Network


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

Hotspot


Dive into the research topics where Takeshi Yuasa is active.

Publication


Featured researches published by Takeshi Yuasa.


Cell Transplantation | 2008

Reestablishment of Microenvironment is Necessary to Maintain In Vitro and In Vivo Human Islet Function

Nalu Navarro-Alvarez; Jorge David Rivas-Carrillo; Alejandro Soto-Gutierrez; Takeshi Yuasa; Teru Okitsu; Hirofumi Noguchi; Shinichi Matsumoto; Jiro Takei; Noriaki Tanaka; Naoya Kobayashi

Islet transplantation is associated with an elevated rate of early graft failure. The isolation process leads to structural and functional abnormalities. The reestablishment of the cell–matrix relationship is important to modulate the survival and function of islets. Thus, we evaluated the effect of human fibronectin (hFN) and self-assembling peptide nanofiber (SAPNF) in the ability to support islet function in vitro and after transplantation into streptozotocin (STZ)-induced diabetic severe combined immunodeficiency (SCID) mice. Human isolated islets were cultured with hFN or SAPNF for 7 days. Their ability to maintain insulin production/glucose responsiveness over time was evaluated. Islets embedded in hFN, SAPNF, or alone were transplanted into STZ-induced diabetic SCID mice. Islet grafts were removed after 14 days to evaluate insulin content, insulin expression, and apoptosis. SAPNF-entrapped islets maintained satisfactory morphology/viability and capability of glucose-dependent insulin secretion for over 7 days, whereas islets cultured in hFN underwent widespread deterioration. In vivo grafts containing human islets in SAPNF showed remarkably higher insulin content and expression when compared with human islets in hFn or alone. RT-PCR revealed lower caspase-3 expression in SAPNF islets grafts. These studies indicate that the reestablishment of the cell–matrix interactions by a synthetic matrix in the immediate postisolation period is a useful tool to maintain islet functions in vitro and in vivo.


Cell Transplantation | 2010

Hepatic differentiation of mouse iPS cells in vitro.

Masaya Iwamuro; Toshiyuki Komaki; Yasuhiro Kubota; Masayuki Seita; Hironobu Kawamoto; Takeshi Yuasa; Javed M. Shahid; Reham A. R. A. Hassan; Wael A. R. A. Hassan; Shuhei Nakaji; Yuriko Nishikawa; Eisaku Kondo; Kazuhide Yamamoto; Ira J. Fox; Naoya Kobayashi

Induced pluripotent stem (iPS) cells are pluripotent and are able to unlimitedly proliferate in vitro. This technical breakthrough in creating iPS cells from somatic cells has noteworthy implications for overcoming the immunological rejection and the ethical issues associated with the derivation of embryonic stem cells from embryos. In the current work, we present an efficient hepatic differentiation of mouse iPS cells in vitro. iPS cells were cultured free floating to induce the formation of embryoid bodies (EB) for 5 days. EB were transferred to a gelatin-coated plate and treated with 100 ng/ml activin A and 100 ng/ml basic fibroblast growth factor (bFGF) for 3 days to induce definitive endoderm. Cells were further cultured for 8 days with 100 ng/ml hepatocyte growth factor (HGF) to generate hepatocytes. Characterization was performed by RT-PCR assay. Functional analysis for albumin secretion and ammonia removal was also carried out. iPS cell-derived hepatocyte-like cells (iPS-Heps) were obtained at the end of the differentiation program. Expression levels of a gestational hepatocyte gene and lineage-specific hepatic genes intensified in iPS-Heps. The production of albumin increased in a time-dependent manner. iPS-Heps were capable of metabolizing ammonia. We present here instant hepatic differentiation of mouse iPS cells using combined 3-day treatments of activin A and bFGF with subsequent 8-day HGF. Our study will be an important step to generate hepatocytes from human iPS cells as a new source for liver-targeted cell therapies.


Cell Transplantation | 2006

Self-Assembling Peptide Nanofiber as a Novel Culture System for Isolated Porcine Hepatocytes

Nalu Navarro-Alvarez; Alejandro Soto-Gutierrez; Jorge David Rivas-Carrillo; Yong Chen; Tsuyoshi Yamamoto; Takeshi Yuasa; Haruo Misawa; Jiro Takei; Noriaki Tanaka; Naoya Kobayashi

Freshly isolated porcine hepatocytes are a very attractive cell source in the cell-based therapies to treat liver failure because of unlimited availability. However, due to the loss of hepatocyte functions in vitro, there is a need to develop a functional culture system to keep the cells metabolically active. Here we compared the effect of a self-assembling peptide nanofiber (SAPNF) as an extracellular matrix (ECM) with collagen type I on hepatocyte metabolic and secretion activities following hepatocyte isolation. Isolated porcine hepatocytes were cultured in SAPNF and collagen type I. Morphological assessment at different time points was performed by using SEM and phase contrast microscope. Metabolic and secretion activities were comparatively performed in the groups, by means of ammonia, lidocaine, and diazepam as well as albumin. Hepatocytes cultured on SAPNF revealed a three-dimensional spheroidal formation, thus maintaining cell differentiation status during 2 weeks of culture. On the other hand, hepatocytes in collagen revealed a spread shape, and by day 14 no hepatocyte-like cells were observed, but cells with long shape were present, thus revealing a degree of dedifferentiation in collagen culture. Hepatocytes in SAPNF were capable of drug-metabolizing activities and albumin secretion in higher ratio than those cultured on collagen. The present work clearly demonstrates the usefulness of SAPNF for maintaining differentiated functions of porcine hepatocytes in culture.


Cell Transplantation | 2010

Characteristics of CD133(+) human colon cancer SW620 cells.

Hironobu Kawamoto; Takeshi Yuasa; Yasuhiro Kubota; Masayuki Seita; Hiromi Sasamoto; Javed M. Shahid; Takahiro Hayashi; Hiroyuki Nakahara; Reham A. R. A. Hassan; Masaya Iwamuro; Eisaku Kondo; Shuhei Nakaji; Noriaki Tanaka; Naoya Kobayashi

Worldwide, colorectal cancer is the third most common type of cancer affecting both sexes. It has been proposed that a small subset of cancer cells (cancer stem cells) within each tumor is able to initiate tumor growth. In 2007, two research groups simultaneously identified a colon cancer stem cell population in human tumors by the use of CD133 expression. In the present study, we used a human colon cancer cell line, SW620, to analyze the cancer stem cell-like characteristics of CD133+ cells in vitro and in vivo. In vitro, CD133+ SW620 cells had a higher proliferative capacity, were more irradiation- and chemotherapy-resistant, and had a higher expression of β-catenin compared with CD133- cells. Injections of either CD133+ or CD133- cells into the skin or rectal mucosa of NOD/SCID mice led to tumors; however, injection of CD133+ cells resulted in the formation of larger tumors. Tumors derived from injections of CD133- cells did not contain any CD133+ cells, whereas tumors derived from injections of CD133+ cells did contain CD133+ cells, suggesting self-renewing capability. However, the proportion of CD133+ cells in the newly formed tumors in vivo was lower than the proportion of CD133+ cells in vitro. In conclusion, the human colon cancer cell line, SW620, contains both CD133+ and CD133- phenotypes, and the CD133+ phenotype has characteristics consistent with those of cancer stem cells.


Cell Transplantation | 2010

Isolation and propagation of a human CD133(-) colon tumor-derived cell line with tumorigenic and angiogenic properties.

Nalu Navarro-Alvarez; Eisaku Kondo; Hironobu Kawamoto; Wael A. R. A. Hassan; Takeshi Yuasa; Yasuhiro Kubota; Masayuki Seita; Hiroyuki Nakahara; Takahiro Hayashi; Yuriko Nishikawa; Reham A. R. A. Hassan; Shahid M. Javed; Hirofumi Noguchi; Shinichi Matsumoto; Shuhei Nakaji; Noriaki Tanaka; Naoya Kobayashi; Alejandro Soto-Gutierrez

It has been proposed in human colorectal cancers (CRC) a minority subset of cancer cells within tumors able to initiate tumor growth, defined as cancer stem cells (CSC). Solid human primary colonic and its ovarian metastatic cancer tissues were collected from fresh surgical samples and subsequent xenografts were established in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. The resulting tumors were disaggregated into single-cell suspensions and a CD133- cell line (NANK) was newly established and analyzed by flow cytometry. Surface markers of progenitor cells were immunophenotypically analyzed, and expression of stem cell and cancer-related genes was characterized. Secreted angiogenesis-associated molecules were investigated by proteomic array technology. Finally, different numbers of NANK were implanted and their tumor-initiating properties were investigated in NOD/SCID mice. Intraperitoneal injection of NANK in NOD/SCID mice induced tumors with developing progressive peritoneal dissemination and ascites. NANK cells maintained a differentiated phenotype and reproduced the full morphologic and phenotypic heterogeneity of their parental lesions. Noticeably, NANK lacked the expression of conventional CSC markers CD133 and CD44, self-renewal genes Oct-4 and Nanog, but showed the expression of an important gastrointestinal development marker CDX-2 and BMI-1 that is essential in regulating the proliferative activity of normal and leukemic stem cells. In addition, NANK secreted high amounts of important angiogeneic cytokines. These results provide a novel and extensive model in human CSC for studying the generation and maintenance of phenotypic heterogeneity in CRC.


Transplantation | 2009

Bone repair by transplantation of hTERT-immortalized human mesenchymal stem cells in mice

Hiroyuki Nakahara; Haruo Misawa; Takahiro Hayashi; Eisaku Kondo; Takeshi Yuasa; Yasuhiro Kubota; Masayuki Seita; Hironobu Kawamoto; Wael A. R. A. Hassan; Reham A. R. A. Hassan; Shahid M. Javed; Masato Tanaka; Hirosuke Endo; Hirofumi Noguchi; Shinichi Matsumoto; Katsuyoshi Takata; Yuichi Tashiro; Shuhei Nakaji; Toshifumi Ozaki; Naoya Kobayashi

Background. Human mesenchymal stem cells (hMSCs) are multipotent stem cells found in the adult bone marrow that have the capacity to differentiate into various mesenchymal cell types. The hMSCs may provide a potential therapy to restore damaged tissues or organs of mesenchymal origin; however, a drawback is their limited life span in vitro. Methods. We immortalized normal hMSCs with retrovirally transmitted human telomerase reverse transcriptase cDNA. One of the immortalized clones (YKNK-12) was established, and the biological characteristics were investigated in vitro and in vivo. Results. YKNK-12 cells were capable of differentiating adipocytes, osetoblasts, and chondrocytes. Osteogenically differentiated YKNK-12 cells produced significant levels of growth factors BMP4, BMP6, FGF6, FGF7, transforming growth factor-&bgr;1, and transforming growth factor-&bgr;3.. Microcomputer tomography T and soft X-ray assays showed an excellent calvarial bone healing in mice after transplantation of osteogenically differentiated YKNK-12 cells. These cells expressed human-specific osteocalcin and increased the gene expression of runt-related transcription factor 2, alkaline phosphatase, osteocalcin, and osterix in the bone regenerating area. YKNK-12 cell transplant corrected the bone defect without inducing any adverse effects. Conclusions. We conclude that hMSCs immortalized by transduction with human telomerase reverse transcriptase may provide an unlimited source of cells for therapeutic use in bone regeneration.


Cell Transplantation | 2009

Regenerative Medicine for Diabetes Mellitus

Naoya Kobayashi; Takeshi Yuasa; Teru Okitsu

In diabetes, a loss of pancreatic β-cells causes insulin dependency. When insulin dependency is caused by type 1 diabetes or pancreatic diabetes, for example, pancreatic β-cells need to be regenerated for definitive treatment. The methods for generating pancreatic β-cells include a method of creating pancreatic β-cells in vitro and implanting them into the body and a method of regenerating pancreatic β-cells in the body via gene introduction or the administration of differential proliferation factors to the body. Moreover, the number of pancreatic β-cells is also low in type 2 diabetes, caused by the compounding factors of insulin secretory failure and insulin resistance; therefore, if pancreatic β-cells can be regenerated in a living body, then a further amelioration of the pathology can be expected. The development of pancreatic β-cell-targeting regenerative medicine can lead to the next generation of diabetes treatment.


Diabetes | 2007

Cell-permeable pentapeptide V5 inhibits apoptosis and enhances insulin secretion, allowing experimental single-donor islet transplantation in mice

Jorge David Rivas-Carrillo; Alejandro Soto-Gutierrez; Nalu Navarro-Alvarez; Hirofumi Noguchi; Teru Okitsu; Yong Chen; Takeshi Yuasa; Kimiaki Tanaka; Michiki Narushima; Atsushi Miki; Haruo Misawa; Yasuhiko Tabata; Hee-Sook Jun; Shinichi Matsumoto; Ira J. Fox; Noriaki Tanaka; Naoya Kobayashi

OBJECTIVE—Treatment of diabetic patients by pancreatic islet transplantation often requires the use of islets from two to four donors to produce insulin independence in a single recipient. Following isolation and transplantation, islets are susceptible to apoptosis, which limits their function and probably long-term islet graft survival. RESEARCH DESIGN AND METHODS—To address this issue, we examined the effect of the cell-permeable apoptosis inhibitor pentapeptide Val-Pro-Met-Leu-Lys, V5, on pancreatic islets in a mouse model. RESULTS—V5 treatment upregulated expression of anti-apoptotic proteins Bcl-2 and XIAP (X-linked inhibitor of apoptosis protein) by more than 3- and 11-fold and downregulated expression of apoptosis-inducing proteins Bax, Bad, and nuclear factor-κB–p65 by 10, 30, and nearly 50%, respectively. Treatment improved the recovered islet mass following collagenase digestion and isolation by 44% and in vitro glucose-responsive insulin secretion nearly fourfold. Following transplantation in streptozotocin-induced diabetic mice, 150 V5-treated islet equivalents functioned as well as 450 control untreated islet equivalents in normalizing blood glucose. CONCLUSIONS—These studies indicate that inhibition of apoptosis by V5 significantly improves islet function following isolation and improves islet graft function following transplantation. Use of this reagent in clinical islet transplantation could have a dramatic impact on the number of patients that might benefit from this therapy and could affect long-term graft survival.


Cell Transplantation | 2010

Treatment of Acute Liver Failure in Mice by Hepatocyte Xenotransplantation

Tsuyoshi Yamamoto; Nalu Navarro-Alvarez; Alejandro Soto-Gutierrez; Takeshi Yuasa; Masaya Iwamuro; Yasuhiro Kubota; Masayuki Seita; Hironobu Kawamoto; Shahid M. Javed; Eisaku Kondo; Hirofumi Noguchi; Satoru Kobayashi; Shuhei Nakaji; Naoya Kobayashi

Liver diseases still have a high mortality even though liver transplantation has become a standard treatment. Currently, hepatocyte transplantation has been proposed as another promising strategy. One limitation is the availability of human livers as a source of hepatocytes. Because of an unlimited supply, the use of porcine hepatocytes might address this problem. Regardless of the source, once isolated hepatocytes lose specific functionality due to the loss of the natural microenvironment. For this reason, we tested the ability of a self-assembling peptide nanofiber (SAPNF) to provide a provisional three-dimensional (3D) support to interact with cells to control their function in vivo. Isolated porcine hepatocytes were embedded in SAPNF, or collagen type I and transplanted by direct injection into the splenic pulp of SCID mice suffering from acute liver failure (ALF) by 90% hepatectomy. SAPNF porcine hepatocyte transplantation produced engraftment that was far superior to that obtained using collagen and prolonged the survival of mice with ALF, in contrast with controls. An ultrastructural evaluation using transmission electron microscopy indicated extensive cell–cell communication and preservation of hepatocyte architecture. The transplanted SAPNF hepatocytes showed higher expression of albumin and PAS and lower apoptotic events assessed by TUNEL staining. Hepatocytes culture in a truly 3D network allows in vivo maintaining of differentiated functions, and once transplanted between widely divergent species can function to correct acute liver failure in mice and prolong their survival.


Cell Transplantation | 2010

Comparative analysis of endoderm formation efficiency between mouse ES cells and iPS cells.

Masaya Iwamuro; Toshiyuki Komaki; Yasuhiro Kubota; Masayuki Seita; Hironobu Kawamoto; Takeshi Yuasa; Javed M. Shahid; Reham A. R. A. Hassan; Wael A. R. A. Hassan; Shuhei Nakaji; Yuriko Nishikawa; Eisaku Kondo; Kazuhide Yamamoto; Naoya Kobayashi

Definitive endoderm (DE) derived from stem cells holds potential to differentiate into hepatocytes. Stem cell therapy using those cells has potential for a treatment of liver disease. To date, various ways of inducing hepatocytes from embryonic stem (ES) cells have been reported by researchers. However, it has not been proved enough that induced pluripotent stem (iPS) cells behave in the same manner as ES cells in endoderm differentiation. The purpose of this study was to establish an efficient method to induce DE from iPS cells, through comparatively analyzing the efficacy of endoderm formation from mouse ES cells. Furthermore, the efficiency of a serum-free medium in the differentiation into DE was investigated. Mouse ES cells and iPS cells were floated in culture medium for 2 or 5 days and embryoid bodies (EB) were formed. Subsequently, DE was induced with 100 ng/ml activin A and 100 ng/ml basic fibroblast growth factor (bFGF). RT-PCR and real-time PCR analyses were carried out at each step to determine the gene expression of EB markers. The difference in cellular proliferation between serum-containing and serum-free media was examined by an MTS assay in EB and DE induction. iPS cells showed the paralleled mRNA expression to ES cells in each step of differentiation into EB, but the levels of expression of Sox17 and Foxa2 were relatively higher in ES cell-derived DE, whereas Cxcr4 expression was higher in iPS cell-derived DE. The utilization of serum-free medium for iPS cells showed significantly favorable cellular proliferation during EB formation and subsequent DE induction. Forming EB for 5 days and subsequently DE induction with activin A and bFGF with serum-free medium was an appropriate protocol in iPS cells. This may represent an important step for generating hepatocytes from iPS cells for the development of cell therapy.

Collaboration


Dive into the Takeshi Yuasa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shuhei Nakaji

Okayama University of 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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge