Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int J Mol Sci ; 21(1)2019 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-31887985

RESUMO

In this study, we reveal that liver organoid transplantation through the portal vein is a safe and effective method for the treatment of chronic liver damage. The liver organoids significantly reconstituted the hepatocytes; hence, the liver was significantly enlarged in this group, compared to the monolayer cell transplantation group in the retrorsine/partial hepatectomy (RS/PH) model. In the liver organoid transplantation group, the bile ducts were located in the donor area and connected to the recipient bile ducts. Thus, the rate of bile reconstruction in the liver was significantly higher compared to that in the monolayer group. By transplanting liver organoids, we saw a level of 70% replacement of the damaged liver. Consequently, in the transplantation group, diminished ductular reaction and a decrease of placental glutathione S-transferase (GST-p) precancerous lesions were observed. After trans-portal injection, the human induced pluripotent stem cell (hiPSC)-derived liver organoids revealed no translocation outside the liver; in contrast, the monolayer cells had spread to the lungs. The hiPSC-derived liver organoids were attached to the liver in the immunodeficient RS/PH rats. This study clearly demonstrates that liver organoid transplantation through the portal vein is a safe and effective method for the treatment of chronic liver damage in rats.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/terapia , Transplante de Fígado/métodos , Organoides/citologia , Veia Porta/cirurgia , Alcaloides de Pirrolizidina/efeitos adversos , Animais , Células Cultivadas , Feminino , Glutationa Transferase/metabolismo , Hepatectomia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Regeneração Hepática , Técnicas de Cultura de Órgãos , Ratos , Resultado do Tratamento
2.
Sci Transl Med ; 16(757): eadg0338, 2024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39047116

RESUMO

Donor organ shortages for transplantation remain a serious global concern, and alternative treatment is in high demand. Fetal cells and tissues have considerable therapeutic potential as, for example, organoid technology that uses human induced pluripotent stem cells (hiPSCs) to generate unlimited human fetal-like cells and tissues. We previously reported the in vivo vascularization of early fetal liver-like hiPSC-derived liver buds (LBs) and subsquent improved survival of recipient mice with subacute liver failure. Here, we show hiPSC-liver organoids (LOs) that recapitulate midgestational fetal liver promote de novo liver generation when grafted onto the surface of host livers in chemical fibrosis models, thereby recovering liver function. We found that fetal liver, a hematopoietic tissue, highly expressed macrophage-recruiting factors and antifibrotic M2 macrophage polarization factors compared with the adult liver, resulting in fibrosis reduction because of CD163+ M2-macrophage polarization. Next, we created midgestational fetal liver-like hiPSC-LOs by fusion of hiPSC-LBs to induce static cell-cell interactions and found that these contained complex structures such as hepatocytes, vasculature, and bile ducts after transplantation. This fusion allowed the generation of a large human tissue suitable for transplantation into immunodeficient rodent models of liver fibrosis. hiPSC-LOs showed superior liver function compared with hiPSC-LBs and improved survival and liver function upon transplantation. In addition, hiPSC-LO transplantation ameliorated chemically induced liver fibrosis, a symptom of liver cirrhosis that leads to organ dysfunction, through immunomodulatory effects, particularly on CD163+ phagocytic M2-macrophage polarization. Together, our results suggest hiPSC-LO transplantation as a promising therapeutic option for liver fibrosis.


Assuntos
Imunomodulação , Células-Tronco Pluripotentes Induzidas , Cirrose Hepática , Fígado , Organoides , Humanos , Cirrose Hepática/patologia , Cirrose Hepática/terapia , Animais , Fígado/patologia , Macrófagos , Transplante de Fígado , Camundongos
3.
Sci Rep ; 13(1): 14996, 2023 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-37696857

RESUMO

Drug-induced liver fibrosis models are used in normal and immunosuppressed small animals for transplantation and regenerative medicine to improve liver fibrosis. Although large animal models are needed for pre-clinical studies, they are yet to be established owing to drug sensitivity in animal species and difficulty in setting doses. In this study, we evaluated liver fibrosis by administering thioacetamide (TA) to normal microminipig and thymectomized microminipig; 3 times for 1 week (total duration: 8 weeks). The pigs treated with TA showed elevated blood cytokine levels and a continuous liver injury at 8 weeks. RNA-seq of the liver showed increased expression of fibrosis-related genes after TA treatment. Histopathological examination showed degenerative necrosis of hepatocytes around the central vein, and revealed fibrogenesis and hepatocyte proliferation. TA treatment caused CD3-positive T cells and macrophages scattered within the hepatic lobule to congregate near the center of the lobule and increased αSMA-positive cells. Thymectomized pigs showed liver fibrosis similar to that of normal pigs, although the clinical signs tended to be milder. This model is similar to pathogenesis of liver fibrosis reported in other animal models. Therefore, it is expected to contribute to research as a drug discovery and pre-clinical transplantation models.


Assuntos
Cirrose Hepática , Tioacetamida , Animais , Suínos , Tioacetamida/toxicidade , Cirrose Hepática/induzido quimicamente , Proliferação de Células , Citocinas
4.
Cells ; 11(8)2022 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-35455938

RESUMO

Human induced pluripotent stem cells (hiPSCs) are a promising cell source for elucidating disease pathology and therapy. The mass supply of hiPSC-derived cells is technically feasible. Carriers that can contain a large number of hiPSC-derived cells and evaluate their functions in vivo-like environments will become increasingly important for understanding disease pathogenesis or treating end-stage organ failure. hiPSC-derived hepatocyte-like cells (hiPSC-HLCs; 5 × 108) were seeded into decellularized organ-derived scaffolds under circumfusion culture. The scaffolds were implanted into immunodeficient microminiature pigs to examine their applicability in vivo. The seeded hiPSC-HLCs demonstrated increased albumin secretion and up-regulated cytochrome P450 activities compared with those in standard two-dimensional culture conditions. Moreover, they showed long-term survival accompanied by neovascularization in vivo. The decellularized organ-derived scaffold is a promising carrier for hiPSC-derived cells for ex vivo and in vivo use and is an essential platform for regenerative medicine and research.


Assuntos
Células-Tronco Pluripotentes Induzidas , Animais , Diferenciação Celular , Hepatócitos , Humanos , Medicina Regenerativa , Suínos
5.
Stem Cell Res Ther ; 11(1): 425, 2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32993816

RESUMO

BACKGROUND: As a critical cellular component in the hepatic stem cell niche, hepatic stellate cells (HSCs) play critical roles in regulating the expansion of hepatic stem cells, liver regeneration, and fibrogenesis. However, the signaling of HSCs, particularly that involved in promoting hepatic stem cell expansion, remains unclear. While the overexpression of galectins has been identified in regenerating liver tissues, their involvement in cell-cell interactions between HSCs and hepatic stem cells remains to be elucidated. METHODS: To generate a liver regeneration rat model and establish a hepatic oval cell microenvironment as a stem cell niche, 2-acetylaminofluorene treatment plus partial hepatectomy was performed. Immunofluorescence staining was conducted to detect the emergence of hepatic stem cells and their niche. Liver parenchymal cells, non-parenchymal cells, and HSCs were isolated for gene and protein expression analysis by qPCR or western blotting. To evaluate the effect of galectins on the colony-forming efficiency of hepatic stem cells, c-Kit-CD29+CD49f+/lowCD45-Ter-119- cells were cultured with recombinant galectin protein, galectin antibody, galectin-producing HSCs, and galectin-knockdown HSCs. RESULTS: Following liver injury, the cytokeratin 19+ ductal cells were robustly induced together with the emergence of OV6+CD44+CD133+EpCAM+ hepatic stem cells. The activated desmin+ HSCs were recruited around the periportal area and markedly enriched in the galectin-positive domain compared to the other non-parenchymal cells. Notably, the HSC fraction isolated from regenerating liver was accompanied by dramatically elevated gene and protein expression of galectins. Hepatic stem cells co-cultured with HSCs significantly enhanced colony-forming efficiency. Conversely, single or double knockdown of galectin-1 and galectin-3 led into a significant function loss, impaired the co-cultured hepatic stem cells to attenuated colony size, inhibited colony frequency, and reduced total cell numbers in colonies. On the other hand, the promotive function of galectins was further confirmed by recombinant galectin protein supplementation and galectins blocking antibodies. CONCLUSIONS: Our findings, for the first time, demonstrated that galectins from activated HSCs contribute to hepatic stem cell expansion during liver regeneration, suggesting that galectins serve as important stem cell niche components.


Assuntos
Células Estreladas do Fígado , Regeneração Hepática , Animais , Galectinas/genética , Fígado , Ratos , Nicho de Células-Tronco
6.
Cell Transplant ; 29: 963689720964384, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33103476

RESUMO

Transplantation of liver organoids has been investigated as a treatment alternative to liver transplantation for chronic liver disease. Transportal approach can be considered as a method of delivering organoids to the liver. It is important to set the allowable organoid amount and verify translocation by intraportal transplantation. We first examined the transplantation tolerance and translocation of porcine fetal liver-derived allogeneic organoids using piglets. Fetal liver-derived organoids generated from the Kusabira Orange-transduced pig were transplanted to the 10-day-old piglet liver through the left branch of the portal vein. All recipients survived without any observable adverse events. In contrast, both local and main portal pressures increased transiently during transplantation. In necropsy samples, Kusabira Orange-positive donor cells were detected primarily in the target lobe of the liver and partly in other areas, including the lungs and brain. As we confirmed the transplantation allowance by porcine fetal liver-derived organoids, we performed intraportal transplantation of human-induced pluripotent stem cell (iPSC)-derived liver organoid, which we plan to use in clinical trials, and portal pressure and translocation were investigated. Human iPSC-derived liver organoids were transplanted into the same 10-day-old piglet. Portal hypertension and translocation of human iPSC-derived liver organoids to the lungs were observed in one of two transplanted animals. Translocation occurred in the piglet in which patent ductus venosus (PDV) was observed. Therefore, a 28-day-old piglet capable of surgically ligating PDV was used, and after the PDV was ligated, human iPSC-derived liver organoids with the amount of which is scheduled in clinical trials were transplanted. This procedure inhibited the translocation of human iPSC-derived liver organoids to extrahepatic sites without no portal hypertension. In conclusion, human iPSC-derived liver organoids can be safely transplanted through the portal vein. Ligation of the ductus venosus prior to transplantation was effective in inhibiting extrahepatic translocation in newborns and infants.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Organoides/citologia , Animais , Veia Porta/metabolismo , Transplante de Células-Tronco/métodos , Suínos
7.
Stem Cell Res Ther ; 9(1): 29, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29402311

RESUMO

BACKGROUND: Mature human hepatocytes are critical in preclinical research and therapy for liver disease, but are difficult to manipulate and expand in vitro. Hepatic stem cells (HpSCs) may be an alternative source of functional hepatocytes for cell therapy and disease modeling. Since these cells play an import role in regenerative medicine, the precise characterization that determines specific markers used to isolate these cells as well as whether they contribute to liver regeneration still remain to be shown. METHOD: In this study, human HpSCs were isolated from human primary fetal liver cells (FLCs) by flow cytometry using CDCP1, CD90, and CD66 antibodies. The isolated CDCP1+CD90+CD66- HpSCs were cultured on dishes coated with type IV collagen in DMEM nutrient mixture F-12 Ham supplemented with FBS, human γ-insulin, nicotinamide, dexamethasone, and L-glutamine for at least 2 weeks, and were characterized by transcriptomic profiling, quantitative real-time PCR, immunocytochemistry, and in-vivo transplantation. RESULTS: The purified CDCP1+CD90+CD66- subpopulation exhibited clonal expansion and self-renewal capability, and bipotential capacity was further identified in single cell-derived colonies containing distinct hepatocytes and cholangiocytes. Moreover, in-vivo liver repopulation assays demonstrated that human CDCP1+CD90+CD66- HpSCs repopulated over 90% of the mouse liver and differentiated into functional hepatocytes with drug metabolism activity. CONCLUSIONS: We identified a human hepatic stem/progenitor population in the CDCP1+CD90+CD66- subpopulation in human FLCs, indicating CDCP1 marker could potentially be utilized to identify and isolate HpSCs for further cytotherapy of liver disease.


Assuntos
Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Feto/metabolismo , Fígado/metabolismo , Proteínas de Neoplasias/metabolismo , Células-Tronco/metabolismo , Antígenos de Neoplasias , Técnicas de Cultura de Células , Células Cultivadas , Feto/citologia , Humanos , Fígado/citologia , Células-Tronco/citologia
8.
Stem Cell Res Ther ; 6: 51, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25881300

RESUMO

INTRODUCTION: The therapeutic potential of acyclic retinoid (ACR), a synthetic retinoid, has been confirmed in experimental and clinical studies. Therapeutic targets include precancerous and cancer stem cells. As ACR is also involved in developmental processes, its effect on normal hepatic stem cells (HpSCs) should be investigated for understanding the underlying mechanisms. Here, we examined effects of the acyclic retinoid peretinoin on fresh isolated murine HpSCs. METHODS: We isolated c-kit-CD29+CD49f+/lowCD45-Ter119- cells from murine fetal livers using flow cytometry. To evaluate the effect of ACR, we traced clonal expansion and analyzed cell differentiation as well as apoptosis during the induction process by immunofluorescent staining and marker gene expression. RESULTS: ACR dose-dependently inhibited HpSCs expansion. Stem cell clonal expansion was markedly inhibited during the culture period. Moreover, ACR showed a significant promotion of HpSC differentiation and induction of cellular apoptosis. The expression of stem cell marker genes, Afp, Cd44, and Dlk, was downregulated, while that of mature hepatocyte genes, Alb and Tat, and apoptosis-related genes, Annexin V and Caspase-3, were upregulated. Flow cytometry showed that the proportion of Annexin V-positive cells increased after ACR incubation compared with the control. Data obtained by immunofluorescent staining for albumin and Caspase-3 corroborated the data on gene expression. Finally, we found that ACR directly regulates the expression of retinoic acid receptors and retinoid X receptors. CONCLUSIONS: These findings indicate that ACR inhibits the clonal expansion of normal HpSCs in vitro and promotes the differentiation of immature cells by regulating receptors of retinoic acid.


Assuntos
Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Regeneração Hepática/fisiologia , Fígado/citologia , Tretinoína/análogos & derivados , Animais , Anexina A5/biossíntese , Proteínas de Ligação ao Cálcio , Caspase 3/biossíntese , Células Cultivadas , Regulação para Baixo , Citometria de Fluxo , Produtos do Gene tat/biossíntese , Receptores de Hialuronatos/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Receptores do Ácido Retinoico/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Tretinoína/farmacologia , Regulação para Cima , alfa-Fetoproteínas/biossíntese
9.
Stem Cell Res Ther ; 6: 49, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25889844

RESUMO

INTRODUCTION: Chimeric mice with humanized livers were recently established by transplanting human hepatocytes. This mouse model that is repopulated with functional human hepatocytes could be a useful tool for investigating human hepatic cell biology and drug metabolism and for other preclinical applications. Successfully transplanting human hepatocytes into mice requires that recipient mice with liver failure do not reject these human cells and provide a suitable microenvironment (supportive niche) to promote human donor cell expansion and differentiation. To overcome the limitations of current mouse models, we used Alb-TRECK/SCID mice for in vivo human immature hepatocyte differentiation and humanized liver generation. METHODS: 1.5 µg/kg diphtheria toxin was administrated into 8-week-old Alb-TRECK/SCID mice, and the degree of liver damage was assessed by serum aspartate aminotransferase activity levels. Forty-eight hours later, mice livers were sampled for histological analyses, and the human donor cells were then transplanted into mice livers on the same day. Chimeric rate and survival rate after cell transplantation was evaluated. Expressions of human hepatic-related genes were detected. A human albumin enzyme-linked immunosorbent assay was performed after 50 days of transplantation. On day 60 after transplantation, drug metabolism was examined in mice. RESULTS: Both human primary fetal liver cells and hepatic stem cells were successfully repopulated in the livers of Alb-TRECK/SCID mice that developed lethal fulminant hepatic failure after administering diphtheria toxin; the repopulation rate in some mice was nearly 100%. Compared with human primary fetal liver cells, human hepatic stem cell transplantation rescued Alb-TRECK/SCID mice with lethal fulminant hepatic failure, and human hepatic stem cell-derived humanized livers secreted more human albumin into mouse sera and also functioned as a "human liver" that could metabolize the drugs ketoprofen and debrisoquine. CONCLUSION: Our model of a humanized liver in Alb-TRECK/SCID mice may provide for functional applications such as drug metabolism, drug to drug interactions, and promote other in vivo and in vitro studies.


Assuntos
Hepatócitos/transplante , Inativação Metabólica/fisiologia , Falência Hepática Aguda/terapia , Regeneração Hepática/fisiologia , Transplante de Células-Tronco/métodos , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Quimera , Debrisoquina/metabolismo , Toxina Diftérica/administração & dosagem , Modelos Animais de Doenças , Hepatócitos/citologia , Humanos , Cetoprofeno/metabolismo , Fígado/citologia , Falência Hepática Aguda/induzido quimicamente , Camundongos , Camundongos Knockout , Camundongos SCID , Células-Tronco/metabolismo , Transplante Heterólogo
10.
Stem Cells Dev ; 23(18): 2237-49, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24804872

RESUMO

Hepatocellular carcinoma (HCC) is a malignant tumor associated with a generally poor prognosis and a high rate of recurrence. HCC usually develops in the context of chronic liver diseases, and long-lasting premalignant conditions precede cancer development. A promising therapeutic approach is to eliminate precancerous cells, which are considered as the precursors of cancer stem cells, to prevent further malignant transformation. In this study, we identified a subpopulation of precancerous cells in a rat liver carcinogenesis model, which were enriched in CD133(+)CD44(+)CD45(-)HIS49(-) cells that formed part of the hepatic oval cells fraction. Prospective isolation of the precancerous cells using flow cytometry identified stem cell properties such as the ability to expand clonally and differentiate into bi-lineage cell types. Furthermore, an acyclic retinoid, which was recently shown to improve overall survival after HCC resection, directly inhibited the extensive expansion of the isolated precancerous cells in vitro and decreased the emergence of the precancerous cells and their progeny in vivo. Long-term follow-up after the acyclic retinoid treatment confirmed reduction in precancerous changes, ultimately resulting in suppression of HCC development. These findings, together with data from recent clinical trials showing marked reduction in intrahepatic recurrence, suggest that acyclic retinoid directly prevents de novo HCC by inhibiting the development of precancerous cells. Given recent advances in diagnostic techniques and the establishment of surveillance programs, the targeting of precancerous cells may have a huge impact on preventative cancer therapies.


Assuntos
Antígenos CD/metabolismo , Carcinoma Hepatocelular/patologia , Glicoproteínas/metabolismo , Receptores de Hialuronatos/metabolismo , Neoplasias Hepáticas/patologia , Terapia de Alvo Molecular , Peptídeos/metabolismo , Lesões Pré-Cancerosas/patologia , Antígeno AC133 , Animais , Carcinoma Hepatocelular/metabolismo , Proliferação de Células , Separação Celular , Glutationa Transferase/metabolismo , Neoplasias Hepáticas/metabolismo , Masculino , Lesões Pré-Cancerosas/metabolismo , Ratos Endogâmicos F344
11.
J Hepatobiliary Pancreat Sci ; 19(6): 621-5, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22878837

RESUMO

Cancer is the leading cause of death worldwide and mortality due to cancer continues to rise. Cancer cell resistance to chemoradiotherapy is hindering treatment efforts in clinics. Prevention strategies and early detection thus may reduce mortality. In this study, we have proposed the concept of using precancerous cells and their progeny in cancer therapy, which could provide unique insights for early cancer diagnosis, treatment, and preventive therapy. In addition to discussing the nature and characteristics of precancerous cells and their progeny, we have also introduced an effective precancerous cell-targeted therapy based on an animal model of hepatocellular carcinoma. Anti-precancerous cell drug development should be a major target during cancer elimination and it may lead to preventive therapies for individuals with a high risk of developing cancer.


Assuntos
Carcinoma Hepatocelular/terapia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Neoplasias Hepáticas/terapia , Células-Tronco Neoplásicas/patologia , Lesões Pré-Cancerosas/diagnóstico , Lesões Pré-Cancerosas/terapia , Carcinoma Hepatocelular/patologia , Humanos , Neoplasias Hepáticas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA