Network


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

Hotspot


Dive into the research topics where Yuichiro Higuchi is active.

Publication


Featured researches published by Yuichiro Higuchi.


Molecular Brain | 2016

Pathological classification of human iPSC-derived neural stem/progenitor cells towards safety assessment of transplantation therapy for CNS diseases

Keiko Sugai; Ryuji Fukuzawa; Tomoko Shofuda; Hayato Fukusumi; Soya Kawabata; Yuichiro Nishiyama; Yuichiro Higuchi; Kenji Kawai; Miho Isoda; Daisuke Kanematsu; Tomoko Hashimoto-Tamaoki; Jun Kohyama; Akio Iwanami; Hiroshi Suemizu; Eiji Ikeda; Morio Matsumoto; Yonehiro Kanemura; Masaya Nakamura; Hideyuki Okano

The risk of tumorigenicity is a hurdle for regenerative medicine using induced pluripotent stem cells (iPSCs). Although teratoma formation is readily distinguishable, the malignant transformation of iPSC derivatives has not been clearly defined due to insufficient analysis of histology and phenotype. In the present study, we evaluated the histology of neural stem/progenitor cells (NSPCs) generated from integration-free human peripheral blood mononuclear cell (PBMC)-derived iPSCs (iPSC-NSPCs) following transplantation into central nervous system (CNS) of immunodeficient mice. We found that transplanted iPSC-NSPCs produced differentiation patterns resembling those in embryonic CNS development, and that the microenvironment of the final site of migration affected their maturational stage. Genomic instability of iPSCs correlated with increased proliferation of transplants, although no carcinogenesis was evident. The histological classifications presented here may provide cues for addressing potential safety issues confronting regenerative medicine involving iPSCs.


Xenobiotica | 2014

The human hepatic cell line HepaRG as a possible cell source for the generation of humanized liver TK-NOG mice

Yuichiro Higuchi; Kenji Kawai; Hiroshi Yamazaki; Masato Nakamura; Françoise Bree; Christiane Guguen-Guillouzo; Hiroshi Suemizu

Abstract 1. Humanized-liver mice, in which the liver has been repopulated with human hepatocytes, have been used to study aspects of human liver physiology such as drug metabolism, toxicology and hepatitis infection. However, the procurement of human hepatocytes is a major problem in producing humanized-liver mice because of the finite nature of the patient-derived resource. 2. In order to overcome this limitation, the human hepatic cell line HepaRG® were evaluated as promising donor cells for liver reconstitution in the TK-NOG mouse model. 3. We demonstrate that, in vivo, transplanted confluent culture or differentiated HepaRG® cells proliferated and differentiated toward both hepatocyte-like and biliary-like cells within the recipient liver. In contrast, proliferative HepaRG® cells could engraft TK-NOG mouse liver but could differentiate only toward biliary-like cells. The differentiation to hepatocyte-like cells was characterized by the detection of human albumin in the recipient mouse serum and was confirmed by immunohistochemical staining for human leukocyte antigen, human albumin, cytochrome P450 3A4, and multidrug resistance-associated protein 2. Biliary-like cells were characterized by positive staining for cytokeratin-19. 4. These results indicated that the differentiated HepaRG® cells are a possible cell source for generating humanized-liver mice, which are a useful model for in vivo studies of liver physiology.


Molecular Brain | 2016

In vitro characterization of neurite extension using induced pluripotent stem cells derived from lissencephaly patients with TUBA1A missense mutations

Yohei Bamba; Tomoko Shofuda; Mitsuhiro Kato; Ritsuko K. Pooh; Yoko Tateishi; Jun-ichi Takanashi; Hidetsuna Utsunomiya; Miho Sumida; Daisuke Kanematsu; Hiroshi Suemizu; Yuichiro Higuchi; Wado Akamatsu; Denis Gallagher; Freda D. Miller; Mami Yamasaki; Yonehiro Kanemura; Hideyuki Okano

BackgroundLissencephaly, or smooth brain, is a severe congenital brain malformation that is thought to be associated with impaired neuronal migration during corticogenesis. However, the exact etiology of lissencephaly in humans remains unknown. Research on congenital diseases is limited by the shortage of clinically derived resources, especially for rare pediatric diseases. The research on lissencephaly is further limited because gyration in humans is more evolved than that in model animals such as mice. To overcome these limitations, we generated induced pluripotent stem cells (iPSCs) from the umbilical cord and peripheral blood of two lissencephaly patients with different clinical severities carrying alpha tubulin (TUBA1A) missense mutations (Patient A, p.N329S; Patient B, p.R264C).ResultsNeural progenitor cells were generated from these iPSCs (iPSC-NPCs) using SMAD signaling inhibitors. These iPSC-NPCs expressed TUBA1A at much higher levels than undifferentiated iPSCs and, like fetal NPCs, readily differentiated into neurons. Using these lissencephaly iPSC-NPCs, we showed that the neurons derived from the iPSCs obtained from Patient A but not those obtained from Patient B showed abnormal neurite extension, which correlated with the pathological severity in the brains of the patients.ConclusionWe established iPSCs derived from lissencephaly patients and successfully modeled one aspect of the pathogenesis of lissencephaly in vitro using iPSC-NPCs and iPSC-derived neurons. The iPSCs from patients with brain malformation diseases helped us understand the mechanism underlying rare diseases and human corticogenesis without the use of postmortem brains.


PLOS ONE | 2013

A versatile technique for the in vivo imaging of human tumor xenografts using near-infrared fluorochrome-conjugated macromolecule probes.

Hiroshi Suemizu; Kenji Kawai; Yuichiro Higuchi; Haruo Hashimoto; Tomoyuki Ogura; Toshio Itoh; Erika Sasaki; Masato Nakamura

Here, we present a versatile method for detecting human tumor xenografts in vivo, based on the enhanced permeability and retention (EPR) effect, using near-infrared (NIR) fluorochrome-conjugated macromolecule probes. Bovine serum albumin (BSA) and two immunoglobulins—an anti-human leukocyte antigen (HLA) monoclonal antibody and isotype control IgG2a—were labeled with XenoLight CF770 fluorochrome and used as NIR-conjugated macromolecule probes to study whole-body imaging in a variety of xenotransplantation mouse models. NIR fluorescent signals were observed in subcutaneously transplanted BxPC-3 (human pancreatic cancer) cells and HCT 116 (colorectal cancer) cells within 24 h of NIR-macromolecule probe injection, but the signal from the fluorochrome itself or from the NIR-conjugated small molecule (glycine) injection was not observed. The accuracy of tumor targeting was confirmed by the localization of the NIR-conjugated immunoglobulin within the T-HCT 116 xenograft (in which the orange-red fluorescent protein tdTomato was stably expressed by HCT 116 cells) in the subcutaneous transplantation model. However, there was no significant difference in the NIR signal intensity of the region of interest between the anti-HLA antibody group and the isotype control group in the subcutaneous transplantation model. Therefore, the antibody accumulation within the tumor in vivo is based on the EPR effect. The liver metastasis generated by an intrasplenic injection of T-HCT 116 cells was clearly visualized by the NIR-conjugated anti-HLA probe but not by the orange-red fluorescent signal derived from the tdTomato reporter. This result demonstrated the superiority of the NIR probes over the tdTomato reporter protein at enhancing tissue penetration. In another xenograft model, patient-derived xenografts (PDX) of LC11-JCK (human non-small cell lung cancer) were successfully visualized using the NIR-conjugated macromolecule probe without any genetic modification. These results suggested that NIR-conjugated macromolecule, preferably, anti-HLA antibody probe is a valuable tool for the detection of human tumors in experimental metastasis models using whole-body imaging.


Liver Transplantation | 2014

Hepatocytes buried in the cirrhotic livers of patients with biliary atresia proliferate and function in the livers of urokinase-type plasminogen activator-NOG mice.

Hiroshi Suemizu; Kazuaki Nakamura; Kenji Kawai; Yuichiro Higuchi; Mureo Kasahara; Junichiro Fujimoto; Akito Tanoue; Masato Nakamura

The pathogenesis of biliary atresia (BA), which leads to end‐stage cirrhosis in most patients, has been thought to inflame and obstruct the intrahepatic and extrahepatic bile ducts. BA is not believed to be caused by abnormalities in parenchymal hepatocytes. However, there has been no report of a detailed analysis of hepatocytes buried in the cirrhotic livers of patients with BA. Therefore, we evaluated the proliferative potential of these hepatocytes in immunodeficient, liver‐injured mice [the urokinase‐type plasminogen activator (uPA) transgenic NOD/Shi‐scid IL2rγnull (NOG); uPA‐NOG strain]. We succeeded in isolating viable hepatocytes from the livers of patients with BA who had various degrees of fibrosis. The isolated hepatocytes were intrasplenically transplanted into the livers of uPA‐NOG mice. The hepatocytes of only 3 of the 9 BA patients secreted detectable amounts of human albumin in sera when they were transplanted into mice. However, human leukocyte antigen–positive hepatocyte colonies were detected in 7 of the 9 mice with hepatocyte transplants from patients with BA. We demonstrated that hepatocytes buried in the cirrhotic livers of patients with BA retained their proliferative potential. A liver that was reconstituted with hepatocytes from patients with BA was shown to be a functioning human liver with a drug‐metabolizing enzyme gene expression pattern that was representative of mature human liver and biliary function, as ascertained by fluorescent dye excretion into the bile canaliculi. These results imply that removing the primary etiology via an earlier portoenterostomy may increase the quantity of functionally intact hepatocytes remaining in a cirrhotic liver and may contribute to improved outcomes. Liver Transpl 20:1127–1137, 2014.


Experimental Animals | 2014

A Novel Enhanced Green FluorescentProtein-Expressing NOG Mouse for Analyzingthe Microenvironment of Xenograft Tissues

Yuichiro Higuchi; Kenji Kawai; Masafumi Yamamoto; Miyuki Kuronuma; Yasuhiko Ando; Ikumi Katano; Masato Nakamura; Hiroshi Suemizu

The interaction between transplanted cells and host tissues is important for the growth and maintenance of transplanted cells. To analyze the mechanisms of these interactions, a systemic fluorescent protein-expressing mouse is a useful recipient. In this study, we generated a novel NOG strain, which strongly expresses enhanced green fluorescent protein (EGFP; PgkEGFP-NOG), especially in the liver, kidney, gastrointestinal tract, and testis. Because the host tissues expressed EGFP, xenotransplanted human cancer cells were clearly identified as EGFP-negative colonies in PgkEGFP-NOG mice. Immunohistochemical analysis revealed that EGFP-expressing stromal tissues formed a complicated tumor microenvironment within xenograft tissues. Moreover, a similar microenvironment was observed in human iPS cell-derived teratomas. Collectively, these results indicated that a suitable microenvironment is essential for the growth and maintenance of xenotransplanted cells and that PgkEGFP-NOG mice represent a useful animal model for analyzing the mechanisms of microenvironment formation.


Xenobiotica | 2018

Expression and inducibility of cytochrome P450s in human hepatocytes isolated from chimeric mice with humanised livers

Shotaro Uehara; Yuichiro Higuchi; Nao Yoneda; Hiroshi Yamazaki; Hiroshi Suemizu

Abstract The evaluation of drug-mediated cytochrome P450 (P450) induction using human hepatocytes is important for predicting drug interactions. In this study, we prepared hepatocytes from chimeric mice with humanised livers (Hu-Liver mice) and evaluated the expression and inducibility of P450s in these hepatocytes. Up to 95% of the Hu-Liver cells stained positive for human leukocyte antigen and the mean viability exceeded 85% (n = 10). Monolayer-cultured Hu-Liver cells displayed a similar morphology to cultures of the corresponding human hepatocytes used as transplantation donors. The mRNA expression levels in Hu-Liver cells of 16 P450 forms belonging to P450 subfamilies 1–4 correlated well with the expression levels of the same enzymes in human hepatocytes. The variations in individual P450 mRNA levels between Hu-Liver cells and the corresponding human hepatocytes were within five-fold for 13 P450 forms. The production of 6β-hydroxytestosterone in Hu-Liver cells was significantly increased (p < .05) following treatment with the CYP3A inducer, rifampicin. Hu-Liver cells have characteristics similar to those of human hepatocytes in terms of mRNA expression levels and the inducibility of the various P450 forms. Thus, Hu-Liver cells can potentially be used for in vitro drug-mediated induction assays of human hepatic P450s.


Asian Spine Journal | 2017

Generation of induced pluripotent stem cells and neural stem/progenitor cells from newborns with Spina bifida aperta

Yohei Bamba; Masahiro Nonaka; Natsu Sasaki; Tomoko Shofuda; Daisuke Kanematsu; Hiroshi Suemizu; Yuichiro Higuchi; Ritsuko K. Pooh; Yonehiro Kanemura; Hideyuki Okano; Mami Yamasaki

Study Design We established induced pluripotent stem cells (iPSCs) and neural stem/progenitor cells (NSPCs) from three newborns with spina bifida aperta (SBa) using clinically practical methods. Purpose We aimed to develop stem cell lines derived from newborns with SBa for future therapeutic use. Overview of Literature SBa is a common congenital spinal cord abnormality that causes defects in neurological and urological functions. Stem cell transplantation therapies are predicted to provide beneficial effects for patients with SBa. However, the availability of appropriate cell sources is inadequate for clinical use because of their limited accessibility and expandability, as well as ethical issues. Methods Fibroblast cultures were established from small fragments of skin obtained from newborns with SBa during SBa repair surgery. The cultured cells were transfected with episomal plasmid vectors encoding reprogramming factors necessary for generating iPSCs. These cells were then differentiated into NSPCs by chemical compound treatment, and NSPCs were expanded using neurosphere technology. Results We successfully generated iPSC lines from the neonatal dermal fibroblasts of three newborns with SBa. We confirmed that these lines exhibited the characteristics of human pluripotent stem cells. We successfully generated NSPCs from all SBa newborn-derived iPSCs with a combination of neural induction and neurosphere technology. Conclusions We successfully generated iPSCs and iPSC-NSPCs from surgical samples obtained from newborns with SBa with the goal of future clinical use in patients with SBa.


Cellular and Molecular Biology | 2015

Forced expression of PDX-1 gene makes hepatoma cells to acquire glucose-responsive insulin secretion while maintaining hepatic characteristic.

Haruo Hashimoto; Yuichiro Higuchi; Kenji Kawai


Experimental Animals | 2011

Vinyl isolator breeding induces insulin resistance in C57BL/6JJcl mice.

Haruo Hashimoto; Susumu Ebukuro; Ryoko Nozu; Masami Ueno; Toshiro Arai; Kenji Kawai; Hiroshi Hirata; Maya Ogawa; Takuma Mizusawa; Kuniyasu Imai; Yuichiro Higuchi; Hiroshi Suemizu; Mamoru Ito; Muneo Saito; Kyoji Hioki

Collaboration


Dive into the Yuichiro Higuchi's collaboration.

Top Co-Authors

Avatar

Hiroshi Suemizu

Central Institute for Experimental Animals

View shared research outputs
Top Co-Authors

Avatar

Kenji Kawai

Central Institute for Experimental Animals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tomoko Shofuda

National Institute of Advanced Industrial Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Yonehiro Kanemura

National Institute of Advanced Industrial Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Haruo Hashimoto

Central Institute for Experimental Animals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiroshi Yamazaki

Showa Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Masahiro Nonaka

Kansai Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge