Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 4941, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38866781

RESUMO

Despite widespread adoption of tissue clearing techniques in recent years, poor access to suitable light-sheet fluorescence microscopes remains a major obstacle for biomedical end-users. Here, we present descSPIM (desktop-equipped SPIM for cleared specimens), a low-cost ($20,000-50,000), low-expertise (one-day installation by a non-expert), yet practical do-it-yourself light-sheet microscope as a solution for this bottleneck. Even the most fundamental configuration of descSPIM enables multi-color imaging of whole mouse brains and a cancer cell line-derived xenograft tumor mass for the visualization of neurocircuitry, assessment of drug distribution, and pathological examination by false-colored hematoxylin and eosin staining in a three-dimensional manner. Academically open-sourced ( https://github.com/dbsb-juntendo/descSPIM ), descSPIM allows routine three-dimensional imaging of cleared samples in minutes. Thus, the dissemination of descSPIM will accelerate biomedical discoveries driven by tissue clearing technologies.


Assuntos
Encéfalo , Imageamento Tridimensional , Microscopia de Fluorescência , Animais , Camundongos , Encéfalo/diagnóstico por imagem , Humanos , Microscopia de Fluorescência/métodos , Microscopia de Fluorescência/instrumentação , Imageamento Tridimensional/métodos , Linhagem Celular Tumoral
2.
Biochem Biophys Res Commun ; 658: 1-9, 2023 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-37004297

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a serious disease with poor prognosis and prone to chemotherapy resistance. It is speculated that the tumor microenvironment (TME) of PDAC contributes to these characteristics. However, the detailed mechanisms of interactions between pancreatic cancer cells and stroma in the TME are unclear. Therefore, the aim of this study was to establish a co-culture system that mimics the TME, using cancer cells derived from PDAC patient specimens and stellate cells from human induced pluripotent stem cells as stromal cells. We succeeded in observing the interaction between cancer cells and stellate cells and reproduced some features of PDAC in vitro using our co-culture systems. In addition, we demonstrated the applicability of our co-culture system for drug treatment in vitro. To conclude, we propose our co-culture system as a novel method to analyze cell-cell interactions, especially in the TME of PDAC.


Assuntos
Carcinoma Ductal Pancreático , Células-Tronco Pluripotentes Induzidas , Neoplasias Pancreáticas , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Técnicas de Cocultura , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/patologia , Microambiente Tumoral , Células Estreladas do Pâncreas/patologia , Linhagem Celular Tumoral , Neoplasias Pancreáticas
3.
Stem Cell Rev Rep ; 18(8): 2995-3007, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35661077

RESUMO

For safe regenerative medicines, contaminated or remaining tumorigenic undifferentiated cells in cell-derived products must be rigorously assessed through sensitive assays. Although in vitro nucleic acid tests offer particularly sensitive tumorigenicity-associated assays, the human pluripotent stem cell (hPSC) detectability is partly constrained by the small input amount of RNA per test. To overcome this limitation, we developed reverse transcription loop-mediated isothermal amplification (RT-LAMP) assays that are highly gene specific and robust against interfering materials. LAMP could readily assay microgram order of input sample per test and detected an equivalent model of 0.00002% hiPSC contamination in a simple one-pot reaction. For the evaluation of cell-derived total RNA, RT-LAMP detected spiked-in hPSCs among hPSC-derived trilineage cells utilizing multiple pluripotency RNAs. We also developed multiplex RT-LAMP assays and further applied for in situ cell imaging, achieving specific co-staining of pluripotency proteins and RNAs. Our attempts uncovered the utility of RT-LAMP approaches for tumorigenicity-associated assays, supporting practical applications of regenerative medicine.


Assuntos
Técnicas de Amplificação de Ácido Nucleico , Células-Tronco Pluripotentes , Humanos , Sensibilidade e Especificidade , Técnicas de Amplificação de Ácido Nucleico/métodos , RNA
4.
Cells ; 10(12)2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34944048

RESUMO

For practical use of pluripotent stem cells (PSCs) for disease modelling, drug screening, and regenerative medicine, the cell differentiation process needs to be properly refined to generate end products with consistent and high quality. To construct and optimize a robust cell-induction process, a myriad of cell culture conditions should be considered. In contrast to inefficient brute-force screening, statistical design of experiments (DOE) approaches, such as factorial design, orthogonal array design, response surface methodology (RSM), definitive screening design (DSD), and mixture design, enable efficient and strategic screening of conditions in smaller experimental runs through multifactorial screening and/or quantitative modeling. Although DOE has become routinely utilized in the bioengineering and pharmaceutical fields, the imminent need of more detailed cell-lineage specification, complex organoid construction, and a stable supply of qualified cell-derived material requires expedition of DOE utilization in stem cell bioprocessing. This review summarizes DOE-based cell culture optimizations of PSCs, mesenchymal stem cells (MSCs), hematopoietic stem cells (HSCs), and Chinese hamster ovary (CHO) cells, which guide effective research and development of PSC-derived materials for academic and industrial applications.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Proliferação de Células , Árvores de Decisões , Humanos , Projetos de Pesquisa
5.
Front Genet ; 12: 759366, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745227

RESUMO

Recent progress in the field of organoid-based cell culture systems has enabled the use of patient-derived cells in conditions that resemble those in cancer tissue, which are better than two-dimensional (2D) cultured cell lines. In particular, organoids allow human cancer cells to be handled in conditions that resemble those in cancer tissue, resulting in more efficient establishment of cells compared with 2D cultured cell lines, thus enabling the use of multiple patient-derived cells with cells from different genetic background, in keeping with the heterogeneity of the cells. One of the most valuable points of using organoids is that human cells from either healthy or cancerous tissue can be used. Using genome editing technology such as clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein, organoid genomes can be modified to, for example, cancer-prone genomes. The normal, cancer, or genome-modified organoids can be used to evaluate whether chemicals have genotoxic or non-genotoxic carcinogenic activity by evaluating the cancer incidence, cancer progression, and cancer metastasis. In this review, the organoid technology and the accompanying technologies were summarized and the advantages of organoid-based toxicology and its application to pancreatic cancer study were discussed.

6.
Int J Cancer ; 148(1): 193-202, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-32984947

RESUMO

To generate a reliable preclinical model system exhibiting the molecular features of salivary adenoid cystic carcinoma (ACC) whose biology is still unclear due to the paucity of stable cell cultures. To develop new in vitro and in vivo models of ACC, the techniques of organoid culture and patient-derived tumor xenograft (PDX), which have attracted attention in other malignancies in recent years, were applied. Tumor specimens from surgically resected salivary ACC were proceeded for the preparation of PDX and organoid culture. The orthotopic transplantation of patient-derived or PDX-derived organoids was demonstrated into submandibular glands of NSG mice and those histology was evaluated. PDX-derived organoid cells were evaluated for the presence of MYB-mediated fusion genes and proceeded for in vitro drug sensitivity assay. Human ACC-derived organoids were successfully generated in three-dimensional culture and confirmed the ability of these cells to form tumors by orthotopic injection. Short-term organoid cell cultures from two individual ACC PDX tumors were also established that maintain the characteristic MYBL1 translocation and histological features of the original parent and PDX tumors. Finally, the establishment of drug sensitivity tests on these short-term cultured cells was confirmed using three different agents. This is the first to report an approach for the generation of human ACC-derived organoids as in vitro and in vivo cancer models, providing insights into understanding of the ACC biology and creating personalized therapy design for patients with ACC.


Assuntos
Carcinoma Adenoide Cístico/patologia , Cultura Primária de Células/métodos , Neoplasias das Glândulas Salivares/patologia , Animais , Carcinoma Adenoide Cístico/genética , Carcinoma Adenoide Cístico/cirurgia , Feminino , Humanos , Masculino , Camundongos , Proteínas de Fusão Oncogênica/genética , Organoides , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-myb/genética , Neoplasias das Glândulas Salivares/genética , Neoplasias das Glândulas Salivares/cirurgia , Glândulas Salivares/patologia , Glândulas Salivares/cirurgia , Transativadores/genética , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Sci Rep ; 10(1): 17937, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-33087763

RESUMO

Advances in organoid technology have broadened the number of target diseases and conditions in which human induced pluripotent stem cell (iPSC)-based regenerative medicine can be applied; however, mass production of organoids and the development of chemically defined, animal origin-free (CD-AOF) media and supplements are unresolved issues that hamper the clinical applicability of these approaches. CD-AOF media and supplements ensure the quality and reproducibility of culture systems by lowering lot-to-lot variations and the risk of contamination with viruses or toxins. We previously generated liver organoids from iPSCs, namely iPSC-liver buds (iPSC-LBs), by mimicking the organogenic interactions among hepatocytes, endothelial cells (ECs), and mesenchymal cells (MCs) and recently reported the mass production of iPSC-LBs derived entirely from iPSCs (all iPSC-LBs), which should facilitate their large-scale production for the treatment of liver failure. However, in previous studies we used media originating from animals for differentiation except for the maintenance of undifferentiated iPSCs. Therefore, we developed a CD-AOF medium to generate all iPSC-LBs. We first developed a CD-AOF medium for hepatocytes, ECs, and stage-matched MCs, i.e., septum transversum mesenchyme (STM), in 2D cultures. We next generated all iPSC-LBs by incubating individual cell types in ultra-low attachment micro-dimple plates. The hepatic functions of all iPSC-LBs generated using the CD-AOF medium were equivalent to those of all iPSC-LBs generated using the conventional medium both in vitro and in vivo. Furthermore, we found that this CD-AOF medium could be used in several cell culture settings. Taken together, these results demonstrate the successful development of a CD-AOF medium suitable for all iPSC-LBs. The protocol developed in this study will facilitate the clinical applicability of all iPSC-LBs in the treatment of liver diseases.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Meios de Cultura , Células-Tronco Pluripotentes Induzidas/fisiologia , Fígado/citologia , Medicina Regenerativa/métodos , Animais , Células Cultivadas , Células Endoteliais , Hepatócitos , Humanos , Células-Tronco Mesenquimais
8.
Cell Rep ; 23(6): 1620-1629, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29742420

RESUMO

Clinical transplantation of tissue fragments, including islets, faces a critical challenge because of a lack of effective strategies that ensure efficient engraftment through the timely integration of vascular networks. We recently developed a complex organoid engineering method by "self-condensation" culture based on mesenchymal cell-dependent contraction, thereby enabling dissociated heterotypic lineages including endothelial cells to self-organize in a spatiotemporal manner. Here, we report the successful adaptation of this method for generating complex tissues from diverse tissue fragments derived from various organs, including pancreatic islets. The self-condensation of human and mouse islets with endothelial cells not only promoted functionalization in culture but also massively improved post-transplant engraftment. Therapeutically, fulminant diabetic mice were more efficiently treated by a vascularized islet transplant compared with the conventional approach. Given the general limitations of post-transplant vascularization associated with 3D tissue-based therapy, our approach offers a promising means of enhancing efficacy in the context of therapeutic tissue transplantation.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/irrigação sanguínea , Neovascularização Fisiológica , Engenharia Tecidual/métodos , Animais , Endotélio/fisiologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Reperfusão
9.
Stem Cell Reports ; 10(3): 780-793, 2018 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-29429958

RESUMO

Early endoderm progenitors naturally possess robust propagating potential to develop a majority of meter-long gastrointestinal tracts and are therefore considered as a promising source for therapy. Here, we demonstrated the reproducible generation of human CDX2+ posterior gut endoderm cells (PGECs) from five induced pluripotent stem cell clones by manipulating FGF, TGF, and WNT signaling. Transcriptome analysis suggested that putative PGECs harbored an intermediate signature profile between definitive endoderm and organ-specific endoderm. We found that combinatorial EGF, VEGF, FGF2, Chir99021, and A83-01 treatments selectively amplify storable PGECs up to 1021 cell scale without any gene transduction or feeder use. PGECs, compared with induced pluripotent stem cells, showed stable differentiation propensity into multiple endodermal lineages without teratoma formation. Furthermore, transplantation of PGEC-derived liver bud organoids showed therapeutic potential against fulminant liver failure. Together, the robustly amplified PGECs may be a promising cellular source for endoderm-derived organoids in studying human development, modeling disease, and, ultimately, therapy.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Intestinos/citologia , Fígado/citologia , Organoides/citologia , Fator de Transcrição CDX2/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Endoderma/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fígado/metabolismo , Organoides/metabolismo , Transdução de Sinais/fisiologia
10.
Cell Rep ; 21(10): 2661-2670, 2017 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-29212014

RESUMO

Organoid technology provides a revolutionary paradigm toward therapy but has yet to be applied in humans, mainly because of reproducibility and scalability challenges. Here, we overcome these limitations by evolving a scalable organ bud production platform entirely from human induced pluripotent stem cells (iPSC). By conducting massive "reverse" screen experiments, we identified three progenitor populations that can effectively generate liver buds in a highly reproducible manner: hepatic endoderm, endothelium, and septum mesenchyme. Furthermore, we achieved human scalability by developing an omni-well-array culture platform for mass producing homogeneous and miniaturized liver buds on a clinically relevant large scale (>108). Vascularized and functional liver tissues generated entirely from iPSCs significantly improved subsequent hepatic functionalization potentiated by stage-matched developmental progenitor interactions, enabling functional rescue against acute liver failure via transplantation. Overall, our study provides a stringent manufacturing platform for multicellular organoid supply, thus facilitating clinical and pharmaceutical applications especially for the treatment of liver diseases through multi-industrial collaborations.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Fígado/enzimologia , Organoides/citologia , Organoides/embriologia , Células-Tronco Pluripotentes/citologia , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Fígado/citologia
11.
Nature ; 546(7659): 533-538, 2017 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-28614297

RESUMO

Conventional two-dimensional differentiation from pluripotency fails to recapitulate cell interactions occurring during organogenesis. Three-dimensional organoids generate complex organ-like tissues; however, it is unclear how heterotypic interactions affect lineage identity. Here we use single-cell RNA sequencing to reconstruct hepatocyte-like lineage progression from pluripotency in two-dimensional culture. We then derive three-dimensional liver bud organoids by reconstituting hepatic, stromal, and endothelial interactions, and deconstruct heterogeneity during liver bud development. We find that liver bud hepatoblasts diverge from the two-dimensional lineage, and express epithelial migration signatures characteristic of organ budding. We benchmark three-dimensional liver buds against fetal and adult human liver single-cell RNA sequencing data, and find a striking correspondence between the three-dimensional liver bud and fetal liver cells. We use a receptor-ligand pairing analysis and a high-throughput inhibitor assay to interrogate signalling in liver buds, and show that vascular endothelial growth factor (VEGF) crosstalk potentiates endothelial network formation and hepatoblast differentiation. Our molecular dissection reveals interlineage communication regulating organoid development, and illuminates previously inaccessible aspects of human liver development.


Assuntos
Comunicação Celular , Diferenciação Celular , Linhagem da Célula , Fígado/citologia , Fígado/embriologia , Organogênese , Técnicas de Cultura de Tecidos/métodos , Idoso , Hipóxia Celular , Movimento Celular , Endotélio/citologia , Células Epiteliais/citologia , Matriz Extracelular/metabolismo , Feminino , Feto/citologia , Hepatócitos/citologia , Humanos , Masculino , Pessoa de Meia-Idade , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Análise de Sequência de RNA , Transdução de Sinais , Análise de Célula Única , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem
12.
Sci Rep ; 8: 45751, 2017 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-28367998

RESUMO

Cancer stem cells (CSCs), a small population of cancer cells, have been considered to be the origin of cancer initiation, recurrence, and metastasis. Tumor microenvironment provides crucial signals for CSCs to maintain stem cell properties and promotes tumorigenesis. Therefore, establishment of an appropriate cell culture system to mimic the microenvironment for CSC studies is an important issue. In this study, we grew colon and hepatocellular carcinoma (HCC) cells on chitosan membranes and evaluated the tumor progression and the CSC properties. Experimental results showed that culturing cancer cells on chitosan increased cell motility, drug resistance, quiescent population, self-renewal capacity, and the expression levels of stemness and CSC marker genes, such as OCT4, NANOG, CD133, CD44, and EpCAM. Furthermore, we demonstrated that chitosan might activate canonical Wnt/ß-catenin-CD44 axis signaling in CD44positive colon cancer cells and noncanonical Wnt-STAT3 signaling in CD44negative HCC cells. In conclusion, chitosan as culture substrates activated the essential signaling of CSCs and promoted CSC properties. The chitosan culture system provides a convenient platform for the research of CSC biology and screening of anticancer drugs.


Assuntos
Carcinoma Hepatocelular/patologia , Proliferação de Células/efeitos dos fármacos , Quitosana/farmacologia , Neoplasias do Colo/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Via de Sinalização Wnt/efeitos dos fármacos , Anticolesterolemiantes/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Molécula de Adesão da Célula Epitelial/genética , Molécula de Adesão da Célula Epitelial/metabolismo , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células Tumorais Cultivadas
13.
Methods Mol Biol ; 1597: 207-216, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28361320

RESUMO

Here, we describe a protocol to develop a three-dimensional (3D) liver bud-like tissue from human iPSCs in vitro. This method mainly consists of two parts: (1) hepatic endoderm (HE) differentiation from human iPSCs in 2D culture and (2) co-culturing iPSC-HE with endothelial and mesenchymal cells. First, iPSCs were differentiated into definitive endoderm (DE) cells, and the DE cells were differentiated into HE cells, which were then co-cultured with endothelial cells and mesenchymal cells on Matrigel-coated plastic plates or micropattern plates. The cells rapidly condensed to generate 3D tissue masses. We named these iPSC liver buds (iPSC-LBs) because they resemble the developing liver bud from the perspective of gene expression, cell proliferation, and cell proportion. This liver bud culture system provides a novel approach for future clinical applications, for drug development, and as a tool for studying human development.


Assuntos
Regeneração Hepática/fisiologia , Fígado/citologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Endoderma/citologia , Células Endoteliais/citologia , Hepatócitos/citologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos
14.
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
15.
In Vivo ; 28(3): 361-5, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24815839

RESUMO

BACKGROUND: Patients diagnosed with pancreatic cancer have a high mortality rate relating to the highly malignant and refractory nature of their disease, and reputedly linked to the presence of cancerous pancreatic stem cells. These stem cells are believed to be deeply involved in distant metastasis. Therefore, the present study examined whether pancreatic cancer stem cells (CSCs) exhibit organ-specific migration patterns during metastasis. MATERIALS AND METHODS: Pancreatic cancer cells derived from primary tumors isolated from a mouse model of pancreatic cancer were subcutaneously injected into wild-type mice to form tumor allografts. Allografts were isolated and dissociated into single cells prior to cell sorting using flow cytometry. Sorted cancer cells were injected into the tail vein or spleen of recipient wild-type mice, and analyzed for engraftment three weeks post-transplantation. RESULTS: Pancreatic cancer cells metastasized either to the liver or lungs. Furthermore, we compared the number and size of metastatic foci in the liver and lungs; metastatic liver foci were larger compared with those in the lungs. CONCLUSION: Our results showed that pancreatic CSCs metastasize to distinct organs with direct access to the transplantation site via the circulation. Clarifying the interaction between pancreatic CSCs and the liver microenvironment will lead to improved prognosis and treatments for pancreatic cancer.


Assuntos
Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Xenoenxertos , Humanos , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Knockout , Metástase Neoplásica
16.
Hepatology ; 60(1): 323-33, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24497168

RESUMO

UNLABELLED: Polycomb-group (PcG) proteins play crucial roles in self-renewal of stem cells by suppressing a host of genes through histone modifications. Identification of the downstream genes of PcG proteins is essential for elucidation of the molecular mechanisms of stem cell self-renewal. However, little is known about the PcG target genes in tissue stem cells. We found that the PcG protein, Ring1B, which regulates expression of various genes through monoubiquitination of histone H2AK119, is essential for expansion of hepatic stem/progenitor cells. In mouse embryos with a conditional knockout of Ring1B, we found that the lack of Ring1B inhibited proliferation and differentiation of hepatic stem/progenitor cells and thereby inhibited hepatic organogenesis. These events were characterized by derepression of cyclin-dependent kinase inhibitors (CDKIs) Cdkn1a and Cdkn2a, known negative regulators of cell proliferation. We conducted clonal culture experiments with hepatic stem/progenitor cells to investigate the individual genetic functions of Ring1B, Cdkn1a, and Cdkn2a. The data showed that the cell-cycle inhibition caused by Ring1B depletion was reversed when Cdkn1a and Cdkn2a were suppressed simultaneously, but not when they were suppressed individually. CONCLUSION: Our results show that expansion of hepatic stem/progenitor cells requires Ring1B-mediated epigenetic silencing of Cdkn1a and Cdkn2a, demonstrating that Ring1B simultaneously regulates multiple CDKIs in tissue stem/progenitor cells.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Células-Tronco Embrionárias/citologia , Fígado/citologia , Complexo Repressor Polycomb 1/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Epigênese Genética/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fígado/embriologia , Fígado/fisiologia , Masculino , Camundongos , Camundongos Knockout , Organogênese/fisiologia , Complexo Repressor Polycomb 1/genética , Gravidez , Ubiquitina-Proteína Ligases/genética
17.
Nat Protoc ; 9(2): 396-409, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24457331

RESUMO

Generation of functional and vascularized organs from human induced pluripotent stem cells (iPSCs) will facilitate our understanding of human developmental biology and disease modeling, hopefully offering a drug-screening platform and providing novel therapies against end-stage organ failure. Here we describe a protocol for the in vitro generation of a 3D liver bud from human iPSC cultures and the monitoring of further hepatic maturation after transplantation at various ectopic sites. iPSC-derived specified hepatic cells are dissociated and suspended with endothelial cells and mesenchymal stem cells. These mixed cells are then plated onto a presolidified matrix, and they form a 3D spherical tissue mass termed a liver bud (iPSC-LB) in 1-2 d. To facilitate additional maturation, 4-d-old iPSC-LBs are transplanted in the immunodeficient mouse. Live imaging has identified functional blood perfusion into the preformed human vascular networks. Functional analyses show the appearance of multiple hepatic functions in a chronological manner in vivo.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Fígado/crescimento & desenvolvimento , Organogênese/fisiologia , Medicina Regenerativa/métodos , Transplante de Células-Tronco/métodos , Animais , Humanos , Transplante de Fígado/métodos , Camundongos , Técnicas de Cultura de Tecidos
18.
Nature ; 499(7459): 481-4, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23823721

RESUMO

A critical shortage of donor organs for treating end-stage organ failure highlights the urgent need for generating organs from human induced pluripotent stem cells (iPSCs). Despite many reports describing functional cell differentiation, no studies have succeeded in generating a three-dimensional vascularized organ such as liver. Here we show the generation of vascularized and functional human liver from human iPSCs by transplantation of liver buds created in vitro (iPSC-LBs). Specified hepatic cells (immature endodermal cells destined to track the hepatic cell fate) self-organized into three-dimensional iPSC-LBs by recapitulating organogenetic interactions between endothelial and mesenchymal cells. Immunostaining and gene-expression analyses revealed a resemblance between in vitro grown iPSC-LBs and in vivo liver buds. Human vasculatures in iPSC-LB transplants became functional by connecting to the host vessels within 48 hours. The formation of functional vasculatures stimulated the maturation of iPSC-LBs into tissue resembling the adult liver. Highly metabolic iPSC-derived tissue performed liver-specific functions such as protein production and human-specific drug metabolism without recipient liver replacement. Furthermore, mesenteric transplantation of iPSC-LBs rescued the drug-induced lethal liver failure model. To our knowledge, this is the first report demonstrating the generation of a functional human organ from pluripotent stem cells. Although efforts must ensue to translate these techniques to treatments for patients, this proof-of-concept demonstration of organ-bud transplantation provides a promising new approach to study regenerative medicine.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Fígado/irrigação sanguínea , Fígado/fisiologia , Medicina Regenerativa/métodos , Animais , Diferenciação Celular , Linhagem da Célula , Doença Hepática Induzida por Substâncias e Drogas/terapia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/transplante , Perfilação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Fígado/embriologia , Fígado/metabolismo , Falência Hepática/terapia , Transplante de Fígado , Mesoderma/citologia , Mesoderma/metabolismo , Mesoderma/transplante , Camundongos , Técnicas de Cultura de Tecidos
19.
Biomaterials ; 34(23): 5785-91, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23642538

RESUMO

Islet transplantation can induce a substantial improvement in the treatment of type 1 diabetes mellitus. However, the clinical application of islet transplantation is severely limited by the shortage of donor organs. It is thus essential to improve the engraftment rate to achieve the expected outcome in the treatment of diabetes mellitus using a limited amount of donor islets. In this manuscript, we describe the generation of ß-cell spheroids using mouse insulinoma cells (MIN6) as a model of ß-cells. We established a 3D culture system that simulates microgravity using a 3D clinostat. Using this method, we were able to produce 100 spheroids per mL of culture media. The optimization of the culture conditions in the clinostat produced spheroids with a size of approximately 250 µm, which is a size that is known to induce good graft survival after islet transplantation. The spheroids produced in the clinostat expressed several ß-cell signature genes at higher levels than the levels that were found in MIN6 cells that were cultured in a standard 2D culture dish (MIN6-2D). The transplantation of the spheroids into the portal vein of streptozotocin-induced diabetic mice ameliorates hyperglycemia, whereas the transplantation of the equivalent number of 2D-cultured cells failed to cure diabetes. These results indicate that the clinostat culture provides a new method for the reconstitution of a large number of functional ß-cell spheroids for diabetes treatment.


Assuntos
Técnicas de Cultura de Células/instrumentação , Células Secretoras de Insulina/citologia , Esferoides Celulares/citologia , Simulação de Ausência de Peso/instrumentação , Ausência de Peso , Animais , Reatores Biológicos , Linhagem Celular Tumoral , Diabetes Mellitus Experimental/terapia , Modelos Animais de Doenças , Imuno-Histoquímica , Células Secretoras de Insulina/transplante , Camundongos , Perfusão , Esferoides Celulares/transplante , Esferoides Celulares/ultraestrutura
20.
J Hepatobiliary Pancreat Sci ; 19(6): 594-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22961010

RESUMO

Enormous efforts have been made to establish pancreatic stem/progenitor cells as a source for regenerative medicine for the treatment of diabetes mellitus. In recent years, it has been recognized that the self-renewal of beta cells is the dominant process involved in postnatal beta-cell regeneration and expansion. Nevertheless, several in-vitro studies have suggested that ductal or as yet unidentified cells are candidates for pancreatic stem/progenitor cells that can differentiate into multilineage cells, including insulin(+) cells. The question remains as to whether beta cells are generated postnatally from stem/progenitor cells other than pre-existing beta cells. Furthermore, mutated pancreatic stem cells are considered to be prospective candidates for cancer stem cells or tumor-initiating cells. This review highlights recent progress in pancreatic stem/progenitor cell research.


Assuntos
Pâncreas/citologia , Pancreatopatias/cirurgia , Medicina Regenerativa/métodos , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Diferenciação Celular , Humanos , Células Secretoras de Insulina/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA