Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 7.686
1.
Sci Rep ; 14(1): 10846, 2024 05 13.
Article En | MEDLINE | ID: mdl-38736008

Human liver organoids are in vitro three dimensionally (3D) cultured cells that have a bipotent stem cell phenotype. Translational research of human liver organoids for drug discovery has been limited by the challenge of their low hepatic function compared to primary human hepatocytes (PHHs). Various attempts have been made to develop functional hepatocyte-like cells from human liver organoids. However, none have achieved the same level of hepatic functions as PHHs. We here attempted to culture human liver organoids established from cryopreserved PHHs (PHH-derived organoids), using HYDROX, a chemically defined 3D nanofiber. While the proliferative capacity of PHH-derived organoids was lost by HYDROX-culture, the gene expression levels of drug-metabolizing enzymes were significantly improved. Enzymatic activities of cytochrome P450 3A4 (CYP3A4), CYP2C19, and CYP1A2 in HYDROX-cultured PHH-derived organoids (Org-HYDROX) were comparable to those in PHHs. When treated with hepatotoxic drugs such as troglitazone, amiodarone and acetaminophen, Org-HYDROX showed similar cell viability to PHHs, suggesting that Org-HYDROX could be applied to drug-induced hepatotoxicity tests. Furthermore, Org-HYDROX maintained its functions for up to 35 days and could be applied to chronic drug-induced hepatotoxicity tests using fialuridine. Our findings demonstrated that HYDROX could possibly be a novel biomaterial for differentiating human liver organoids towards hepatocytes applicable to pharmaceutical research.


Cell Differentiation , Hepatocytes , Nanofibers , Organoids , Humans , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/cytology , Organoids/drug effects , Organoids/metabolism , Organoids/cytology , Cell Differentiation/drug effects , Nanofibers/chemistry , Cells, Cultured , Liver/cytology , Liver/drug effects , Liver/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/metabolism , Cell Survival/drug effects , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A/genetics
2.
Front Immunol ; 15: 1385022, 2024.
Article En | MEDLINE | ID: mdl-38694507

Liver failure represents a critical medical condition with a traditionally grim prognosis, where treatment options have been notably limited. Historically, liver transplantation has stood as the sole definitive cure, yet the stark disparity between the limited availability of liver donations and the high demand for such organs has significantly hampered its feasibility. This discrepancy has necessitated the exploration of hepatocyte transplantation as a temporary, supportive intervention. In light of this, our review delves into the burgeoning field of hepatocyte transplantation, with a focus on the latest advancements in maintaining hepatocyte function, co-microencapsulation techniques, xenogeneic hepatocyte transplantation, and the selection of materials for microencapsulation. Our examination of hepatocyte microencapsulation research highlights that, to date, most studies have been conducted in vitro or using liver failure mouse models, with a notable paucity of experiments on larger mammals. The functionality of microencapsulated hepatocytes is primarily inferred through indirect measures such as urea and albumin production and the rate of ammonia clearance. Furthermore, research on the mechanisms underlying hepatocyte co-microencapsulation remains limited, and the practicality of xenogeneic hepatocyte transplantation requires further validation. The potential of hepatocyte microencapsulation extends beyond the current scope of application, suggesting a promising horizon for liver failure treatment modalities. Innovations in encapsulation materials and techniques aim to enhance cell viability and function, indicating a need for comprehensive studies that bridge the gap between small-scale laboratory success and clinical applicability. Moreover, the integration of bioengineering and regenerative medicine offers novel pathways to refine hepatocyte transplantation, potentially overcoming the challenges of immune rejection and ensuring the long-term functionality of transplanted cells. In conclusion, while hepatocyte microencapsulation and transplantation herald a new era in liver failure therapy, significant strides must be made to translate these experimental approaches into viable clinical solutions. Future research should aim to expand the experimental models to include larger mammals, thereby providing a clearer understanding of the clinical potential of these therapies. Additionally, a deeper exploration into the mechanisms of cell survival and function within microcapsules, alongside the development of innovative encapsulation materials, will be critical in advancing the field and offering new hope to patients with liver failure.


Cell Encapsulation , Cell Survival , Hepatocytes , Animals , Humans , Cell Encapsulation/methods , Hepatocytes/transplantation , Hepatocytes/cytology , Liver Failure/therapy , Transplantation, Heterologous
3.
Nat Commun ; 15(1): 4034, 2024 May 13.
Article En | MEDLINE | ID: mdl-38740814

Mechanisms underlying human hepatocyte growth in development and regeneration are incompletely understood. In vitro, human fetal hepatocytes (FH) can be robustly grown as organoids, while adult primary human hepatocyte (PHH) organoids remain difficult to expand, suggesting different growth requirements between fetal and adult hepatocytes. Here, we characterize hepatocyte organoid outgrowth using temporal transcriptomic and phenotypic approaches. FHs initiate reciprocal transcriptional programs involving increased proliferation and repressed lipid metabolism upon initiation of organoid growth. We exploit these insights to design maturation conditions for FH organoids, resulting in acquisition of mature hepatocyte morphological traits and increased expression of functional markers. During PHH organoid outgrowth in the same culture condition as for FHs, the adult transcriptomes initially mimic the fetal transcriptomic signatures, but PHHs rapidly acquire disbalanced proliferation-lipid metabolism dynamics, resulting in steatosis and halted organoid growth. IL6 supplementation, as emerged from the fetal dataset, and simultaneous activation of the metabolic regulator FXR, prevents steatosis and promotes PHH proliferation, resulting in improved expansion of the derived organoids. Single-cell RNA sequencing analyses reveal preservation of their fetal and adult hepatocyte identities in the respective organoid cultures. Our findings uncover mitogen requirements and metabolic differences determining proliferation of hepatocytes changing from development to adulthood.


Cell Proliferation , Hepatocytes , Lipid Metabolism , Organoids , Transcriptome , Humans , Hepatocytes/metabolism , Hepatocytes/cytology , Organoids/metabolism , Fetus/metabolism , Adult , Interleukin-6/metabolism , Interleukin-6/genetics , Cells, Cultured
4.
J Cell Sci ; 137(10)2024 May 15.
Article En | MEDLINE | ID: mdl-38700490

Hepatocyte organoids (HOs) generated in vitro are powerful tools for liver regeneration. However, previously reported HOs have mostly been fetal in nature with low expression levels of metabolic genes characteristic of adult liver functions, hampering their application in studies of metabolic regulation and therapeutic testing for liver disorders. Here, we report development of novel culture conditions that combine optimized levels of triiodothyronine (T3) with the removal of growth factors to enable successful generation of mature hepatocyte organoids (MHOs) of both mouse and human origin with metabolic functions characteristic of adult livers. We show that the MHOs can be used to study various metabolic functions including bile and urea production, zonal metabolic gene expression, and metabolic alterations in both alcoholic liver disease and non-alcoholic fatty liver disease, as well as hepatocyte proliferation, injury and cell fate changes. Notably, MHOs derived from human fetal hepatocytes also show improved hepatitis B virus infection. Therefore, these MHOs provide a powerful in vitro model for studies of human liver physiology and diseases. The human MHOs are potentially also a robust research tool for therapeutic development.


Hepatocytes , Liver , Organoids , Hepatocytes/metabolism , Hepatocytes/cytology , Organoids/metabolism , Organoids/cytology , Humans , Animals , Mice , Liver/metabolism , Liver/cytology , Mice, Inbred C57BL , Cell Differentiation
5.
Cell Transplant ; 33: 9636897241253700, 2024.
Article En | MEDLINE | ID: mdl-38770981

Hepatocyte transplantation (HCT) is a potential bridging therapy or an alternative to liver transplantation. Conventionally, single-cell hepatocytes are injected via the portal vein. This strategy, however, has yet to overcome poor cell engraftment and function. Therefore, we developed an orthotopic HCT method using a liver-derived extracellular matrix (L-ECM) gel. PXB cells (flesh mature human hepatocytes) were dispersed into the hydrogel solution in vitro, and the gel solution was immediately gelated in 37°C incubators to investigate the affinity between mature human hepatocyte and the L-ECM gel. During the 3-day cultivation in hepatocyte medium, PXB cells formed cell aggregates via cell-cell interactions. Quantitative analysis revealed human albumin production in culture supernatants. For the in vivo assay, PXB cells were encapsulated in the L-ECM gel and transplanted between the liver lobes of normal rats. Pathologically, the L-ECM gel was localized at the transplant site and retained PXB cells. Cell survival and hepatic function marker expression were verified in another rat model wherein thioacetamide was administered to induce liver fibrosis. Moreover, cell-cell interactions and angiogenesis were enhanced in the L-ECM gel compared with that in the collagen gel. Our results indicate that L-ECM gels can help engraft transplanted hepatocytes and express hepatic function as a scaffold for cell transplantation.


Cell Communication , Hepatocytes , Liver Cirrhosis , Hepatocytes/cytology , Hepatocytes/transplantation , Hepatocytes/metabolism , Animals , Humans , Liver Cirrhosis/therapy , Liver Cirrhosis/pathology , Rats , Neovascularization, Physiologic , Extracellular Matrix/metabolism , Male , Liver , Hydrogels/chemistry , Tissue Engineering/methods , Rats, Sprague-Dawley , Cells, Cultured , Angiogenesis
7.
Nat Commun ; 15(1): 3940, 2024 May 15.
Article En | MEDLINE | ID: mdl-38750036

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.


Cell Dedifferentiation , Cell Differentiation , Epithelial Cells , Hepatocytes , Animals , Hepatocytes/cytology , Hepatocytes/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Mice , Organoids/cytology , Organoids/metabolism , Epithelial-Mesenchymal Transition , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Cells, Cultured , Signal Transduction , Vimentin/metabolism , Hippo Signaling Pathway , Liver/cytology , Liver/metabolism , Mice, Inbred C57BL , Male , Cell Culture Techniques/methods
8.
Sci Rep ; 14(1): 12168, 2024 05 28.
Article En | MEDLINE | ID: mdl-38806510

The bioartificial liver (BAL) system can potentially rescue acute liver failure (ALF) patients by providing partial liver function until a suitable donor liver can be found or the native liver has self-regenerated. In this study, we established a suitable cryopreservation process for the development of an off-the-shelf BAL system. The viability of hepatocyte spheroids cryopreserved in liquid nitrogen was comparable to that of fresh primary hepatocyte spheroids. When hepatocyte spheroids were subjected to cryopreservation in a deep freezer, no statistically significant differences were observed in ammonia removal rate or urea secretion rate based on the cryopreservation period. However, the functional activity of the liver post-cryopreservation in a deep freezer was significantly lower than that observed following liquid nitrogen cryopreservation. Moreover, cryopreserving spheroid hydrogel beads in a deep freezer resulted in a significant decrease (approximately 30%) in both ammonia removal and urea secretion rates compared to the group cryopreserved in liquid nitrogen. The viabilities of spheroid hydrogel beads filled into the bioreactor of a BAL system were similar across all four groups. However, upon operating the BAL system for 24 h, the liver function activity was significantly higher in the group comprising hydrogel beads generated after thawing hepatocyte spheroids cryopreserved in liquid nitrogen. Consequently, the manufacturing of beads after the cryopreservation of hepatocyte spheroids is deemed the most suitable method, considering efficiency, economic feasibility, and liver function activity, for producing a BAL system.


Cryopreservation , Hepatocytes , Liver, Artificial , Spheroids, Cellular , Hepatocytes/metabolism , Hepatocytes/cytology , Cryopreservation/methods , Spheroids, Cellular/metabolism , Spheroids, Cellular/cytology , Animals , Cell Survival , Male , Temperature , Rats , Urea/metabolism , Humans , Ammonia/metabolism , Liver Failure, Acute/therapy , Liver Failure, Acute/metabolism , Liver/metabolism , Liver/cytology
9.
Int J Mol Sci ; 25(10)2024 May 12.
Article En | MEDLINE | ID: mdl-38791320

Nuclear speckles are compartments enriched in splicing factors present in the nucleoplasm of eucaryote cells. Speckles have been studied in mammalian culture and tissue cells, as well as in some non-mammalian vertebrate cells and invertebrate oocytes. In mammals, their morphology is linked to the transcriptional and splicing activities of the cell through a recruitment mechanism. In rats, speckle morphology depends on the hormonal cycle. In the present work, we explore whether a similar situation is also present in non-mammalian cells during the reproductive cycle. We studied the speckled pattern in several tissues of a viviparous reptile, the lizard Sceloporus torquatus, during two different stages of reproduction. We used immunofluorescence staining against splicing factors in hepatocytes and oviduct epithelium cells and fluorescence and confocal microscopy, as well as ultrastructural immunolocalization and EDTA contrast in Transmission Electron Microscopy. The distribution of splicing factors in the nucleoplasm of oviductal cells and hepatocytes coincides with the nuclear-speckled pattern described in mammals. Ultrastructurally, those cell types display Interchromatin Granule Clusters and Perichromatin Fibers. In addition, the morphology of speckles varies in oviduct cells at the two stages of the reproductive cycle analyzed, paralleling the phenomenon observed in the rat. The results show that the morphology of speckles in reptile cells depends upon the reproductive stage as it occurs in mammals.


Cell Nucleus , Hepatocytes , Lizards , Animals , Female , Lizards/anatomy & histology , Lizards/physiology , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Hepatocytes/cytology , Viviparity, Nonmammalian/physiology , Oviducts/metabolism , Oviducts/ultrastructure , Oviducts/cytology
10.
Cell Mol Life Sci ; 81(1): 200, 2024 Apr 30.
Article En | MEDLINE | ID: mdl-38684535

BACKGROUND AND AIM: Cellular senescence of hepatocytes involves permanent cell cycle arrest, disrupted cellular bioenergetics, resistance to cell death, and the release of pro-inflammatory cytokines. This 'zombie-like' state perpetuates harmful effects on tissues and holds potential implications for liver disease progression. Remarkably, senescence exhibits heterogeneity, stemming from two crucial factors: the inducing stressor and the cell type. As such, our present study endeavors to characterize stressor-specific changes in senescence phenotype, its related molecular patterns, and cellular bioenergetics in primary mouse hepatocytes (PMH) and hepatocyte-derived liver organoids (HepOrgs). METHODS: PMH, isolated by collagenase-perfused mouse liver (C57B6/J; 18-23 weeks), were cultured overnight in William's E-medium supplemented with 2% FBS, L-glutamine, and hepatocyte growth supplements. HepOrgs were developed by culturing cells in a 3D matrix for two weeks. The senescence was induced by DNA damage (doxorubicin, cisplatin, and etoposide), oxidative stress (H2O2, and ethanol), and telomere inhibition (BIBR-1532), p53 activation (nutlin-3a), DNA methyl transferase inhibition (5-azacitidine), and metabolism inhibitors (galactosamine and hydroxyurea). SA-ß galactosidase activity, immunofluorescence, immunoblotting, and senescence-associated secretory phenotype (SASP), and cellular bioenergetics were used to assess the senescence phenotype. RESULTS: Each senescence inducer triggers a unique combination of senescence markers in hepatocytes. All senescence inducers, except hydroxyurea and ethanol, increased SA-ß galactosidase activity, the most commonly used marker for cellular senescence. Among the SASP factors, CCL2 and IL-10 were consistently upregulated, while Plasminogen activator inhibitor-1 exhibited global downregulation across all modes of senescence. Notably, DNA damage response was activated by DNA damage inducers. Cell cycle markers were most significantly reduced by doxorubicin, cisplatin, and galactosamine. Additionally, DNA damage-induced senescence shifted cellular bioenergetics capacity from glycolysis to oxidative phosphorylation. In HepOrgs exposed to senescence inducers, there was a notable increase in γH2A.X, p53, and p21 levels. Interestingly, while showing a similar trend, SASP gene expression in HepOrgs was significantly higher compared to PMH, demonstrating a several-fold increase. CONCLUSION: In our study, we demonstrated that each senescence inducer activates a unique combination of senescence markers in PMH. Doxorubicin demonstrated the highest efficacy in inducing senescence, followed by cisplatin and H2O2, with no impact on apoptosis. Each inducer prompted DNA damage response and mitochondrial dysfunction, independent of MAPK/AKT.


Cellular Senescence , DNA Damage , Hepatocytes , Mice, Inbred C57BL , Oxidative Stress , Animals , Cellular Senescence/drug effects , Hepatocytes/metabolism , Hepatocytes/drug effects , Hepatocytes/cytology , Mice , Oxidative Stress/drug effects , Cells, Cultured , Senescence-Associated Secretory Phenotype , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Doxorubicin/pharmacology , Energy Metabolism/drug effects , Liver/metabolism , Liver/drug effects , Male
11.
Nat Genet ; 56(5): 953-969, 2024 May.
Article En | MEDLINE | ID: mdl-38627598

The mechanism by which mammalian liver cell responses are coordinated during tissue homeostasis and perturbation is poorly understood, representing a major obstacle in our understanding of many diseases. This knowledge gap is caused by the difficulty involved with studying multiple cell types in different states and locations, particularly when these are transient. We have combined Stereo-seq (spatiotemporal enhanced resolution omics-sequencing) with single-cell transcriptomic profiling of 473,290 cells to generate a high-definition spatiotemporal atlas of mouse liver homeostasis and regeneration at the whole-lobe scale. Our integrative study dissects in detail the molecular gradients controlling liver cell function, systematically defining how gene networks are dynamically modulated through intercellular communication to promote regeneration. Among other important regulators, we identified the transcriptional cofactor TBL1XR1 as a rheostat linking inflammation to Wnt/ß-catenin signaling for facilitating hepatocyte proliferation. Our data and analytical pipelines lay the foundation for future high-definition tissue-scale atlases of organ physiology and malfunction.


Homeostasis , Liver Regeneration , Liver , Wnt Signaling Pathway , Animals , Liver Regeneration/genetics , Mice , Liver/metabolism , Wnt Signaling Pathway/genetics , Hepatocytes/metabolism , Hepatocytes/cytology , Cell Proliferation/genetics , Single-Cell Analysis , Gene Regulatory Networks , Gene Expression Profiling/methods , Transcriptome , Mice, Inbred C57BL , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Male
12.
Sci Bull (Beijing) ; 69(10): 1448-1457, 2024 May 30.
Article En | MEDLINE | ID: mdl-38490890

Liver-tissue engineering has proven valuable in treating liver diseases, but the construction of liver tissues with high fidelity remains challenging. Here, we present a novel three-dimensional (3D)-imprinted cell-sheet strategy for the synchronous construction of biomimetic hepatic microtissues with high accuracy in terms of cell type, density, and distribution. To achieve this, the specific composition of hepatic cells in a normal human liver was determined using a spatial proteogenomics dataset. The data and biomimetic hepatic micro-tissues with hexagonal hollow cross-sections indicate that cell information was successfully generated using a homemade 3D-imprinted device for layer-by-layer imprinting and assembling the hepatic cell sheets. By infiltrating vascular endothelial cells into the hollow section of the assembly, biomimetic hepatic microtissues with vascularized channels for nutrient diffusion and drug perfusion can be obtained. We demonstrate that the resultant vascularized biomimetic hepatic micro-tissues can not only be integrated into a microfluidic drug-screening liver-on-a-chip but also assembled into an enlarged physiological structure to promote liver regeneration. We believe that our 3D-imprinted cell sheets strategy will open new avenues for biomimetic microtissue construction.


Biomimetics , Hepatocytes , Liver , Tissue Engineering , Humans , Liver/cytology , Tissue Engineering/methods , Biomimetics/methods , Hepatocytes/cytology , Hepatocytes/metabolism , Liver Regeneration , Lab-On-A-Chip Devices , Biomimetic Materials/chemistry
13.
Macromol Biosci ; 24(5): e2300411, 2024 May.
Article En | MEDLINE | ID: mdl-38326219

Liver fibrosis occurs in many chronic liver diseases, while severe fibrosis can lead to liver failure. A chitosan-phenol based self-healing hydrogel (CP) integrated with decellularized liver matrix (DLM) is proposed in this study as a 3D gel matrix to carry hepatocytes for possible therapy of liver fibrosis. To mimic the physiological liver microenvironment, DLM is extracted from pigs and mixed with CP hydrogel to generate DLM-CP self-healing hydrogel. Hepatocyte spheroids coated with endothelial cells (ECs) are fabricated using a customized method and embedded in the hydrogel. Hepatocytes injured by exposure to CCl4-containing medium are used as the in vitro toxin-mediated liver fibrosis model, where the EC-covered hepatocyte spheroids embedded in the hydrogel are co-cultured with the injured hepatocytes. The urea synthesis of the injured hepatocytes reaches 91% of the normal level after 7 days of co-culture, indicating that the hepatic function of injured hepatocytes is rescued by the hybrid spheroid-laden DLM-CP hydrogel. Moreover, the relative lactate dehydrogenase activity of the injured hepatocytes is decreased 49% by the hybrid spheroid-laden DLM-CP hydrogel after 7 days of co-culture, suggesting reduced damage in the injured hepatocytes. The combination of hepatocyte/EC hybrid spheroids and DLM-CP hydrogel presents a promising therapeutic strategy for hepatic fibrosis.


Coculture Techniques , Endothelial Cells , Hepatocytes , Hydrogels , Liver , Spheroids, Cellular , Hepatocytes/metabolism , Hepatocytes/cytology , Animals , Spheroids, Cellular/cytology , Hydrogels/chemistry , Hydrogels/pharmacology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Liver/injuries , Liver/pathology , Swine , Decellularized Extracellular Matrix/chemistry , Decellularized Extracellular Matrix/pharmacology , Chitosan/chemistry , Chitosan/pharmacology , Humans , Liver Cirrhosis/pathology , Liver Cirrhosis/therapy , Extracellular Matrix/metabolism , Carbon Tetrachloride
14.
Adv Sci (Weinh) ; 11(17): e2309899, 2024 May.
Article En | MEDLINE | ID: mdl-38380546

The emerging stem cell-derived hepatocyte-like cells (HLCs) are the alternative cell sources of hepatocytes for treatment of highly lethal acute liver failure (ALF). However, the hostile local environment and the immature cell differentiation may compromise their therapeutic efficacy. To this end, human adipose-derived mesenchymal stromal/stem cells (hASCs) are engineered into different-sized multicellular spheroids and co-cultured with 3D coaxially and hexagonally patterned human umbilical vein endothelial cells (HUVECs) in a liver lobule-like manner to enhance their hepatic differentiation efficiency. It is found that small-sized hASC spheroids, with a diameter of ≈50 µm, show superior pro-angiogenic effects and hepatic differentiation compared to the other counterparts. The size-dependent functional enhancements are mediated by the Wnt signaling pathway. Meanwhile, co-culture of hASCs with HUVECs, at a HUVECs/hASCs seeding density ratio of 2:1, distinctly promotes hepatic differentiation and vascularization both in vitro and in vivo, especially when endothelial cells are patterned into hollow hexagons. After subcutaneous implantation, the mini-liver, consisting of HLC spheroids and 3D-printed interconnected vasculatures, can effectively improve liver regeneration in two ALF animal models through amelioration of local oxidative stress and inflammation, reduction of liver necrosis, as well as increase of cell proliferation, thereby showing great promise for clinical translation.


Human Umbilical Vein Endothelial Cells , Mesenchymal Stem Cells , Printing, Three-Dimensional , Spheroids, Cellular , Spheroids, Cellular/cytology , Humans , Animals , Mesenchymal Stem Cells/cytology , Mice , Cell Differentiation/physiology , Tissue Engineering/methods , Liver , Hepatocytes/cytology , Disease Models, Animal , Liver Failure/therapy , Coculture Techniques/methods
15.
Int J Med Sci ; 21(3): 508-518, 2024.
Article En | MEDLINE | ID: mdl-38250613

This study aimed to explore the role of connexin 32 (Cx32) in the directional differentiation of induced pluripotent stem cells (iPSCs) into hepatocytes. Urine-derived epithelial cells were collected from the fresh urine of a healthy donor and transducted with reprogramming plasmid mixture to generate iPSCs. The iPSCs were then directionally differentiated into hepatocytes. During the differentiation, the upregulated and downregulated groups were treated with vitamin K2 (VK2) and 2-aminoethoxyboronate diphenylester (2-APB) to increase and inhibit Cx32 expression, respectively. The control group was not treated with the regulatory factor. Expression of Cx32 and hepatocyte-specific markers, including AFP, hepatocyte nuclear factor 4α (HNF-4α), albumin (ALB) and cytokeratin 18 (CK18) were detected. It indicated that Cx32 expression was not observed in iPSCs, but gradually increased during the process of hepatic differentiation from iPSCs. Upregulation of Cx32 expression by VK2 treatment promoted hepatocyte maturation and enhanced the expression of the aforementioned hepatic specific markers, whereas downregulation of Cx32 expression by 2-APB treatment had the opposite effects. In conclusion, urine-derived iPSCs could be directionally differentiated into hepatocytes. Up-regulation of Cx32 improves the efficiency and maturity of differentiation of iPSCs into hepatocytes, and Cx32 may be a promoting factor during the process of hepatic differentiation from iPSCs.


Cell Differentiation , Gap Junction beta-1 Protein , Hepatocytes , Induced Pluripotent Stem Cells , Down-Regulation , Gap Junction beta-1 Protein/genetics , Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , Vitamin K 2 , Humans
16.
Adv Healthc Mater ; 13(13): e2302943, 2024 May.
Article En | MEDLINE | ID: mdl-38266310

Decellularization of discarded whole livers and their recellularization with patient-specific induced pluripotent stem cells (iPSCs) to develop a functional organ is a promising approach to increasing the donor pool. The effect of extracellular matrix (ECM) of marginal livers on iPSC-hepatocyte differentiation and function has not been shown. To test the effect of donor liver ECM age and steatosis, young and old, as well as no, low, and high steatosis livers, are decellularized. All livers are decellularized successfully. High steatosis livers have fat remaining on the ECM after decellularization. Old donor liver ECM induces lower marker expression in early differentiation stages, compared to young liver ECM, while this difference is closed at later stages and do not affect iPSC-hepatocyte function significantly. High steatosis levels of liver ECM lead to higher albumin mRNA expression and secretion while at later stages of differentiation expression of major cytochrome (CYP) 450 enzymes is highest in low steatosis liver ECM. Both primary human hepatocytes and iPSC-hepatocytes show an increase in fat metabolism marker expression with increasing steatosis levels most likely induced by excess fat remaining on the ECM. Overall, removal of excess fat from liver ECM may be needed for inducing proper hepatic function after recellularization.


Cell Differentiation , Extracellular Matrix , Fatty Liver , Hepatocytes , Induced Pluripotent Stem Cells , Liver , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Fatty Liver/metabolism , Fatty Liver/pathology , Extracellular Matrix/metabolism , Hepatocytes/metabolism , Hepatocytes/cytology , Liver/metabolism , Liver/pathology , Adult , Tissue Donors , Middle Aged , Cells, Cultured , Age Factors , Tissue Scaffolds/chemistry
17.
Prog Mol Biol Transl Sci ; 199: 109-127, 2023.
Article En | MEDLINE | ID: mdl-37678968

Conventional two-dimensional (2-D) cultivation are easy to utilize for human pluripotent stem (hPS) cell cultivation in standard techniques and are important for analysis or development of the signal pathways to keep pluripotent state of hPS cells cultivated on 2-D cell culture materials. However, the most efficient protocol to prepare hPS cells is the cell culture in a three dimensional (3-D) cultivation unit because huge numbers of hPS cells should be utilized in clinical treatment. Some 3-D cultivation strategies for hPS cells are considered: (a) microencapsulated cell cultivation in suspended hydrogels, (b) cell cultivation on microcarriers (MCs), (c) cell cultivation on self-aggregated spheroid [cell aggregates; embryoid bodies (EBs) and organoids], (d) cell cultivation on microfibers or nanofibers, and (e) cell cultivation in macroporous scaffolds. These cultivation ways are described in this chapter.


Cell Culture Techniques, Three Dimensional , Cell Differentiation , Stem Cells , Humans , Stem Cells/cytology , Cell Culture Techniques , Cell Culture Techniques, Three Dimensional/methods , Hepatocytes/cytology , Hydrogels , Cell Encapsulation , Tissue Scaffolds , Tissue Engineering
18.
Life Sci ; 329: 121967, 2023 Sep 15.
Article En | MEDLINE | ID: mdl-37487274

AIMS: Hepatocytes resume proliferation following liver injuries to compensate for the loss of liver mass. Robust liver regeneration is an intrinsic and pivotal process that facilitates restoration of liver anatomy and function. In the present study we investigated the role of ubiquitin-specific peptidase 47 (USP47) in liver regeneration. METHODS AND MATERIALS: Proliferation of hepatocytes was evaluated by Ki67 staining in vivo and EdU incorporation in vitro. DNA-protein interaction was evaluated by chromatin immunoprecipitation (ChIP). RESULTS: USP47 expression was up-regulated in hepatocytes isolated from mice subjected to partial hepatectomy (PHx) or exposed to HGF treatment. Ingenuity pathway analysis revealed E2F1 as a primary regulator of USP47 transcription. Reporter assay and ChIP assay confirmed that E2F1 directly bound to the USP47 promoter and activated USP47 transcription. Consistently, E2F1 knockdown abrogated USP47 induction by HGF. Compared to the wild type littermates, USP47 knockout mice displayed compromised liver regeneration following PHx. In addition, USP47 inhibition by a small-molecule compound impaired liver regeneration in mice. On the contrary, USP47 over-expression enhanced proliferation of hepatocytes in vitro and promoted liver regeneration in mice. Importantly, a positive correlation between USP47 expression and hepatocyte proliferation was identified in patients with acute liver failure (ALF). SIGNIFICANCE: Our data suggest that USP47, transcriptionally activated by E2F1, plays an essential role in liver regeneration.


Hepatocytes , Liver Regeneration , Ubiquitin-Specific Proteases , Male , Animals , Mice , Ubiquitin-Specific Proteases/metabolism , Hepatocytes/cytology , Cell Proliferation , Mice, Knockout , E2F1 Transcription Factor/metabolism , Humans
19.
Nature ; 611(7936): 563-569, 2022 Nov.
Article En | MEDLINE | ID: mdl-36352220

Malaria infection involves an obligatory, yet clinically silent liver stage1,2. Hepatocytes operate in repeating units termed lobules, exhibiting heterogeneous gene expression patterns along the lobule axis3, but the effects of hepatocyte zonation on parasite development at the molecular level remain unknown. Here we combine single-cell RNA sequencing4 and single-molecule transcript imaging5 to characterize the host and parasite temporal expression programmes in a zonally controlled manner for the rodent malaria parasite Plasmodium berghei ANKA. We identify differences in parasite gene expression in distinct zones, including potentially co-adaptive programmes related to iron and fatty acid metabolism. We find that parasites develop more rapidly in the pericentral lobule zones and identify a subpopulation of periportally biased hepatocytes that harbour abortive infections, reduced levels of Plasmodium transcripts and parasitophorous vacuole breakdown. These 'abortive hepatocytes', which appear predominantly with high parasite inoculum, upregulate immune recruitment and key signalling programmes. Our study provides a resource for understanding the liver stage of Plasmodium infection at high spatial resolution and highlights the heterogeneous behaviour of both the parasite and the host hepatocyte.


Gene Expression Regulation , Hepatocytes , Liver , Malaria , Parasites , Plasmodium berghei , Single-Cell Analysis , Animals , Hepatocytes/cytology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/parasitology , Liver/anatomy & histology , Liver/cytology , Liver/immunology , Liver/parasitology , Malaria/genetics , Malaria/immunology , Malaria/parasitology , Parasites/genetics , Parasites/immunology , Parasites/metabolism , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Plasmodium berghei/metabolism , Single Molecule Imaging , Sequence Analysis, RNA , Iron/metabolism , Fatty Acids/metabolism , Transcription, Genetic , Genes, Protozoan/genetics , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology
20.
Proc Natl Acad Sci U S A ; 119(30): e2203849119, 2022 07 26.
Article En | MEDLINE | ID: mdl-35867815

Cell proliferation is tightly controlled by inhibitors that block cell cycle progression until growth signals relieve this inhibition, allowing cells to divide. In several tissues, including the liver, cell proliferation is inhibited at mitosis by the transcriptional repressors E2F7 and E2F8, leading to formation of polyploid cells. Whether growth factors promote mitosis and cell cycle progression by relieving the E2F7/E2F8-mediated inhibition is unknown. We report here on a mechanism of cell division control in the postnatal liver, in which Wnt/ß-catenin signaling maintains active hepatocyte cell division through Tbx3, a Wnt target gene. The TBX3 protein directly represses transcription of E2f7 and E2f8, thereby promoting mitosis. This cascade of sequential transcriptional repressors, initiated by Wnt signals, provides a paradigm for exploring how commonly active developmental signals impact cell cycle completion.


Hepatocytes , Mitosis , Repressor Proteins , Wnt Signaling Pathway , Animals , Hepatocytes/cytology , Hepatocytes/metabolism , Mice , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Box Domain Proteins/metabolism , beta Catenin/metabolism
...