Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31.646
Filter
1.
Sci Rep ; 14(1): 10846, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38736008

ABSTRACT

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.


Subject(s)
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.
Cell Mol Biol Lett ; 29(1): 67, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724891

ABSTRACT

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.


Subject(s)
Endothelial Cells , Energy Metabolism , Fatty Acids , Liver , Oxidative Phosphorylation , Animals , Liver/metabolism , Liver/cytology , Endothelial Cells/metabolism , Mice , Fatty Acids/metabolism , Mitochondria/metabolism , Carnitine/metabolism , Carnitine/analogs & derivatives , Palmitic Acid/metabolism , Mice, Inbred C57BL , Male , Mitochondria, Liver/metabolism , Cells, Cultured , Oxidation-Reduction
3.
Biomaterials ; 310: 122621, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38815455

ABSTRACT

In vitro models of the human liver are promising alternatives to animal tests for drug development. Currently, primary human hepatocytes (PHHs) are preferred for pharmacokinetic and cytotoxicity tests. However, they are unable to recapitulate the flow of bile in hepatobiliary clearance owing to the lack of bile ducts, leading to the limitation of bile analysis. To address the issue, a liver organoid culture system that has a functional bile duct network is desired. In this study, we aimed to generate human iPSC-derived hepatobiliary organoids (hHBOs) consisting of hepatocytes and bile ducts. The two-step differentiation process under 2D and semi-3D culture conditions promoted the maturation of hHBOs on culture plates, in which hepatocyte clusters were covered with monolayered biliary tubes. We demonstrated that the hHBOs reproduced the flow of bile containing a fluorescent bile acid analog or medicinal drugs from hepatocytes into bile ducts via bile canaliculi. Furthermore, the hHBOs exhibited pathophysiological responses to troglitazone, such as cholestasis and cytotoxicity. Because the hHBOs can recapitulate the function of bile ducts in hepatobiliary clearance, they are suitable as a liver disease model and would be a novel in vitro platform system for pharmaceutical research use.


Subject(s)
Bile Ducts , Hepatocytes , Induced Pluripotent Stem Cells , Organoids , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Organoids/drug effects , Organoids/cytology , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/cytology , Cell Differentiation/drug effects , Pharmaceutical Research/methods
4.
Nat Commun ; 15(1): 3940, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750036

ABSTRACT

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.


Subject(s)
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
5.
Int J Mol Sci ; 25(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38791172

ABSTRACT

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.


Subject(s)
Fluorouracil , Humans , Fluorouracil/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Toxicity Tests/methods , Hydrogen Peroxide/pharmacology , Cell Survival/drug effects , Animals , Coculture Techniques/methods , Ultraviolet Rays , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Liver/drug effects , Liver/metabolism , Liver/cytology , Ibuprofen/pharmacology , Cells, Cultured , Rifampin/pharmacology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/drug effects
6.
J Cell Sci ; 137(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38700490

ABSTRACT

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.


Subject(s)
Hepatocytes , Liver , Organoids , Hepatocytes/metabolism , Hepatocytes/cytology , Organoids/metabolism , Organoids/cytology , Humans , Animals , Mice , Liver/metabolism , Liver/cytology , Mice, Inbred C57BL , Cell Differentiation
7.
Biofabrication ; 16(3)2024 May 28.
Article in English | MEDLINE | ID: mdl-38749417

ABSTRACT

Accurate simulation of different cell type interactions is crucial for physiological and precisein vitrodrug testing. Human tissue-resident macrophages are critical for modulating disease conditions and drug-induced injuries in various tissues; however, their limited availability has hindered their use inin vitromodeling. Therefore, this study aimed to create macrophage-containing organoid co-culture models by directly incorporating human-induced pluripotent stem cell (hiPSC)-derived pre-macrophages into organoid and scaffold cell models. The fully differentiated cells in these organoids exhibited functional characteristics of tissue-resident macrophages with enriched pan-macrophage markers and the potential for M1/M2 subtype specialization upon cytokine stimulation. In a hepatic organoid model, the integrated macrophages replicated typical intrinsic properties, including cytokine release, polarization, and phagocytosis, and the co-culture model was more responsive to drug-induced liver injury than a macrophage-free model. Furthermore, alveolar organoid models containing these hiPSC-derived macrophages also showed increased drug and chemical sensitivity to pulmonary toxicants. Moreover, 3D adipocyte scaffold models incorporating macrophages effectively simulated in vivo insulin resistance observed in adipose tissue and showed improved insulin sensitivity on exposure to anti-diabetic drugs. Overall, the findings demonstrated that incorporating hiPSC-derived macrophages into organoid culture models resulted in more physiological and sensitivein vitrodrug evaluation and screening systems.


Subject(s)
Coculture Techniques , Induced Pluripotent Stem Cells , Macrophages , Organoids , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Humans , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Cell Differentiation/drug effects , Liver/cytology , Liver/drug effects , Models, Biological , Animals
8.
FEBS Open Bio ; 14(6): 922-941, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38710666

ABSTRACT

The regenerative capability of the liver is remarkable, but further research is required to understand the role that neutrophils play in this process. In the present study, we reanalyzed single-cell RNA sequencing data from a mouse partial hepatectomy (PH) model to track the transcriptional changes in hepatocytes and non-parenchymal cells. Notably, we unraveled the regenerative capacity of hepatocytes at diverse temporal points after PH, unveiling the contributions of three distinct zones in the liver regeneration process. In addition, we observed that the depletion of neutrophils reduced the survival and liver volume after PH, confirming the important role of neutrophils in liver regeneration. CellChat analysis revealed an intricate crosstalk between neutrophils and macrophages promoting liver regeneration and, using weighted gene correlation network analysis, we identified the most significant genetic module associated with liver regeneration. Our study found that hepatocytes in the periportal zone of the liver are more active than in other zones, suggesting that the crosstalk between neutrophils and macrophages might be a potential target for liver regeneration treatment.


Subject(s)
Hepatectomy , Hepatocytes , Liver Regeneration , Macrophages , Neutrophils , Liver Regeneration/genetics , Animals , Neutrophils/metabolism , Mice , Macrophages/metabolism , Hepatocytes/metabolism , Liver/metabolism , Liver/cytology , Mice, Inbred C57BL , Male
9.
Nature ; 630(8015): 158-165, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38693268

ABSTRACT

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.


Subject(s)
Acetaminophen , Cell Movement , Cell Proliferation , Hepatocytes , Liver Failure, Acute , Liver Regeneration , Liver , Liver Regeneration/physiology , Humans , Animals , Acetaminophen/pharmacology , Mice , Hepatocytes/cytology , Male , Liver Failure, Acute/pathology , Liver Failure, Acute/chemically induced , Liver/cytology , Necrosis , Disease Models, Animal , Wound Healing , Female , Single-Cell Analysis , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology , Chemical and Drug Induced Liver Injury/pathology , Cell Lineage , Mice, Inbred C57BL
10.
Sci Rep ; 14(1): 12168, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806510

ABSTRACT

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.


Subject(s)
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
11.
Nature ; 630(8015): 166-173, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38778114

ABSTRACT

For many adult human organs, tissue regeneration during chronic disease remains a controversial subject. Regenerative processes are easily observed in animal models, and their underlying mechanisms are becoming well characterized1-4, but technical challenges and ethical aspects are limiting the validation of these results in humans. We decided to address this difficulty with respect to the liver. This organ displays the remarkable ability to regenerate after acute injury, although liver regeneration in the context of recurring injury remains to be fully demonstrated. Here we performed single-nucleus RNA sequencing (snRNA-seq) on 47 liver biopsies from patients with different stages of metabolic dysfunction-associated steatotic liver disease to establish a cellular map of the liver during disease progression. We then combined these single-cell-level data with advanced 3D imaging to reveal profound changes in the liver architecture. Hepatocytes lose their zonation and considerable reorganization of the biliary tree takes place. More importantly, our study uncovers transdifferentiation events that occur between hepatocytes and cholangiocytes without the presence of adult stem cells or developmental progenitor activation. Detailed analyses and functional validations using cholangiocyte organoids confirm the importance of the PI3K-AKT-mTOR pathway in this process, thereby connecting this acquisition of plasticity to insulin signalling. Together, our data indicate that chronic injury creates an environment that induces cellular plasticity in human organs, and understanding the underlying mechanisms of this process could open new therapeutic avenues in the management of chronic diseases.


Subject(s)
Cell Transdifferentiation , Hepatocytes , Liver , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Single-Cell Analysis , TOR Serine-Threonine Kinases , Humans , Hepatocytes/metabolism , Hepatocytes/cytology , Hepatocytes/pathology , Liver/pathology , Liver/metabolism , Liver/cytology , Chronic Disease , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Organoids/metabolism , Organoids/pathology , Cell Plasticity , Liver Regeneration , Insulin/metabolism , Male , Liver Diseases/pathology , Liver Diseases/metabolism , RNA-Seq , Female , Disease Progression , Biopsy , Biliary Tract/cytology , Biliary Tract/metabolism , Biliary Tract/pathology , Epithelial Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/pathology
12.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(5): 435-443, 2024 May 09.
Article in Chinese | MEDLINE | ID: mdl-38636997

ABSTRACT

Objective: To explore the biological process of liver tissue-derived extracellular vesicle (LT-EV) in promoting osteogenic differentiation of mesenchymal stem cells and healing of jaw defects to provide a feasible treatment method for the clinical treatment of jaw bone defects. Methods: Enzymatic hydrolysis and differential centrifugation were used to extract LT-EV, scanning electron microscopy, Western blotting, and nanoparticle tracking analyzers were used to identify and characterize LT-EV, and further to explore the biological functions of LT-EV through proteomics and Kyoto Encyclopedia of Genes and Genomes. Flow cytometry was used to detect LT-EV plasma concentration and to calculate the plasma half-life of LT-EV. Small animal in vivo imaging system was used to detect the biological distribution of LT-EV 24 hours after injection. Six C57BL/6 mice were divided into control group and LT-EV group (3 mice in each group) by simple random sampling method. All mice underwent jaw bone defect surgery and tail vein injection every 7 days (the control group was injected with phosphoric buffer saline, LT-EV group was injected with LT-EV), micro-CT was used to evaluate the degree of mouse jaw bone healing 28 days after surgery, HE staining was used to analyze the multi-organ biosafety of LT-EV, and immunofluorescence staining was used to detect the jaw bone expression of osteogenic marker proteins in the defect area. Human jaw bone mesenchymal stem cells (hJBMSC) induced by osteogenic differentiation were treated with LT-EV (obtained from orthognathic surgery patients provided by the Department of Traumatology and Orthognathic Surgery of School of Stomatology of The Fourth Military Medical University resected normal jaw bone fragments), and the difference in osteogenic differentiation ability between the hJBMSC group and the control group (phosphate buffer saline treatment) was compared, and the in vitro bone differentiation promoting effect of LT-EV was verified through alkaline phosphatase (ALP) staining and real-time fluorescence quantitative PCR. Results: The yield of LT-EV was high, and proteomics and Kyoto Encyclopedia of Genes and Genomes showed that LT-EV contained a series of proteins that regulated cell biological functions. LT-EV injected into the tail vein could reach the mouse jaw bone defect area and promote the regeneration and repair of the jaw bone defect [the bone volume fractions of the LT-EV group and the control group were (36.06±4.20)% and (18.58±5.61)%, respectively; t=4.32, P=0.013], and had good biosafety. LT-EV could promote osteogenic differentiation of hJBMSC in vitro. Compared to the control group, ALP staining and osteogenic gene expression levels were significantly enhanced after osteogenic differentiation of hJBMSC (P<0.05). Conclusions: LT-EV exhibits a high yield, ease of acquisition, high biological safety, and excellent bone-promoting effects. It holds promise as a novel cell-free therapy strategy for regenerating craniofacial bone defects.


Subject(s)
Cell Differentiation , Extracellular Vesicles , Liver , Mesenchymal Stem Cells , Mice, Inbred C57BL , Osteogenesis , Animals , Mesenchymal Stem Cells/cytology , Extracellular Vesicles/metabolism , Mice , Liver/cytology , Jaw/cytology , Bone Regeneration
13.
Lab Chip ; 24(10): 2747-2761, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38660778

ABSTRACT

Human liver organoids (HLOs) hold significant potential for recapitulating the architecture and function of liver tissues in vivo. However, conventional culture methods of HLOs, forming Matrigel domes in 6-/24-well plates, have technical limitations such as high cost and low throughput in organoid-based assays for predictive assessment of compounds in clinical and pharmacological lab settings. To address these issues, we have developed a unique microarray 3D bioprinting protocol of progenitor cells in biomimetic hydrogels on a pillar plate with sidewalls and slits, coupled with a clear bottom, 384-deep well plate for scale-up production of HLOs. Microarray 3D bioprinting, a droplet-based printing technology, was used to generate a large number of small organoids on the pillar plate for predictive hepatotoxicity assays. Foregut cells, differentiated from human iPSCs, were mixed with Matrigel and then printed on the pillar plate rapidly and uniformly, resulting in coefficient of variation (CV) values in the range of 15-18%, without any detrimental effect on cell viability. Despite utilizing 10-50-fold smaller cell culture volume compared to their counterparts in Matrigel domes in 6-/24-well plates, HLOs differentiated on the pillar plate exhibited similar morphology and superior function, potentially due to rapid diffusion of nutrients and oxygen at the small scale. Day 25 HLOs were robust and functional on the pillar plate in terms of their viability, albumin secretion, CYP3A4 activity, and drug toxicity testing, all with low CV values. From three independent trials of in situ assessment, the IC50 values calculated for sorafenib and tamoxifen were 6.2 ± 1.6 µM and 25.4 ± 8.3 µM, respectively. Therefore, our unique 3D bioprinting and miniature organoid culture on the pillar plate could be used for scale-up, reproducible generation of HLOs with minimal manual intervention for high-throughput assessment of compound hepatotoxicity.


Subject(s)
Bioprinting , Liver , Organoids , Humans , Organoids/cytology , Organoids/metabolism , Bioprinting/instrumentation , Liver/cytology , Printing, Three-Dimensional , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Hydrogels/chemistry , Cell Survival/drug effects
14.
Dev Comp Immunol ; 156: 105178, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38599553

ABSTRACT

In the present study, using transgenic frogs that express GFP specifically in myeloid cells under the myeloperoxidase enhancer sequence, we found that myeloperoxidase-positive cells are localized in the liver cortex at the late tadpole stages. Immunohistochemical analysis revealed that myelopoiesis in the liver cortex became evident after st. 50 and reached its peak by st. 56. Transplantation experiments indicated that cells with a high density at the liver cortex were derived from the dorso-lateral plate tissue in the neurula embryo. Analysis of smear samples of the cells isolated from collagenase-treated liver tissues of the transgenic tadpoles indicated that myeloid cells were the major population of blood cells in the larval liver and that, in addition to myeloid colonies, erythroid colonies expanded in entire liver after metamorphosis. Cells that were purified from the livers of transgenic tadpoles according to the GFP expression exhibited the multi-lobed nuclei. The results of present study provide evidence that the liver cortex of the Xenopus tadpole is a major site of granulopoiesis.


Subject(s)
Animals, Genetically Modified , Larva , Liver , Myeloid Cells , Xenopus laevis , Animals , Liver/cytology , Myelopoiesis , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Peroxidase/metabolism , Metamorphosis, Biological
15.
Redox Biol ; 72: 103162, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38669864

ABSTRACT

Protein disulfide isomerases (PDIs) are involved in many intracellular and extracellular processes, including cell adhesion and cytoskeletal reorganisation, but their contribution to the regulation of fenestrations in liver sinusoidal endothelial cells (LSECs) remains unknown. Given that fenestrations are supported on a cytoskeleton scaffold, this study aimed to investigate whether endothelial PDIs regulate fenestration dynamics in primary mouse LSECs. PDIA3 and PDIA1 were found to be the most abundant among PDI isoforms in LSECs. Taking advantage of atomic force microscopy, the effects of PDIA1 or PDIA3 inhibition on the fenestrations in LSECs were investigated using a classic PDIA1 inhibitor (bepristat) and novel aromatic N-sulfonamides of aziridine-2-carboxylic acid derivatives as PDIA1 (C-3389) or PDIA3 (C-3399) inhibitors. The effect of PDIA1 inhibition on liver perfusion was studied in vivo using dynamic contrast-enhanced magnetic resonance imaging. Additionally, PDIA1 inhibitors were examined in vitro in LSECs for effects on adhesion, cytoskeleton organisation, bioenergetics, and viability. Inhibition of PDIA1 with bepristat or C-3389 significantly reduced the number of fenestrations in LSECs, while inhibition of PDIA3 with C-3399 had no effect. Moreover, the blocking of free thiols by the cell-penetrating N-ethylmaleimide, but not by the non-cell-penetrating 4-chloromercuribenzenesulfonate, resulted in LSEC defenestration. Inhibition of PDIA1 did not affect LSEC adhesion, viability, and bioenergetics, nor did it induce a clear-cut rearrangement of the cytoskeleton. However, PDIA1-dependent defenestration was reversed by cytochalasin B, a known fenestration stimulator, pointing to the preserved ability of LSECs to form new pores. Importantly, systemic inhibition of PDIA1 in vivo affected intra-parenchymal uptake of contrast agent in mice consistent with LSEC defenestration. These results revealed the role of intracellular PDIA1 in the regulation of fenestration dynamics in LSECs, and in maintaining hepatic sinusoid homeostasis.


Subject(s)
Endothelial Cells , Liver , Protein Disulfide-Isomerases , Animals , Male , Mice , Cell Adhesion , Cells, Cultured , Cytoskeleton/metabolism , Endothelial Cells/metabolism , Endothelial Cells/cytology , Enzyme Inhibitors/pharmacology , Liver/metabolism , Liver/cytology , Protein Disulfide-Isomerases/metabolism , Protein Disulfide-Isomerases/antagonists & inhibitors
16.
Immunol Cell Biol ; 102(5): 381-395, 2024.
Article in English | MEDLINE | ID: mdl-38629182

ABSTRACT

Resident macrophages of various mammalian organs are characterized by several distinctive features in their gene expression profile and phenotype, including involvement in the regulation of organ functions, as well as reduced sensitivity to proinflammatory activation factors. The reasons for the formation of such a specific phenotype remain the subject of intensive research. Some papers emphasize the role of the origin of organ macrophages. Other studies indicate that monocytes that develop in the red bone marrow are also able to form resident macrophages with a phenotype characteristic of a particular organ, but this requires appropriate microenvironmental conditions. In this article, we studied the possibility of differentiation of monocyte-derived macrophages into cells with a Kupffer-like phenotype under the influence of the main stromal components of Kupffer cells macrophage niche: Ito cells, liver sinusoid endotheliocytes and hepatocyte growth factor (HGF). It was found that Kupffer cells are characterized by several features, including increased expression of transcription factors Spic and Id3, as well as MUP family genes, Clusterin and Ngp genes. In addition, Kupffer cells were characterized by a higher proliferative activity. The expression of marker genes of Kupffer cells (i.e. Id3, Spic, Marco and Timd4) increased in monocyte-derived macrophages during coculture with Ito cells, liver sinusoid endothelial cells, macrophage colony-stimulating factor and HGF cells only by 3 days. However, the expression level of these genes was always higher in Kupffer cells. In addition, a complete coincidence of the expressed gene profile in monocyte-derived macrophages and Kupffer cells did not occur even after 3 days of culturing.


Subject(s)
Cell Differentiation , Cellular Microenvironment , Kupffer Cells , Macrophages , Phenotype , Kupffer Cells/metabolism , Kupffer Cells/cytology , Macrophages/metabolism , Animals , Monocytes/metabolism , Monocytes/cytology , Hepatocyte Growth Factor/metabolism , Endothelial Cells/metabolism , Coculture Techniques , Humans , Cell Proliferation , Cells, Cultured , Liver/cytology , Liver/metabolism , Mice
17.
Liver Int ; 44(6): 1290-1297, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38451053

ABSTRACT

Since organoids were developed 15 years ago, they are now in their adolescence as a research tool. The ability to generate 'tissue in a dish' has created enormous opportunities for biomedical research. We examine the contributions that hepatic organoids have made to three areas of liver research: as a source of cells and tissue for basic research, for drug discovery and drug safety testing, and for understanding disease pathobiology. We discuss the features that enable hepatic organoids to provide useful models for human liver diseases and identify four types of advances that will enable them to become a mature (i.e., adult) research tool over the next 5 years. During this period, advances in single-cell RNA sequencing and CRISPR technologies coupled with improved hepatic organoid methodology, which enables them to have a wider range of cell types that are present in liver and to be grown in microwells, will generate discoveries that will dramatically advance our understanding of liver development and the pathogenesis of liver diseases. It will generate also new approaches for treating liver fibrosis, which remains a major public health problem with few treatment options.


Subject(s)
Liver Diseases , Liver , Organoids , Humans , Liver/cytology , Liver/pathology , Liver Diseases/pathology , Liver Diseases/therapy , Drug Discovery , Biomedical Research , Single-Cell Analysis
18.
J Pediatr Gastroenterol Nutr ; 78(5): 1047-1058, 2024 May.
Article in English | MEDLINE | ID: mdl-38529852

ABSTRACT

OBJECTIVES: Parenteral nutrition (PN) is used for patients of varying ages with intestinal failure to supplement calories. Premature newborns with low birth weight are at a high risk for developing PN associated liver disease (PNALD) including steatosis, cholestasis, and gallbladder sludge/stones. To optimize nutrition regimens, models are required to predict PNALD. METHODS: We have exploited induced pluripotent stem cell derived liver organoids to provide a testing platform for PNALD. Liver organoids mimic the developing liver and contain the different hepatic cell types. The organoids have an early postnatal maturity making them a suitable model for premature newborns. To mimic PN treatment we used medium supplemented with either clinoleic (80% olive oil/20% soybean oil) or intralipid (100% soybean oil) for 7 days. RESULTS: Homogenous HNF4a staining was found in all organoids and PN treatments caused accumulation of lipids in hepatocytes. Organoids exhibited a dose dependent decrease in CYP3A4 activity and expression of hepatocyte functional genes. The lipid emulsions did not affect overall organoid viability and glucose levels had no contributory effect to the observed results. CONCLUSIONS: Liver organoids could be utilized as a potential screening platform for the development of new, less hepatotoxic PN solutions. Both lipid treatments caused hepatic lipid accumulation, a significant decrease in CYP3A4 activity and a decrease in the RNA levels of both CYP3A4 and CYP1A2 in a dose dependent manner. The presence of high glucose had no additive effect, while Clinoleic at high dose, caused significant upregulation of interleukin 6 and TLR4 expression.


Subject(s)
Cytochrome P-450 CYP3A , Induced Pluripotent Stem Cells , Liver , Organoids , Parenteral Nutrition , Soybean Oil , Organoids/drug effects , Organoids/metabolism , Cytochrome P-450 CYP3A/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Liver/drug effects , Liver/cytology , Soybean Oil/pharmacology , Phospholipids/pharmacology , Phospholipids/metabolism , Emulsions , Fat Emulsions, Intravenous/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Olive Oil/pharmacology , Infant, Newborn , Hepatocyte Nuclear Factor 4/metabolism , Hepatocyte Nuclear Factor 4/genetics
19.
Sci Bull (Beijing) ; 69(10): 1448-1457, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38490890

ABSTRACT

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.


Subject(s)
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
20.
J Biol Chem ; 300(3): 105732, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38336290

ABSTRACT

The manganese (Mn) export protein SLC30A10 is essential for Mn excretion via the liver and intestines. Patients with SLC30A10 deficiency develop Mn excess, dystonia, liver disease, and polycythemia. Recent genome-wide association studies revealed a link between the SLC30A10 variant T95I and markers of liver disease. The in vivo relevance of this variant has yet to be investigated. Using in vitro and in vivo models, we explore the impact of the T95I variant on SLC30A10 function. While SLC30A10 I95 expressed at lower levels than T95 in transfected cell lines, both T95 and I95 variants protected cells similarly from Mn-induced toxicity. Adeno-associated virus 8-mediated expression of T95 or I95 SLC30A10 using the liver-specific thyroxine binding globulin promoter normalized liver Mn levels in mice with hepatocyte Slc30a10 deficiency. Furthermore, Adeno-associated virus-mediated expression of T95 or I95 SLC30A10 normalized red blood cell parameters and body weights and attenuated Mn levels and differential gene expression in livers and brains of mice with whole body Slc30a10 deficiency. While our in vivo data do not indicate that the T95I variant significantly compromises SLC30A10 function, it does reinforce the notion that the liver is a key site of SLC30A10 function. It also supports the idea that restoration of hepatic SLC30A10 expression is sufficient to attenuate phenotypes in SLC30A10 deficiency.


Subject(s)
Amino Acid Substitution , Cation Transport Proteins , Dependovirus , Liver , Manganese , Mutation , Animals , Mice , Body Weight , Brain/metabolism , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line , Dependovirus/genetics , Erythrocytes , Genome-Wide Association Study , Hepatocytes/metabolism , Liver/cytology , Liver/metabolism , Liver Diseases/genetics , Liver Diseases/metabolism , Manganese/metabolism , Manganese Poisoning/metabolism , Phenotype , Promoter Regions, Genetic , Thyroxine-Binding Globulin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...