Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Transl Med ; 16(757): eadg0338, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39047116

RESUMEN

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.


Asunto(s)
Inmunomodulación , Células Madre Pluripotentes Inducidas , Cirrosis Hepática , Hígado , Organoides , Humanos , Cirrosis Hepática/patología , Cirrosis Hepática/terapia , Animales , Hígado/patología , Macrófagos , Trasplante de Hígado , Ratones
2.
Regen Ther ; 26: 219-234, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38903867

RESUMEN

Liver is involved in metabolic reactions, ammonia detoxification, and immunity. Multicellular liver tissue cultures are more desirable for drug screening, disease modeling, and researching transplantation therapy, than hepatocytes monocultures. Hepatocytes monocultures are not stable for long. Further, hepatocyte-like cells induced from pluripotent stem cells and in vivo hepatocytes are functionally dissimilar. Organoid technology circumvents these issues by generating functional ex vivo liver tissue from intrinsic liver progenitor cells and extrinsic stem cells, including pluripotent stem cells. To function as in vivo liver tissue, the liver organoid cells must be arranged precisely in the 3-dimensional space, closely mimicking in vivo liver tissue. Moreover, for long term functioning, liver organoids must be appropriately vascularized and in contact with neighboring epithelial tissues (e.g., bile canaliculi and intrahepatic bile duct, or intrahepatic and extrahepatic bile ducts). Recent discoveries in liver developmental biology allows one to successfully induce liver component cells and generate organoids. Thus, here, in this review, we summarize the current state of knowledge on liver development with a focus on its application in generating different liver organoids. We also cover the future prospects in creating (functionally and structurally) in vivo-like liver organoids using the current knowledge on liver development.

3.
Cells ; 12(23)2023 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-38067145

RESUMEN

Mature hepatocytes (MHs) in an adult rodent liver are categorized into the following three subpopulations based on their proliferative capability: type I cells (MH-I), which are committed progenitor cells that possess a high growth capability and basal hepatocytic functions; type II cells (MH-II), which possess a limited proliferative capability; and type III cells (MH-III), which lose the ability to divide (replicative senescence) and reach the final differentiated state. These subpopulations may explain the liver's development and growth after birth. Generally, small-sized hepatocytes emerge in mammal livers. The cells are characterized by being morphologically identical to hepatocytes except for their size, which is substantially smaller than that of ordinary MHs. We initially discovered small hepatocytes (SHs) in the primary culture of rat hepatocytes. We believe that SHs are derived from MH-I and play a role as hepatocytic progenitors to supply MHs. The population of MH-I (SHs) is distributed in the whole lobules, a part of which possesses a self-renewal capability, and decreases with age. Conversely, injured livers of experimental models and clinical cases showed the emergence of SHs. Studies demonstrate the involvement of SHs in liver regeneration. SHs that appeared in the injured livers are not a pure population but a mixture of two distinct origins, MH-derived and hepatic-stem-cell-derived cells. The predominant cell-derived SHs depend on the proliferative capability of the remaining MHs after the injury. This review will focus on the SHs that appeared in the liver and discuss the significance of SHs in liver regeneration.


Asunto(s)
Hepatocitos , Hígado , Ratas , Animales , Ratas Endogámicas F344 , Diferenciación Celular/fisiología , Células Madre , Mamíferos
4.
Cell Rep ; 42(11): 113420, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37955987

RESUMEN

The aggressiveness of pancreatic ductal adenocarcinoma (PDAC) is affected by the tumor microenvironment (TME). In this study, to recapitulate the PDAC TME ex vivo, we cocultured patient-derived PDAC cells with mesenchymal and vascular endothelial cells derived from human induced pluripotent stem cells (hiPSCs) to create a fused pancreatic cancer organoid (FPCO) in an air-liquid interface. FPCOs were further induced to resemble two distinct aspects of PDAC tissue. Quiescent FPCOs were drug resistant, likely because the TME consisted of abundant extracellular matrix proteins that were secreted from the various types of cancer-associated fibroblasts (CAFs) derived from hiPSCs. Proliferative FPCOs could re-proliferate after anticancer drug treatment, suggesting that this type of FPCO would be useful for studying PDAC recurrence. Thus, we generated PDAC organoids that recapitulate the heterogeneity of PDAC tissue and are a potential platform for screening anticancer drugs.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinoma Ductal Pancreático , Células Madre Pluripotentes Inducidas , Neoplasias Pancreáticas , Humanos , Fibroblastos Asociados al Cáncer/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Endoteliales/metabolismo , Línea Celular Tumoral , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/patología , Células del Estroma/metabolismo , Organoides/metabolismo , Microambiente Tumoral
5.
Sci Rep ; 13(1): 7084, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37127664

RESUMEN

Intrahepatic nerves are involved in the regulation of metabolic reactions and hepatocyte-based regeneration after surgical resection, although their contribution to chronic liver injury remains unknown. Given that intrahepatic nerves are abundant in the periportal tissue, they may be correlated also with cholangiocyte-based regeneration. Here we demonstrate that isoproterenol (ISO), a ß-adrenergic receptor agonist, promoted ductular expansion induced by 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) in vivo. Immunofluorescence analysis shows that nerve fibers positive for tyrosine hydroxylase form synaptophysin-positive nerve endings on epithelial cell adhesion molecule-positive (EpCAM+) cholangiocytes as well as on Thy1+ periportal mesenchymal cells (PMCs) that surround bile ducts, suggesting that the intrahepatic biliary tissue are targeted by sympathetic nerves. In vitro analyses indicate that ISO directly increases cAMP levels in cholangiocytes and PMCs. Mechanistically, ISO expands the lumen of cholangiocyte organoids, resulting in promotion of cholangiocyte proliferation, whereas it increases expression of fibroblast growth factor 7, a growth factor for cholangiocytes, in PMCs. Taken together, the results indicate that intrahepatic sympathetic nerves regulate remodeling of bile ducts during DDC-injury by the activation of ß-adrenergic receptors on cholangiocytes and PMCs.


Asunto(s)
Hígado , Piridinas , Hígado/metabolismo , Piridinas/farmacología , Conductos Biliares/metabolismo , Agonistas Adrenérgicos beta/farmacología
6.
Stem Cell Res Ther ; 14(1): 134, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37194082

RESUMEN

BACKGROUND: Small hepatocyte-like progenitor cells (SHPCs) are hepatocytic progenitor cells that transiently form clusters in rat livers treated with retrorsine (Ret) that underwent 70% partial hepatectomy (PH). We previously reported that transplantation of Thy1+ cells obtained from D-galactosamine-treated livers promotes SHPC expansion, thereby accelerating liver regeneration. Extracellular vesicles (EVs) secreted by Thy1+ cells induce sinusoidal endothelial cells (SECs) and Kupffer cells (KCs) to secrete IL17B and IL25, respectively, thereby activating SHPCs through IL17 receptor B (RB) signaling. This study aimed to identify the inducers of IL17RB signaling and growth factors for SHPC proliferation in EVs secreted by Thy1+ cells (Thy1-EVs). METHODS: Thy1+ cells isolated from the livers of rats treated with D-galactosamine were cultured. Although some liver stem/progenitor cells (LSPCs) proliferated to form colonies, others remained as mesenchymal cells (MCs). Thy1-MCs or Thy1-LSPCs were transplanted into Ret/PH-treated livers to examine their effects on SHPCs. EVs were isolated from the conditioned medium (CM) of Thy1-MCs and Thy1-LSPCs. Small hepatocytes (SHs) isolated from adult rat livers were used to identify factors regulating cell growth in Thy1-EVs. RESULTS: The size of SHPC clusters transplanted with Thy1-MCs was significantly larger than that of SHPC clusters transplanted with Thy1-LSPCs (p = 0.02). A comprehensive analysis of Thy1-MC-EVs revealed that miR-199a-5p, cytokine-induced neutrophil chemoattractant-2 (CINC-2), and monocyte chemotactic protein 1 (MCP-1) were candidates for promoting SHPC growth. Additionally, miR-199a-5p mimics promoted the growth of SHs (p = 0.02), whereas CINC-2 and MCP-1 did not. SECs treated with CINC-2 induced Il17b expression. KCs treated with Thy1-EVs induced the expression of CINC-2, Il25, and miR-199a-5p. CM derived from SECs treated with CINC-2 accelerated the growth of SHs (p = 0.03). Similarly, CM derived from KCs treated with Thy1-EVs and miR-199a-5p mimics accelerated the growth of SHs (p = 0.007). In addition, although miR-199a-overexpressing EVs could not enhance SHPC proliferation, transplantation of miR-199a-overexpressing Thy1-MCs could promote the expansion of SHPC clusters. CONCLUSION: Thy1-MC transplantation may accelerate liver regeneration owing to SHPC expansion, which is induced by CINC-2/IL17RB signaling and miR-199a-5p via SEC and KC activation.


Asunto(s)
Quimiocinas CXC , Vesículas Extracelulares , MicroARNs , Animales , Ratas , Proliferación Celular , Células Endoteliales , Galactosamina , Hepatocitos/metabolismo , Regeneración Hepática/fisiología , MicroARNs/genética , MicroARNs/metabolismo , Ratas Endogámicas F344 , Células Madre/metabolismo , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo
7.
Methods Mol Biol ; 2544: 183-193, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36125719

RESUMEN

Small hepatocyte-like progenitor cells (SHPCs) are known as liver stem/progenitor cells (LSPCs). SHPCs transiently appear and form clusters in rat livers treated with retrorsine (Ret) and a 70% partial hepatectomy (PH). The Ret/PH model has been used widely to analyze the effectiveness of cell transplantation and the mechanisms of LSPC proliferation. Laser microdissection (LMD) is a powerful tool that can excise and collect specific areas of cells from a tissue slice with a laser under a microscope. These cells exhibiting morphological alterations different from the surrounding cells may be analyzed by gene expression profiling. Specific markers of SHPCs have not yet been identified, in part, because it is difficult to isolate SHPCs from the liver using fluorescence or magnetic-activated cell sorting. To examine the underlying mechanism for SHPC growth, we established comprehensive gene expression profiles for SHPCs captured from liver sections using LMD. In this chapter, we introduce a method to isolate SHPCs from liver tissue sections using LMD for gene expression analysis.


Asunto(s)
Hepatectomía , Regeneración Hepática , Animales , Hepatocitos/metabolismo , Captura por Microdisección con Láser , Hígado/cirugía , Alcaloides de Pirrolicidina , Ratas , Ratas Endogámicas F344 , Células Madre/metabolismo
8.
Methods Mol Biol ; 2544: 269-278, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36125726

RESUMEN

Hepatocytes are liver parenchymal cells involved in performing various metabolic reactions. During the development of therapeutic drugs, toxicological assays are conducted using hepatocyte cultures before clinical trials. However, since primary hepatocytes cannot proliferate and rapidly lose their functions in vitro, many efforts have been put into modifying culture conditions to expand primary hepatocytes and induce hepatocyte functions in intrinsic and extrinsic stem/progenitor cells. In this chapter, we summarize recent advances in preparing hepatocyte cultures and induction of hepatocytes from various cellular sources.


Asunto(s)
Hepatocitos , Células Madre , Hepatocitos/metabolismo , Humanos
9.
Semin Fetal Neonatal Med ; 27(1): 101229, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33745829

RESUMEN

The liver emerges from the ventral foregut endoderm around 3 weeks in human and 1 week in mice after fertilization. The fetal liver works as a hematopoietic organ and then develops functions required for performing various metabolic reactions in late fetal and neonatal periods. In parallel with functional differentiation, the liver establishes three dimensional tissue structures. In particular, establishment of the bile excretion system consisting of bile canaliculi of hepatocytes and bile ducts of cholangiocytes is critical to maintain healthy tissue status. This is because hepatocytes produce bile as they functionally mature, and if allowed to remain within the liver tissue can lead to cytotoxicity. In this review, we focus on epithelial tissue morphogenesis in the perinatal period and cholestatic liver diseases caused by abnormal development of the biliary system.


Asunto(s)
Colestasis , Hígado , Animales , Conductos Biliares/metabolismo , Colestasis/metabolismo , Modelos Animales de Enfermedad , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Ratones
10.
Stem Cell Reports ; 16(12): 3050-3063, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34861166

RESUMEN

Hepatic stellate cells (HSCs) play a central role in the progression of liver fibrosis by producing extracellular matrices. The development of drugs to suppress liver fibrosis has been hampered by the lack of human quiescent HSCs (qHSCs) and an appropriate in vitro model that faithfully recapitulates HSC activation. In the present study, we developed a culture system to generate qHSC-like cells from human-induced pluripotent stem cells (hiPSCs) that can be converted into activated HSCs in culture. To monitor the activation process, a red fluorescent protein (RFP) gene was inserted in hiPSCs downstream of the activation marker gene actin alpha 2 smooth muscle (ACTA2). Using qHSC-like cells derived from RFP reporter iPSCs, we screened a repurposing chemical library and identified therapeutic candidates that prevent liver fibrosis. Hence, hiPSC-derived qHSC-like cells will be a useful tool to study the mechanism of HSC activation and to identify therapeutic agents.


Asunto(s)
Técnicas de Cultivo de Célula , Ciclo Celular , Descubrimiento de Drogas , Células Estrelladas Hepáticas/citología , Células Madre Pluripotentes Inducidas/citología , Modelos Biológicos , Animales , Evaluación Preclínica de Medicamentos , Perfilación de la Expresión Génica , Células Estrelladas Hepáticas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Masculino , Ratones Endogámicos C57BL
11.
Nat Commun ; 12(1): 3390, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099675

RESUMEN

In the liver, the bile canaliculi of hepatocytes are connected to intrahepatic bile ducts lined with cholangiocytes, which remove cytotoxic bile from the liver tissue. Although liver organoids have been reported, it is not clear whether the functional connection between hepatocytes and cholangiocytes is recapitulated in those organoids. Here, we report the generation of a hepatobiliary tubular organoid (HBTO) using mouse hepatocyte progenitors and cholangiocytes. Hepatocytes form the bile canalicular network and secrete metabolites into the canaliculi, which are then transported into the biliary tubular structure. Hepatocytes in HBTO acquire and maintain metabolic functions including albumin secretion and cytochrome P450 activities, over the long term. In this study, we establish functional liver tissue incorporating a bile drainage system ex vivo. HBTO enable us to reproduce the transport of hepatocyte metabolites in liver tissue, and to investigate the way in which the two types of epithelial cells establish functional connections.


Asunto(s)
Conductos Biliares Intrahepáticos/citología , Comunicación Celular/fisiología , Hígado/citología , Organoides/fisiología , Cultivo Primario de Células/métodos , Animales , Conductos Biliares Intrahepáticos/fisiología , Diferenciación Celular , Células Cultivadas , Hepatocitos/fisiología , Hígado/fisiología , Ratones , Organoides/citología , Células Madre/fisiología
12.
Stem Cell Res Ther ; 12(1): 312, 2021 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-34051870

RESUMEN

BACKGROUND: Small hepatocyte-like progenitor cells (SHPCs) appear to form transient clusters in rat livers treated with retrorsine (Ret) and 70% partial hepatectomy (PH). We previously reported that the expansion of SHPCs was amplified in Ret/PH-treated rat livers transplanted with Thy1+ cells derived from D-galactosamine-treated injured livers. Extracellular vesicles (EVs) produced by hepatic Thy1+ donor cells activated SHPCs via interleukin (IL)-17 receptor B signaling. As bone marrow-derived mesenchymal cells (BM-MCs) also express Thy1, we aimed to determine whether BM-MCs could also promote the growth of SHPCs. METHODS: BM-MCs were isolated from dipeptidyl-peptidase IV (DPPIV)-positive rats. BM-MCs or BM-MC-derived EVs were administered to DPPIV-negative Ret/PH rat livers, and the growth and the characteristics of SHPC clusters were evaluated 14 days post-treatment. miRNA microarrays and cytokine arrays examined soluble factors within EVs. Small hepatocytes (SHs) isolated from an adult rat liver were used to identify factors enhancing hepatocytic progenitor cells growth. RESULTS: The recipient's livers were enlarged at 2 weeks post-BM-MC transplantation. The number and the size of SHPCs increased remarkably in livers transplanted with BM-MCs. BM-MC-derived EVs also stimulated SHPC growth. Comprehensive analyses revealed that BM-MC-derived EVs contained miR-146a-5p, interleukin-6, and stem cell factor, which could enhance SHs' proliferation. Administration of EVs derived from the miR-146a-5p-transfected BM-MCs to Ret/PH rat livers remarkably enhanced the expansion of SHPCs. CONCLUSIONS: miR-146a-5p involved in EVs produced by BM-MCs may play a major role in accelerating liver regeneration by activating the intrinsic hepatocytic progenitor cells.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Animales , Médula Ósea , Células de la Médula Ósea , Hepatocitos , MicroARNs/genética , Ratas , Ratas Endogámicas F344
13.
Aging (Albany NY) ; 12(19): 18907-18927, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-33001859

RESUMEN

The liver gradually loses its regenerative capabilities with aging. However, it remains unknown whether aging affects drug-induced liver injury. Here, we used acetaminophen induced acute liver injury model to compare tissue injury and regeneration of aged mice (>80 weeks old) with young ones (8-10 weeks old). The mortality of aged mice after acetaminophen injury was higher than that of young mice. Transient increase of serum GOT and decrease of reduced glutathione (GSH) were not returned to original levels in aged mice even at 48 hours. In addition, Foxm1b and its targets Ccnd1 and Cdk1 were upregulated in young but not in aged mice after 48 hours. Moreover, an apoptosis-related gene, Cidea, was upregulated specifically in aged livers, which was consistent with increased number of TUNEL+ hepatocytes. Unexpectedly, damaged hepatocytes were retained in aged liver tissue, which may be caused by impaired recruitment of macrophages to the damaged area, without increases in Ccl2 after acetaminophen injury. Collectively, prolonged oxidative stress due to delayed recovery of GSH and the retention of damaged hepatocytes may suppress tissue repair and hepatocyte proliferation, resulting in exacerbation of acetaminophen injury in aged mice. Thus, aging is a risk factor conferring susceptibility against drug-induced liver injury.

14.
Hepatol Commun ; 4(1): 21-37, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31909353

RESUMEN

In this study, we investigated how the ability of hepatocytic parental progenitor cells (HPPCs) to self-renew can be maintained and how laminin (LN) isoforms play an important role in their self-renewal and maturation. Hepatocytes isolated from adult rat livers were cultured on hyaluronic acid to form colonies consisting of CD44+ small hepatocytes, which could be passaged on dishes coated with Matrigel. When second-passage cells were plated on Matrigel, LN111, or LN511, HPPCs appeared on Matrigel and LN111 but not on LN511. We identified two types of cells among the second-passage cells: Small, round cells and large, flat ones were observed on Matrigel, whereas the former and latter ones were specifically attached on LN111 and LN511, respectively. We hypothesized that small and round cells are the origin of HPPC colonies, and the binding to LN111 could be key to maintaining their self-renewal capability. Among the integrins involved in LN binding, integrins α3 and ß1 were expressed in colonies on LN111 more than in those on LN511, whereas ß4 was more strongly expressed in colonies on LN511. Integrin α3highα6ß1high cells could form HPPC colonies on LN111 but not on LN511, whereas integrin α6ß1low cells could not on either LN111 or LN511. In addition, neutralizing anti-integrin ß1 and anti-LN111 antibodies inhibited the passaged cells' ability to attach and form colonies on LN111 by HPPCs. Matrigel overlay induced second-passage cells growing on LN111 to increase their expression of hepatic functional genes and to form 3-dimensional colonies with bile canalicular networks, whereas such a shift was poorly induced when they were grown onLN511. Conclusion: These results suggest that the self-renewal capability of HPPCs depends on LN111 through integrin ß1 signaling.

15.
Methods Mol Biol ; 1940: 267-273, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30788832

RESUMEN

The liver performs a number of physiologically important functions. Hepatocytes are the liver parenchymal cells performing most of those functions. Therefore, it is important to recover functional hepatocytes after hepatic injury and prepare a mass of hepatocytes for regenerative medicine. We have found that mature hepatocytes dedifferentiate to hepatocyte progenitors in chronically injured mouse liver. Those hepatocyte progenitors can be isolated as CD24+EpCAM- cells from the CD31-CD45- fraction, which clonally proliferate and efficiently re-differentiate to functional hepatocytes both in vitro and in vivo. Here, I describe the methods to isolate hepatocyte progenitors from chronically injured liver, to expand them in vitro, and to induce differentiation into functional hepatocytes.


Asunto(s)
Hepatocitos/citología , Hígado/citología , Hígado/lesiones , Células Madre/citología , Animales , Antígeno CD24/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Molécula de Adhesión Celular Epitelial/metabolismo , Citometría de Flujo , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Endogámicos C57BL , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Piridinas/farmacología , Factor de Transcripción SOX9/metabolismo
16.
Methods Mol Biol ; 1905: 9-17, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30536086

RESUMEN

Liver stem/progenitor cells (LPCs) are defined as bipotential cells differentiating into both hepatocytes and cholangiocytes. The Notch, TGFß, and Hippo pathways have been implicated in lineage determination of LPCs during development and regeneration. However, the molecular mechanisms governing the lineage specification have not been fully elucidated, yet. Epithelial adhesion molecule (EpCAM) is a marker of cholangiocytes and of LPCs. We found that EpCAM+ cells isolated from neonatal liver contain LPCs that clonally proliferate and are bipotential in vitro and in vivo. Furthermore, EpCAM+ progenies keep the capacity of bidirectional differentiation even after long-term culture. These cells are useful to investigate the molecular mechanisms regulating lineage commitment and epithelial differentiation of LPCs.


Asunto(s)
Separación Celular/métodos , Molécula de Adhesión Celular Epitelial/metabolismo , Hígado/citología , Células Madre/citología , Animales , Animales Recién Nacidos , Conductos Biliares/citología , Conductos Biliares/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Citometría de Flujo , Hepatocitos/citología , Hepatocitos/metabolismo , Hígado/metabolismo , Ratones , Células Madre/metabolismo
17.
Development ; 145(9)2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29615468

RESUMEN

Epithelial organs consist of multiple tissue structures, such as epithelial sheets, blood vessels and nerves, which are spatially organized to achieve optimal physiological functions. The hepatic nervous system has been implicated in physiological functions and regeneration of the liver. However, the processes of development and reconstruction of the intrahepatic nerve network and its underlying mechanisms remain unknown. Here, we demonstrate that neural class III ß-tubulin (TUBB3)+ nerve fibers are not distributed in intrahepatic tissue at embryonic day 17.5; instead, they gradually extend along the periportal tissue, including intrahepatic bile ducts (IHBDs), after birth. Nerve growth factor (Ngf) expression increased in biliary epithelial cells (BECs) and mesenchymal cells next to BECs before nerve fiber extension, and Ngf was upregulated by hairy enhancer of slit 1 (Hes family bHLH transcription factor 1; Hes1). Ectopic NGF expression in mature hepatocytes induced nerve fiber extension into the parenchymal region, from where these fibers are normally excluded. Furthermore, after BECs were damaged by the administration of 4,4-diaminodiphenylmethane, the nerve network appeared shrunken; however, it was reconstructed after IHBD regeneration, which depended on the NGF signal. These results suggest that IHBDs guide the extension of nerve fibers by secreting NGF during nerve fiber development and regeneration.


Asunto(s)
Conductos Biliares Intrahepáticos , Células Epiteliales/metabolismo , Hepatocitos/metabolismo , Regeneración Hepática/fisiología , Hígado , Red Nerviosa/embriología , Animales , Conductos Biliares Intrahepáticos/embriología , Conductos Biliares Intrahepáticos/inervación , Hígado/embriología , Hígado/inervación , Ratones , Factor de Crecimiento Nervioso/biosíntesis , Factor de Crecimiento Nervioso/genética , Factor de Transcripción HES-1/genética , Factor de Transcripción HES-1/metabolismo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
18.
Sci Rep ; 7: 46177, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28397810

RESUMEN

The liver has a variety of functions for maintaining homeostasis, and hepatocytes play a major role. In contrast with the high regenerative capacity of mature hepatocytes (MHs) in vivo, they have not been successfully expanded ex vivo. Here we demonstrate that CD44-positive cells sorted from small hepatocyte (SH) colonies derived from a healthy adult rat liver can proliferate on a Matrigel-coated dish in serum-free chemically defined medium; in addition, a subpopulation of the cells can divide more than 50 times in a period of 17 weeks every 4-week-passage. The passage cells retained the capability to recover highly differentiated functions, such as glycogen storage, CYP activity and bile secretion. When Matrigel-treated cells from the third passage were transplanted into retrorsine/partial hepatectomy-treated rat livers, the cells engrafted to differentiate into MHs and cholangiocytes. These results suggest that long-term cultured CD44+ SHs retain hepatocytic characteristics in vitro and the capability to differentiate into hepatocytes and cholangiocytes in vivo. Thus, a newly identified subpopulation of MHs possessing the attributes of hepatocytic stem/progenitor cells can be passaged several times without losing hepatocytic characteristics.


Asunto(s)
Autorrenovación de las Células , Tamaño de la Célula , Hepatocitos/citología , Células Madre/citología , Animales , Canalículos Biliares/citología , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Colágeno/farmacología , Ensayo de Unidades Formadoras de Colonias , Combinación de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/ultraestructura , Receptores de Hialuranos/metabolismo , Laminina/farmacología , Hígado/citología , Proteoglicanos/farmacología , Ratas , Ratas Endogámicas F344 , Trasplante de Células Madre , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Factores de Tiempo
19.
20.
Artículo en Inglés | MEDLINE | ID: mdl-28213465

RESUMEN

Tissue stem/progenitor cells supply multiple types of epithelial cells that eventually acquire specialized functions during organ development. In addition, three-dimensional (3D) tissue structures need to be established for organs to perform their physiological functions. The liver contains two types of epithelial cells, namely, hepatocytes and cholangiocytes, which are derived from hepatoblasts, fetal liver stem/progenitor cells (LPCs), in mid-gestation. Hepatocytes performing many metabolic reactions form cord-like structures, whereas cholangiocytes, biliary epithelial cells, form tubular structures called intrahepatic bile ducts. Analyses for human genetic diseases and mutant mice have identified crucial molecules for liver organogenesis. Functions of those molecules can be examined in in vitro culture systems where LPCs are induced to differentiate into hepatocytes or cholangiocytes. Recent technical advances have revealed 3D epithelial morphogenesis during liver organogenesis. Therefore, the liver is a good model to understand how tissue stem/progenitor cells differentiate and establish 3D tissue architectures during organ development.


Asunto(s)
Hígado/embriología , Animales , Epitelio/embriología , Hepatocitos/fisiología , Humanos , Hígado/citología , Organogénesis , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...