of the necrotic area, enabling collective migration of the hepatocyte sheet to effect wound closure. Depletion of hepatocyte ANXA2 reduces hepatocyte growth factor-induced human and mouse hepatocyte migration in vitro, and abrogates necrotic wound closure following APAP-induced mouse liver injury. Together, our work dissects unanticipated aspects of liver regeneration, demonstrating an uncoupling of wound closure and hepatocyte proliferation and uncovering a novel migratory hepatocyte subpopulation that mediates wound closure following liver injury. Therapies designed to promote rapid reconstitution of normal hepatic microarchitecture and reparation of the gut-liver barrier may advance new areas of therapeutic discovery in regenerative medicine.


Sujet(s)
Défaillance hépatique aigüe , Régénération hépatique , Animaux , Femelle , Humains , Mâle , Souris , Acétaminophène/pharmacologie , Lignage cellulaire , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lésions hépatiques dues aux substances/anatomopathologie , Modèles animaux de maladie humaine , Facteur de croissance des hépatocytes/métabolisme , Facteur de croissance des hépatocytes/pharmacologie , Hépatocytes/cytologie , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Foie/cytologie , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Défaillance hépatique aigüe/anatomopathologie , Défaillance hépatique aigüe/induit chimiquement , Régénération hépatique/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Nécrose/induit chimiquement , Médecine régénérative , Analyse de l'expression du gène de la cellule unique , Cicatrisation de plaie
18.
Nat Commun ; 15(1): 3940, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750036

RÉSUMÉ

Hepatocytes play important roles in the liver, but in culture, they immediately lose function and dedifferentiate into progenitor-like cells. Although this unique feature is well-known, the dynamics and mechanisms of hepatocyte dedifferentiation and the differentiation potential of dedifferentiated hepatocytes (dediHeps) require further investigation. Here, we employ a culture system specifically established for hepatic progenitor cells to study hepatocyte dedifferentiation. We found that hepatocytes dedifferentiate with a hybrid epithelial/mesenchymal phenotype, which is required for the induction and maintenance of dediHeps, and exhibit Vimentin-dependent propagation, upon inhibition of the Hippo signaling pathway. The dediHeps re-differentiate into mature hepatocytes by forming aggregates, enabling reconstitution of hepatic tissues in vivo. Moreover, dediHeps have an unexpected differentiation potential into intestinal epithelial cells that can form organoids in three-dimensional culture and reconstitute colonic epithelia after transplantation. This remarkable plasticity will be useful in the study and treatment of intestinal metaplasia and related diseases in the liver.


Sujet(s)
Dédifférenciation cellulaire , Différenciation cellulaire , Cellules épithéliales , Hépatocytes , Animaux , Hépatocytes/cytologie , Hépatocytes/métabolisme , Cellules épithéliales/cytologie , Cellules épithéliales/métabolisme , Souris , Organoïdes/cytologie , Organoïdes/métabolisme , Transition épithélio-mésenchymateuse , Muqueuse intestinale/cytologie , Muqueuse intestinale/métabolisme , Cellules cultivées , Transduction du signal , Vimentine/métabolisme , Voie de signalisation Hippo , Foie/cytologie , Foie/métabolisme , Souris de lignée C57BL , Mâle , Techniques de culture cellulaire/méthodes
19.
Cell Mol Biol Lett ; 29(1): 67, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38724891

RÉSUMÉ

BACKGROUND: It is generally accepted that endothelial cells (ECs), primarily rely on glycolysis for ATP production, despite having functional mitochondria. However, it is also known that ECs are heterogeneous, and their phenotypic features depend on the vascular bed. Emerging evidence suggests that liver sinusoidal ECs (LSECs), located in the metabolically rich environment of the liver, show high metabolic plasticity. However, the substrate preference for energy metabolism in LSECs remains unclear. METHODS: Investigations were conducted in primary murine LSECs in vitro using the Seahorse XF technique for functional bioenergetic assays, untargeted mass spectrometry-based proteomics to analyse the LSEC proteome involved in energy metabolism pathways, liquid chromatography-tandem mass spectrometry-based analysis of acyl-carnitine species and Raman spectroscopy imaging to track intracellular palmitic acid. RESULTS: This study comprehensively characterized the energy metabolism of LSECs, which were found to depend on oxidative phosphorylation, efficiently fuelled by glucose-derived pyruvate, short- and medium-chain fatty acids and glutamine. Furthermore, despite its high availability, palmitic acid was not directly oxidized in LSEC mitochondria, as evidenced by the acylcarnitine profile and etomoxir's lack of effect on oxygen consumption. However, together with L-carnitine, palmitic acid supported mitochondrial respiration, which is compatible with the chain-shortening role of peroxisomal ß-oxidation of long-chain fatty acids before further degradation and energy generation in mitochondria. CONCLUSIONS: LSECs show a unique bioenergetic profile of highly metabolically plastic ECs adapted to the liver environment. The functional reliance of LSECs on oxidative phosphorylation, which is not a typical feature of ECs, remains to be determined.


Sujet(s)
Cellules endothéliales , Métabolisme énergétique , Acides gras , Foie , Phosphorylation oxydative , Animaux , Foie/métabolisme , Foie/cytologie , Cellules endothéliales/métabolisme , Souris , Acides gras/métabolisme , Mitochondries/métabolisme , Carnitine/métabolisme , Carnitine/analogues et dérivés , Acide palmitique/métabolisme , Souris de lignée C57BL , Mâle , Mitochondries du foie/métabolisme , Cellules cultivées , Oxydoréduction
20.
Int J Mol Sci ; 25(10)2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38791172

RÉSUMÉ

The main focus of in vitro toxicity assessment methods is to assess the viability of the cells, which is usually based on metabolism changes. Yet, when exposed to toxic substances, the cell triggers multiple signals in response. With this in mind, we have developed a promising cell-based toxicity method that observes various cell responses when exposed to toxic substances (either death, division, or remain viable). Based on the collective cell response, we observed and predicted the dynamics of the cell population to determine the toxicity of the toxicant. The method was tested with two different conformations: In the first conformation, we exposed a monoculture model of blood macrophages to UV light, hydrogen peroxide, nutrient deprivation, tetrabromobisphenol A, fatty acids, and 5-fluorouracil. In the second, we exposed a coculture liver model consisting of hepatocytes, hepatic stellate cells, Kupffer cells, and liver sinusoidal endothelial cells to rifampicin, ibuprofen, and 5-fluorouracil. The method showed good accuracy compared to established toxicity assessment methods. In addition, this approach provided more representative information on the toxic effects of the compounds, as it considers the different cellular responses induced by toxic agents.


Sujet(s)
Fluorouracil , Humains , Fluorouracil/pharmacologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Tests de toxicité/méthodes , Peroxyde d'hydrogène/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques , Animaux , Techniques de coculture/méthodes , Rayons ultraviolets , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/cytologie , Ibuprofène/pharmacologie , Cellules cultivées , Rifampicine/pharmacologie , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE