Network


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

Hotspot


Dive into the research topics where Kazuyoshi Uchihashi is active.

Publication


Featured researches published by Kazuyoshi Uchihashi.


Endocrinology | 2008

A New Organotypic Culture of Adipose Tissue Fragments Maintains Viable Mature Adipocytes for a Long Term, Together with Development of Immature Adipocytes and Mesenchymal Stem Cell-Like Cells

Emiko Sonoda; Shigehisa Aoki; Kazuyoshi Uchihashi; Hidenobu Soejima; Sachiko Kanaji; Kenji Izuhara; Seiji Satoh; Noboru Fujitani; Hajime Sugihara; Shuji Toda

Adipose tissue that consists of mature and immature adipocytes is suggested to contain mesenchymal stem cells (MSCs), but a culture system for analyzing their cell types within the tissue has not been established. Here we show that three-dimensional collagen gel culture of rat sc adipose tissue fragments maintained viable mature adipocytes for a long term, producing immature adipocytes and MSC-like cells from the fragments, using immunohistochemistry, ELISA, and real time RT-PCR. Bromodeoxyuridine uptake of mature adipocytes was detected. Adiponectin and leptin, and adipocyte-specific genes of adiponectin, leptin, and PPAR-gamma were detected in culture assembly, whereas the lipogenesis factor insulin (20 mU/ml) and inflammation-related agent TNF-alpha (2 nm) increased and decreased, respectively, all of their displays. Both spindle-shaped cell types with oil red O-positive lipid droplets and those with expression of MSC markers (CD105 and CD44) developed around the fragments. The data indicate that adipose tissue-organotypic culture retains unilocular structure, proliferative ability, and some functions of mature adipocytes, generating both immature adipocytes and CD105+/CD44+ MSC-like cells. This suggests that our method will open up a new way for studying both multiple cell types within adipose tissue and the cell-based mechanisms of obesity and metabolic syndrome.


Cancer Science | 2008

Irradiated fibroblast‐induced bystander effects on invasive growth of squamous cell carcinoma under cancer–stromal cell interaction

Noriyuki Kamochi; Masahiro Nakashima; Shigehisa Aoki; Kazuyoshi Uchihashi; Hajime Sugihara; Shuji Toda; Sho Kudo

The irradiated fibroblast‐induced response of non‐irradiated neighboring cells is called ‘radiation‐induced bystander effect’, but it is unclear in non‐irradiated human squamous cell carcinoma (SCC) cells. The present study shows that irradiated fibroblasts promoted the invasive growth of T3M‐1 SCC cells, but not their apoptosis, more greatly than non‐irradiated fibroblasts, using collagen gel invasion assay, immunohistochemistry and Western blot. The number of irradiated fibroblasts decreased to about 30% of that of non‐irradiated fibroblasts, but irradiated fibroblasts increased the growth marker ki‐67 display of SCC cells more greatly than non‐irradiated fibroblasts. Irradiated fibroblasts did not affect the apoptosis marker ss‐DNA expression of SCC cells. Irradiated fibroblasts enhanced the display of the following growth‐, invasion‐ and motility‐related molecules in SCC cells more greatly than non‐irradiated fibroblasts: c‐Met, Ras, mitogen‐activated protein kinase (MAPK) cascade (Raf‐1, MEK‐1 and ERK‐1/2), matrix metalloproteinase‐1 and ‐9, laminin 5 and filamin A. Irradiated fibroblasts, but not non‐irradiated ones, formed irradiation‐induced foci (IRIF) of the genomic instability marker p53‐binding protein 1 (53BP1) and expressed transforming growth factor‐β1 (TGF‐ β1). Irradiated fibroblasts in turn enabled SCC cells to enhance 53BP1 IRIF formation more extensively than non‐irradiated fibroblasts. Finally, effects of irradiated fibroblasts on growth and apoptosis of another HEp‐2 SCC cell type were similar to those of T3M‐1. These results suggest that irradiated fibroblasts promotes invasion and growth of SCC cells by enhancement of invasive growth‐related molecules above through TGF‐ β1‐mediated bystander mechanism, in which irradiated fibroblast‐induced genomic instability of SCC cells may be involved. (Cancer Sci 2008; 99: 2417–2427)


Bone | 2013

Osteoblast migration into type I collagen gel and differentiation to osteocyte-like cells within a self-produced mineralized matrix: a novel system for analyzing differentiation from osteoblast to osteocyte.

Kazuyoshi Uchihashi; Shigehisa Aoki; Aki Matsunobu; Shuji Toda

Osteoblasts are believed to differentiate into osteocytes, becoming embedded in bone, or to undergo apoptosis after the bone formation phase. The regulation of this terminal differentiation seems to be critical for bone homeostasis. However the mechanism remains unclear and there is no assay system currently available to analyze this process. To address this issue, we developed a new model in which osteoblasts are cultured on a type I collagen gel layer with osteogenic supplements β-glycerophosphate and ascorbic acid. Cellular behavior was analyzed by electron microscopy, immunohistochemistry and real-time RT-PCR. Osteoblasts gradually migrated into the gel, produced collagen fibrils, and differentiated to osteocytic cells with bone lacunae- and canaliculi-like mineralization. Osteocalcin, DMP-1 and SOST protein expression was mainly expressed in the migrated cells within the mid-layer of the gel. Osteoblastic (ALP and osteocalcin) and osteocytic (PHEX, DMP-1 and SOST) mRNA expression was significantly increased compared with those of the cells cultured on plastic dishes alone after 21 days. The number of TUNEL-positive apoptotic cells gradually increased, reaching a maximum at 28 days. The cells were distributed at the surface and in the mid-layer of the gel at 7 days and after 14 days of culture, respectively. These data indicate that our model reproduces transition from osteoblasts to osteocytes, suggesting the following: 1) migration of osteoblasts into collagen gel may play a critical role in osteocytic differentiation; and 2) spatiotemporal gene expression and apoptosis may be involved in the terminal differentiation of osteoblasts. Our model will make it possible to study the mechanism of transition from osteoblast to osteocyte, and both cell type-related diseases including osteoporosis and osteonecrosis.


Organogenesis | 2009

Adipose tissue-organotypic culture system as a promising model for studying adipose tissue biology and regeneration

Shuji Toda; Kazuyoshi Uchihashi; Shigehisa Aoki; Emiko Sonoda; Fumio Yamasaki; Meihua Piao; Akifumi Ootani; Nobuhisa Yonemitsu; Hajime Sugihara

Adipose tissue consists of mature adipocytes, preadipocytes and mesenchymal stem cells (MSCs), but a culture system for analyzing their cell types within the tissue has not been established. We have recently developed “adipose tissue-organotypic culture system” that maintains unilocular structure, proliferative ability and functions of mature adipocytes for a long term, using three-dimensional collagen gel culture of the tissue fragments. In this system, both preadipocytes and MSCs regenerate actively at the peripheral zone of the fragments. Our method will open up a new way for studying both multiple cell types within adipose tissue and the cell-based mechanisms of obesity and metabolic syndrome. Thus, it seems to be a promising model for investigating adipose tissue biology and regeneration. In this article, we introduce adipose tissue-organotypic culture, and propose two theories regarding the mechanism of tissue regeneration that occurs specifically at peripheral zone of tissue fragments in vitro.


Cell and Tissue Research | 2011

Interaction between adipose tissue stromal cells and gastric cancer cells in vitro.

Namie Nomoto-Kojima; Shigehisa Aoki; Kazuyoshi Uchihashi; Aki Matsunobu; Eisuke Koike; Akifumi Ootani; Nobuhisa Yonemitsu; Kazuma Fujimoto; Shuji Toda

Adipose tissue exists in the gastric submucosa and subserosa. Thus, adipose tissue stromal cells (ATSCs), which include mesenchymal stem cells (MSCs), seem critical for the progression of gastric cancer but their interaction with the cancer cells is unknown. We demonstrated an interaction between these cells, using immunohistochemistry, Western blot and the collagen gel invasion assay system, in which the adenocarcinoma cells (well and poorly differentiated types, MKN28 and MKN45, respectively) were cultured on a ATSC-embedded or ATSC-non-embedded gel. ATSCs promoted the expression of the growth marker, proliferation cell nuclear antigen but inhibited that of the apoptosis marker, single-stranded DNA, in the cancer cell types. ATSCs accelerated the invasion of only MKN28 into the gel and promoted the expression of mitogen-activated protein kinase (MAPK, pERK-1/2) but decreased that of the molecularly targeted protein, HER2, in the cancer cells. ATSCs did not affect the expression of the prostaglandin biosynthetic enzyme cyclooxgenase-2 (COX-2) in the cancer cells. The COX-2 inhibitor celecoxib did not affect the morphology or invasion of the cancer cells. The cancer cell types in turn promoted the display of the myofibroblast marker, α-smooth muscle actin, whereas they decreased that of some MSC markers, e.g., CD44 and CD105, in ATSCs. The data suggest that (1) ATSCs influence the progression of gastric cancer by increasing their growth/invasion and decreasing their apoptosis through MAPK activation in a COX-2-independent way; (2) ATSCs adversely affect HER2-targeted therapy; (3) the cancer cells induce the cancer-associated myofibroblast phenotype in ATSCs.


Pathology International | 2009

Non-skin mesenchymal cell types support epidermal regeneration in a mesenchymal stem cell or myofibroblast phenotype-independent manner

Shigehisa Aoki; Toshiaki Takezawa; Kazuyoshi Uchihashi; Hajime Sugihara; Shuji Toda

Skin‐derived fibroblasts, preadipocytes and adipocytes, and non‐skin‐derived bone marrow stromal cells support epidermal regeneration. It remains unclear, however, whether various organ‐derived mesenchymal cell (MC) types other than the aforementioned counterparts affect epidermal regeneration. Using a skin reconstruction model, it is shown here that heart‐, spleen‐, lung‐, liver‐ and kidney‐derived MC support epidermal regeneration by keratinocytes. Immunohistochemistry showed that these MC types described here allowed keratinocytes to express cytokeratin (CK) 10, CK14 and involucrin in a normal fashion, and to retain the epidermal progenitor cell marker, p63, within the basal layer. MC types constantly expressed vimentin, but they were heterogeneous in their expression of the mesenchymal stem cell markers, stage‐specific embryonic antigen‐4, CD105, CD90 and CD44, and the myofibroblast marker, α‐smooth muscle actin. The MC types expressed keratinocyte growth factor, stromal‐derived factor‐1 and interleukin‐6, which are all critical for dermal fibroblast–keratinocyte interaction. These results indicate that vimentin‐positive MC originating from the heart, spleen, lung, liver and kidney can support epidermal regeneration without the involvement of mesenchymal stem cell and myofibroblast phenotypes of MC.


Kidney International | 2010

Adipose tissue explants and MDCK cells reciprocally regulate their morphogenesis in coculture

Kazuma Udo; Shigehisa Aoki; Kazuyoshi Uchihashi; Maki Kawasaki; Aki Matsunobu; Yuji Tokuda; Akifumi Ootani; Shuji Toda; Jiro Uozumi

Adipokine-producing fatty tissues, composed of preadipocytes, adipocytes, and mesenchymal stem cells, surround the kidney. To study the interaction between renal tubular cells and adipose tissue, we cocultured adipose tissue fragments and MDCK cells. MDCK cells in the coculture showed a taller columnar shape with improved organization of their microvilli and basal lamina than that seen in MDCK cell monoculture. The adipose tissue-induced change in morphology was replicated when we added leptin to MDCK cells cultured alone. Adiponectin abolished the leptin effect. Adipose tissue fragments inhibited MDCK cell division and also the formation of single-stranded DNA, an indicator of apoptosis. The fragments promoted the expression of polarity-associated proteins, including the tight junction molecules, ZO-1, atypical protein kinase C, and Cdc42. Further, the fragments also accelerated the expression of pendrin, the chloride/iodide transporter in the MDCK cells. In turn, MDCK cells decreased the number of preadipocytes and CD44+/CD105+ mesenchymal stem cells in the fragments, and promoted adiponectin production from the fragments. Thus, our study shows that adipose tissue fragments promote the hypertrophy, polarization, and differentiation of MDCK cells by attenuating their growth and apoptosis through opposing endocrine or paracrine effects of leptin and adiponectin. Further, MDCK cells inhibit the regeneration of preadipocytes and mesenchymal stem cells in adipose tissue.


Endocrinology | 2011

A Promising Culture Model for Analyzing the Interaction between Adipose Tissue and Cardiomyocytes

Mayumi Anan; Kazuyoshi Uchihashi; Shigehisa Aoki; Aki Matsunobu; Akifumi Ootani; Koichi Node; Shuji Toda

The heart has epicardial adipose tissue that produces adipokines and mesenchymal stem cells. Systemic adipose tissue is involved in the pathophysiology of obesity-related heart diseases. However, the method for analyzing the direct interaction between adipose tissue and cardiomyocytes has not been established. Here we show the novel model, using collagen gel coculture of adipose tissue fragments (ATFs) and HL-1 cardiomyocytes, and electron microscopy, immunohistochemistry, real-time RT-PCR, and ELISA. HL-1 cells formed a stratified layer on ATF-nonembedded gel, whereas they formed almost a monolayer on ATF-embedded gel. ATFs promoted the apoptosis, lipid accumulation, and fatty acid transport protein (FATP) expression of FATP4 and CD36 in HL-1 cells, whereas ATFs inhibited the growth and mRNA expression of myosin, troponin T, and atrial natriuretic peptide. Treatment of leptin (100 ng/ml) and adiponectin (10 μg/ml) neither replicated nor abolished the ATF-induced morphology of HL-1 cells, whereas that of FATP4 and CD36 antibodies (25 μg/ml) never abolished it. HL-1 cells prohibited the development of CD44+/CD105+ mesenchymal stem cell-like cells and lipid-laden preadipocytes from ATFs. HL-1 cells increased the production of adiponectin in ATFs, whereas they decreased that of leptin. The data indicate that our model actively creates adipose tissue-HL-1 cardiomyocyte interaction, suggesting first that ATFs may be related to the lipotoxiciy of HL-1 cells via unknown factors plus FATP4 and CD36 and second that HL-1 cells may help to retain the static state of ATFs, affecting adipokine secretion. Our model will serve to study adipose tissue-cardiomyocyte interaction and mechanisms of obesity-related lipotoxicity and heart diseases.


Pathology International | 2008

Sclerosing variant of epithelioid angiomyolipoma.

Atsuji Matsuyama; Masanori Hisaoka; Kazuhito Ichikawa; Takahiro Fujimori; Kazuma Udo; Kazuyoshi Uchihashi; Shigehisa Aoki; Hiroshi Hashimoto

Presented herein are two unusual epithelioid angiomyolipomas (AML) displaying prominent stromal sclerosis. Both patients were middle‐aged women without a clinical history of tuberous sclerosis. One patient (case 1) had a 2 cm lesion arising in the renal cortex, and another (case 2) had a pararenal retroperitoneal tumor measuring 13 cm. Both tumors were composed of sheets or nests of polygonal epithelioid or short spindle cells having uniform round to oval nuclei and eosinophilic cytoplasm with cords of hyalinized sclerotic stroma between them. The tumor in case 2 had small areas of mature‐looking fat cells. Immunohistochemically, epithelioid tumor cells were diffusely positive for actins and desmin in both cases, and melanoma antigen recognized by T cells (MART)‐1 was positive in patient 2. Scattered HMB‐45‐immunoreactive cells were identified in the sclerotic cords of both tumors, but epithelioid tumor cells were essentially negative for HMB‐45. The characteristic clinicopathological and immunohistochemical features of the present cases are analogous to a subset of epithelioid AML or sclerosing perivascular epithelioid cell tumors previously reported.


Peritoneal Dialysis International | 2011

FLUID FLOW STRESS AFFECTS PERITONEAL CELL KINETICS: POSSIBLE PATHOGENESIS OF PERITONEAL FIBROSIS

Shigehisa Aoki; Junichi Makino; Akinori Nagashima; Toshiaki Takezawa; Namie Nomoto; Kazuyoshi Uchihashi; Aki Matsunobu; Toru Sanai; Hajime Sugihara; Shuji Toda

♦ Background: Peritoneal fibrosis is an essential precursor condition to the development of encapsulating peritoneal sclerosis (EPS). This serious complication leads to a high mortality rate in peritoneal dialysis (PD) patients. Although several factors, including highly concentrated glucose in the dialysis solution, are believed to be potent agents for peritoneal fibrosis, the underlying mechanism remains unclear. During PD, the dialysis solution continuously generates fluid flow stress to the peritoneum under peristalsis and body motion. Fluid flow stress has been implicated as playing a critical role in the physiologic responses of many cell types. We therefore hypothesized that fluid flow stress may be involved in the pathogenesis of peritoneal fibrosis leading to EPS. ♦ Methods: To generate fluid flow stress, culture containers were placed on a rotatory shaker in a thermostatic chamber. In this system, the shaker rotated at a speed of 25 rpm with a radius of 1.5 cm. Mesothelial cells were cultured in low-glucose (1000 mg/L) or high-glucose (4500 mg/L) complete medium with and without flow stress. ♦ Results: Fluid flow stress promoted hyperplasia and epithelial-mesenchymal transition (EMT) of mesothelial cells independent of glucose concentration. Fluid flow stress inhibited expression of ERK (extracellular signal-regulated kinase) and p38 MAPK (mitogen-activated protein kinase) in mesothelial cells. Administration of ERK and p38 MAPK inhibitors replicated the stress-induced morphology of mesothelial cells. ♦ Conclusions: The present data indicate that fluid flow stress promotes hyperplasia and EMT of mesothelial cells via the MAPK axis, suggesting that fluid flow stress may be involved in the pathogenesis of peritoneal fibrosis.

Collaboration


Dive into the Kazuyoshi Uchihashi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hajime Sugihara

International University of Health and Welfare

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Masanori Hisaoka

University of Occupational and Environmental Health Japan

View shared research outputs
Top Co-Authors

Avatar

Toshiaki Takezawa

National Agriculture and Food Research Organization

View shared research outputs
Top Co-Authors

Avatar

Eisuke Shiba

University of Occupational and Environmental Health Japan

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge