Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Rie Utoh is active.

Publication


Featured researches published by Rie Utoh.


American Journal of Pathology | 2004

Near Completely Humanized Liver in Mice Shows Human-Type Metabolic Responses to Drugs

Chise Tateno; Yasumi Yoshizane; Naomi Saito; Miho Kataoka; Rie Utoh; Chihiro Yamasaki; Asato Tachibana; Yoshinori Soeno; Kinji Asahina; Hiroshi Hino; Toshimasa Asahara; Tsuyoshi Yokoi; Toshinori Furukawa; Katsutoshi Yoshizato

Human hepatocytes were transplanted into urokinase-type plasminogen activator-transgenic SCID mice (uPA/SCID mice), which are immunodeficient and undergo liver failure. The transplanted cells were characterized in terms of their in vivo growth potential and functions. The human hepatocytes progressively repopulated the murine host liver. However, the recipients died when the replacement index (RI) of the human hepatocytes exceeded 50%. The hosts (chimeric mice) survived at RI >50% when treated with a drug that has anti-human complement factor activity, and these mice developed livers with RI values as high as 96%. In total, 36 chimeric mice were generated, and the rate of successful engraftment was as high as 92%. The yield of chimeric mice with RI >70% was 32%. The human hepatocytes in the murine host liver expressed mRNAs for a variety of human cytochrome P450 (hCYP) subtypes, in a manner that was similar to the donor liver. The mRNAs for hCYP3A4 and hCYP1A1/2 were induced in the liver in a CYP type-specific manner when the mice were treated with rifampicin and 3-methylcholanthrene, respectively. These results indicate that human hepatocytes that propagate in mice retain their normal pharmacological responses. We conclude that the chimeric mouse developed in the present study is a useful model for assessing the functions and pharmacological responses of human hepatocytes.


Biomaterials | 2012

Controlled formation of heterotypic hepatic micro-organoids in anisotropic hydrogel microfibers for long-term preservation of liver-specific functions

Rie Utoh; Kazuo Ohashi; Kohei Tatsumi; Masayuki Yamato; Teruo Okano; Minoru Seki

We have developed a hydrogel-based cell cultivation platform for forming 3D restiform hepatic micro-organoids consisting of primary rat hepatocytes and feeder cells (Swiss 3T3 cells). Sodium alginate solutions containing hepatocytes/3T3 cells were continuously introduced into a microfluidic channel to produce cell-incorporating anisotropic Ba-alginate hydrogel microfibers, where hepatocytes at the center were closely sandwiched by 3T3 cells. Hydrogel fiber-based cultivation under high oxygen tension enabled the formation of heterotypic micro-organoids with a length of up to 1 mm and a diameter of ∼50 μm, mimicking the hepatic cord structures found in the liver, while maintaining a high hepatocyte viability (∼80%) over 30 days. Long-term observation of up to 90 days revealed a significant enhancement of hepatic functions because of heterotypic and homotypic cell-cell interactions, including albumin secretion and urea synthesis as well as expression of hepatocyte-specific genes, compared with conventional monolayer culture and single cultivation in the hydrogel fibers. The encapsulated hepatic constructs were recovered as scaffold-free micro-organoids by enzymatically digesting the hydrogel matrices using alginate lyase. This technique for creating heterotypic micro-organoids with precisely ordered multiple cell types will be useful for the development of a new liver tissue engineering approach and may be applicable to the fabrication of extracorporeal bioartificial liver (BAL) devices and assessment tools for drug development and testing.


Biomaterials | 2009

Bioengineering of a functional sheet of islet cells for the treatment of diabetes mellitus

Hirofumi Shimizu; Kazuo Ohashi; Rie Utoh; Kazuya Ise; Mitsukazu Gotoh; Masayuki Yamato; Teruo Okano

The present study was designed to establish a novel tissue engineering approach for diabetes mellitus (DM) by fabricating a tissue sheet composed of pancreatic islet cells for in vivo transplantation. Pancreatic islet cell suspensions were obtained from Lewis rats, and plated onto temperature-responsive culture dishes coated with extracellular matrix (ECM) proteins. After the cells reached confluency, islet cells cultured on laminin-5 coated dishes were successfully harvested as a uniformly spread tissue sheet by lowering the culture temperature to 20 degrees C for 20 min. The functional activity of the islet cell sheets was confirmed by histological examination and Insulin secretion assay prior to in vivo transplantation. Histological examination revealed that the harvested islet cell sheet was comprised of insulin- (76%) and glucagon- (19%) positive cells, respectively. In vivo functionality of the islet cell sheet was maintained even 7 days after transplantation into the subcutaneous space of Lewis rats. The present study describes an approach to generate a functional sheet of pancreatic islet cells on laminin-5 coated temperature-responsive dishes, which can be subsequently transplanted in vivo. This study serves as the foundation for the creation of a novel cell-based therapy for DM to provide patients an alternative method.


Journal of Controlled Release | 2014

Cell sheet approach for tissue engineering and regenerative medicine.

Katsuhisa Matsuura; Rie Utoh; Kenichi Nagase; Teruo Okano

After the biotech medicine era, regenerative medicine is expected to be an advanced medicine that is capable of curing patients with difficult-to-treat diseases and physically impaired function. Our original scaffold-free cell sheet-based tissue engineering technology enables transplanted cells to be engrafted for a long time, while fully maintaining their viability. This technology has already been applied to various diseases in the clinical setting, including the cornea, esophagus, heart, periodontal ligament, and cartilage using autologous cells. Transplanted cell sheets not only replace the injured tissue and compensate for impaired function, but also deliver growth factors and cytokines in a spatiotemporal manner over a prolonged period, which leads to promotion of tissue repair. Moreover, the integration of stem cell biology and cell sheet technology with sufficient vascularization opens possibilities for fabrication of human three-dimensional vascularized dense and intact tissue grafts for regenerative medicine to parenchymal organs.


Biomaterials | 2012

Preserved liver-specific functions of hepatocytes in 3D co-culture with endothelial cell sheets.

Kyungsook Kim; Kazuo Ohashi; Rie Utoh; Kyoko Kano; Teruo Okano

Hepatocyte-based tissue engineering is an attractive method that is being developed to treat liver diseases. However, this method is limited by the relatively short lifespan of cultured hepatocytes to maintain their normal function. For this reason, the present study was designed to develop a cell sheet-based hepatocyte co-culture system that enables cultured hepatocytes to preserve their functions for a longer period of time. To achieve this goal, a monolayer cell sheet composed of endothelial cells (EC) was placed on top of a monolayer of hepatocytes (Hep). In this hybrid cell sheet format, histological examination revealed that bile canaliculi networks were formed and well developed among the hepatocytes in the layered Hep-EC sheet group. The albumin secretion level was highly preserved at least for 28 days in the hybrid Hep-EC sheet, whereas the monolayer of hepatocytes exhibited a markedly reduced time course of secretion. The expression levels of hepatocyte-specific genes including albumin, hepatocyte nucleus factor 4 (HNF 4), multidrug resistance-associated protein 2 (MRP 2), and claudin-3 were significantly higher in the Hep-EC sheet compared to the Hep sheet alone after 14-days in culture. In all, this culture system provides a valuable technology to prolong hepatocyte functionality and enable more efficient development of liver tissue engineering approaches to create liver-targeted regenerative therapies.


Biochemical and Biophysical Research Communications | 2013

Tissue factor triggers procoagulation in transplanted mesenchymal stem cells leading to thromboembolism

Kohei Tatsumi; Kazuo Ohashi; Yoshinori Matsubara; Ayako Kohori; Takahiro Ohno; Hiroshi Kakidachi; Akihiro Horii; Kazuko Kanegae; Rie Utoh; Takanori Iwata; Teruo Okano

Mesenchymal stem cells (MSCs) have shown extreme clinical promise as a therapeutic regenerative system in the treatment of numerous types of diseases. A recent report, however, documented lethal pulmonary thromboembolism in a patient following the administration of adipose-derived MSCs (ADSCs). In our study, we designed experiments to examine the role of tissue factor (TF), which is highly expressed at the level of mRNA and localized to the cell surface of cultured MSCs, as a triggering factor in the procoagulative cascade activated by infused MSCs. A high mortality rate of ~85% in mice was documented following intravenous infusion of mouse ADSCs within 24 h due to the observation of pulmonary embolism. Rotation thromboelastometry and plasma clotting assay demonstrated significant procoagulation by the cultured mouse ADSCs, and preconditioning of ADSCs with an anti-TF antibody or usage of factor VII deficient plasma in the assay successfully suppressed the procoagulant properties. These properties were also observed in human ADSCs, and could be suppressed by recombinant human thrombomodulin. In uncultured mouse adipose-derived cells (ADCs), the TF-triggered procoagulant activity was not observed and all mice infused with these uncultured ADCs survived after 24 h. This clearly demonstrated that the process of culturing cells plays a critical role in sensitizing these cells as a procoagulator through the induction of TF expression. Our results would recommend that clinical applications of MSCs to inhibit TF activity using anti-coagulant agents or genetic approaches to maximize clinical benefit to the patients.


Transplantation | 2011

Reversal of diabetes by the creation of neo-islet tissues into a subcutaneous site using islet cell sheets.

Takahiro Saito; Kazuo Ohashi; Rie Utoh; Hirofumi Shimizu; Kazuya Ise; Hiroyuki Suzuki; Masayuki Yamato; Teruo Okano; Mitsukazu Gotoh

Background. There remains a paucity of therapeutic approaches to completely treat diabetes mellitus. This study was designed to develop a dispersed islet cell-based tissue engineering approach to engineer functional neo-islet tissues in the absence of traditional bioabsorbable scaffold matrices. Methods. Specialized coated plastic dishes were prepared by covalently immobilizing a temperature-responsive polymer, poly(N-isopropylacrylamide), onto the plastic followed by coating with laminin-5. Dispersed rat islet cells were plated on the laminin-5-poly(N-isopropylacrylamide) dishes. After 2 days of culturing, islet cells were harvested as a uniformly connected tissue sheet by lowering the culture temperature from 37°C to 20°C for 30 min. Two harvested islet cell sheets were transplanted into the subcutaneous space of streptozotocin-induced diabetic severe combined immunodeficiency (SCID) mice to engineer neo-islet tissues in vivo. Therapeutic effects were investigated after the tissue engineering procedures. Results. In all of the diabetic SCID mice transplanted with the islet sheets, serum hyperglycemia was successfully reverted to a steady normoglycemic level. The recipient SCID mice demonstrated positive for serum rat C-peptide and elevated serum insulin levels. Moreover, the islet cell sheet-transplanted SCID mice demonstrated rapid glucose clearance and return of serum glucose levels after intraperitoneal glucose tolerance test. Histological examination revealed that the transplanted islet cell sheets were structured as flat clusters of islet tissues in which an active vascular network manifested within and surrounding the newly formed tissues. Conclusions. This study describes a new proof-of-concept therapeutic approach to engineer functional neo-islet tissues for the treatment of type 1 diabetes mellitus.


Development Growth & Differentiation | 2002

Lineage of anuran epidermal basal cells and their differentiation potential in relation to metamorphic skin remodeling

Ken-ichi Suzuki; Rie Utoh; Kaori Kotani; Masanobu Obara; Katsutoshi Yoshizato

The anuran remodels the larval epidermis into the adult one during metamorphosis. Larval and adult epidermal cells of the bullfrog were characterized by determining the presence of huge cytoplasmic keratin bundles and the expression profiles of specific marker genes, namely colα1 (collagen α1 (I)), rlk (larval keratin) and rak (adult keratin). We identified four types of epidermal basal cells: (i) basal skein cells that have keratin bundles and express colα1 and rlk; (ii) rak+‐basal skein cells that have keratin bundles and express colα1, rlk, and rak; (iii) larval basal cells that express rlk and rak; and (iv) adult basal cells that express rak. These traits suggested that these basal cells are on the same lineage in which basal skein cells are the original progenitor cells that consecutively differentiate into rak+‐basal skein cells into larval basal cells, and finally into adult basal cells. To directly verify the differentiation potential of larval basal cells into adult ones, the mono‐layered epidermis composed of larval basal cells was cultured in the presence of aldosterone and thyroid hormone. In this culture, larval basal cells differentiated into adult basal cells that reconstituted the adult epidermis. Thus, it was concluded that larval basal cells are the direct progenitor cells of the adult epidermal stem cells.


Journal of Bioscience and Bioengineering | 2013

Preparation of stripe-patterned heterogeneous hydrogel sheets using microfluidic devices for high-density coculture of hepatocytes and fibroblasts

Aoi Kobayashi; Kenta Yamakoshi; Yuya Yajima; Rie Utoh; Minoru Seki

Here we demonstrate the production of stripe-patterned heterogeneous hydrogel sheets for the high-density 3D coculture of multiple cell types, by using microchannel-combined micronozzle devices. The prepared hydrogel sheet, composed of multiple regions with varying physical stiffness, regulates the direction of proliferation of encapsulated cells and enables the formation of arrays of rod-like heterotypic organoids inside the hydrogel matrix. We successfully prepared stripe-patterned hydrogel sheets with a uniform thickness of ~100 μm and a width of several millimeters. Hepatoma cells (HepG2) and fibroblasts (Swiss 3T3) were embedded inside the hydrogel matrix and cocultured, to form heterotypic micro-organoids mimicking in vivo hepatic cord structures. The upregulation of hepatic functions by the 3D coculture was confirmed by analyzing liver-specific functions. The presented heterogeneous hydrogel sheet could be useful, as it provides relatively large, but precisely-controlled, 3-dimensional microenvironments for the high-density coculture of multiple types of cells.


Journal of Endocrinology | 2007

GH enhances proliferation of human hepatocytes grafted into immunodeficient mice with damaged liver

Norio Masumoto; Chise Tateno; Asato Tachibana; Rie Utoh; Yoshio Morikawa; Takashi Shimada; Hiroyuki Momisako; Toshiyuki Itamoto; Toshimasa Asahara; Katsutoshi Yoshizato

We investigated effects of human (h) GH on the proliferation of h-hepatocytes that had been engrafted in the liver of albumin enhancer/promoter driven-urokinase plasminogen activator transgenic/severe combined immunodeficiency disease (uPA/SCID) mice (chimeric mice). The h-hepatocytes therein were considered to be deficient in GH, because hGH receptor (hGHR) is unresponsive to mouse GH. Actually, hIGF-1 was undetectable in chimeric mouse sera. The uPA/SCID mice were transplanted with h-hepatocytes from a 6-year (6Y)-old donor, and were injected with recombinant hGH (rhGH). rhGH stimulated the repopulation speed of h-hepatocytes; and up-regulated hIGF-1, human signal transducers and activators of transcription (hSTAT) 3, and cell cycle regulatory genes such as human forkhead box M1, human cell division cycle 25A, and human cyclin D1. To confirm the reproducibility of these effects of rhGH, similar experiments were run using h-hepatocytes from a 46-year (46Y)-old donor. rhGH similarly enhanced their repopulation speed and up-regulated the expression of the above-tested genes, especially hIGF-1 and hSTAT1. The extent of the enhancement by rhGH was much less than that in 6Y-hepatocyte-chimeric mice most probably due to the difference in GHR expression levels between the two donors. In conclusion, this study clearly demonstrated that rhGH stimulates the proliferation of h-hepatocytes in vivo.

Collaboration


Dive into the Rie Utoh's collaboration.

Top Co-Authors

Avatar

Teruo Okano

National Institute for Materials Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mitsukazu Gotoh

Fukushima Medical University

View shared research outputs
Top Co-Authors

Avatar

Kazuya Ise

Fukushima Medical University

View shared research outputs
Top Co-Authors

Avatar

Hirofumi Shimizu

Fukushima Medical University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge