Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
FEBS Open Bio ; 13(3): 434-446, 2023 03.
Article in English | MEDLINE | ID: mdl-36648092

ABSTRACT

Cancer stem cells (CSCs) are proposed to be involved in tumor initiation and play important roles in cancer relapse, metastasis, and drug resistance. Therefore, the targeting of CSCs has potential for effective anticancer therapies. Curcumin is one of the most widely characterized phytochemicals with tumor-suppressive potential. GO-Y030 is a novel curcumin analogue exhibiting a much stronger growth-inhibitory effect than curcumin. In the present study, we verified the potency of GO-Y030 against a CSC population. We observed that GO-Y030 suppressed CSC sphere-forming ability in several cancer cell lines. Interestingly, a specific inhibitor of heat shock protein (HSP) 70 also exhibited effects similar to GO-Y030 (i.e. inhibition of CSC sphere formation and upregulation of HSP70 and HSP40 protein expression), suggesting that HSP70 and/or HSP40 might be target molecules of GO-Y030. We then performed an in vitro HSP70/HSP40-mediated refolding activity assay and observed that chaperone activity was efficiently inhibited by GO-Y030. Finally, we performed a substrate-binding assay to show that GO-Y030 reduced the binding of both HSP70 and HSP40 with their substrates. HSPs prevent denaturation or unfolding of client proteins under stressful conditions such as high temperature. Because CSCs by nature adapt to various stresses by reinforcing protein-folding activity, the function of HSP70/HSP40 is important for the maintenance of CSC population. Our data suggest that GO-Y030 may impair stress tolerance in CSCs by inhibiting the interaction of HSP70/HSP40 with their substrate proteins and disrupting the function of HSP70/HSP40, thereby contributing to a reduction of the CSC population.


Subject(s)
Curcumin , Humans , Curcumin/pharmacology , HSP40 Heat-Shock Proteins/metabolism , Cell Line, Tumor , Neoplasm Recurrence, Local , HSP70 Heat-Shock Proteins/metabolism , Neoplastic Stem Cells/metabolism
2.
Pathol Int ; 70(3): 171-178, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31944485

ABSTRACT

Desmoplastic small round cell tumor (DSRCT) is a rare aggressive malignant tumor. It is a refractory tumor and the median overall survival is very short. We report two autopsy cases of DSRCT, both of which were already advanced and metastasized at the first medical examination. Both cases showed typical DSRCT findings in terms of localization of the lesions, histopathology and genetics, but the rate of disease progression was quite different. Survival after initial symptoms in Case 1 was only 12 months. On the other hand, survival after primary hospitalization in Case 2 was 42 months. The Case 2 patient initially received chemotherapy for advanced pancreatic carcinoma, because a nodule of the pancreatic tail was found on computed tomography (CT) scan. After chemotherapy, tumor regression was observed on CT scan. It is thus implied that adoption of the regimen for pancreatic carcinoma might have been one of reasons of the long survival in Case 2.


Subject(s)
Desmoplastic Small Round Cell Tumor/diagnostic imaging , Oncogene Proteins, Fusion/genetics , Pancreatic Neoplasms/diagnostic imaging , Adult , Autopsy , Desmoplastic Small Round Cell Tumor/drug therapy , Desmoplastic Small Round Cell Tumor/genetics , Desmoplastic Small Round Cell Tumor/pathology , Humans , In Situ Hybridization, Fluorescence , Male , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Tomography, X-Ray Computed , Translocation, Genetic/genetics , Pancreatic Neoplasms
3.
Int J Mol Sci ; 19(7)2018 Jul 23.
Article in English | MEDLINE | ID: mdl-30041406

ABSTRACT

It has long been known that the gap junction is down-regulated in many tumours. One of the downregulation mechanisms is the translocation of connexin, a gap junction protein, from cell membrane into cytoplasm, nucleus, or Golgi apparatus. Interestingly, as tumours progress and reinforce their malignant phenotype, the amount of aberrantly-localised connexin increases in different malignant tumours including oesophageal squamous cell carcinoma, thus suggesting that such an aberrantly-localised connexin should be oncogenic, although gap junctional connexins are often tumour-suppressive. To define the dual roles of connexin in head and neck squamous cell carcinoma (HNSCC), we introduced the wild-type connexin26 (wtCx26) or the mutant Cx26 (icCx26) gene, the product of which carries the amino acid sequence AKKFF, an endoplasmic reticulum-Golgi retention signal, at the C-terminus and is not sorted to cell membrane, into the human FaDu hypopharyngeal cancer cell line that had severely impaired the expression of connexin during carcinogenesis. wtCx26 protein was trafficked to the cell membrane and formed gap junction, which successfully exerted cell-cell communication. On the other hand, the icCx26 protein was co-localised with a Golgi marker, as revealed by immunofluorescence, and thus was retained on the way to the cell membrane. While the forced expression of wtCx26 suppressed both cell proliferation in vitro and tumorigenicity in mice in vivo, icCx26 significantly enhanced both cell proliferation and tumorigenicity compared with the mock control clones, indicating that an excessive accumulation of connexin protein in intracellular domains should be involved in cancer progression and that restoration of proper subcellular sorting of connexin might be a therapeutic strategy to control HNSCC.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Squamous Cell/metabolism , Connexin 26/metabolism , Head and Neck Neoplasms/metabolism , Animals , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Connexin 26/chemistry , Connexin 26/genetics , Golgi Apparatus/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Protein Sorting Signals , Protein Transport
4.
Int J Urol ; 23(6): 510-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27020040

ABSTRACT

OBJECTIVES: To clarify the interaction between adipose tissue stromal cells and bladder cancer cells. METHODS: Superficial (RT4) and invasive (EJ) urothelial carcinoma cells were cultured on adipose tissue stromal cell-embedded or non-embedded collagen gel. Cells were analyzed by immunohistochemistry, western blot and real-time reverse transcription polymerase chain reaction. RESULTS: Adipose tissue stromal cells inhibited growth of RT4, while they promoted the apoptosis. In contrast, adipose tissue stromal cells promoted growth of EJ, but they did not affect the apoptosis. Adipose tissue stromal cells slightly promoted expression of mitogen-activated protein kinase cascade in RT4 and EJ. Adipose tissue stromal cells promoted display of the molecular-targeted agent human epidermal growth factor receptor-2 in only RT4. In turn, RT4 and EJ enhanced α-smooth muscle actin (myofibroblast marker) and S-100 protein (adipocyte marker) expression of adipose tissue stromal cells, respectively. CONCLUSIONS: These findings suggest that: (i) adipose tissue stromal cells might suppress the progression of superficial-type cancer, whereas they might promote that of invasive type; (ii) adipose tissue stromal cell-activated mitogen-activated protein kinase pathway might play differential roles in both types of bladder cancer; (iii) human epidermal growth factor receptor-2 could represent a critical therapeutic agent for the superficial type under adipose tissue stromal cells-cancer interaction; and (iv) superficial bladder cancer might promote myofibroblast differentiation of adipose tissue stromal cells as a cancer-associate phenotype, whereas invasive bladder cancer might promote their adipocyte differentiation.


Subject(s)
Apoptosis , Carcinoma, Transitional Cell/pathology , Neoplasm Invasiveness , Stromal Cells , Urinary Bladder Neoplasms/pathology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Humans
5.
Am J Pathol ; 186(5): 1180-94, 2016 05.
Article in English | MEDLINE | ID: mdl-26952643

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) develops within the squamous epithelial layer and invades the submucosa to the subadventitia that has adipose tissue (AT). AT seems critical to ESCC progression, but the underlying mechanism is unknown. We aimed to address the association between ESCC and AT in vitro. ESCC cells were cultured on rat or human subcutaneous AT-embedded or -non-embedded collagen gel. AT promoted the growth of ESCC cells and inhibited their apoptosis. AT promoted the expression of the squamous differentiation marker involucrin in ESCC cells. AT accelerated the expression of invasion-related factors in poorly differentiated ESCC cells only. AT promoted the expression of phosphorylated-insulin-like growth factor-1 receptor in ESCC cells, whereas it inhibited that of the human epidermal growth factor receptor 2. Insulin-like growth factor-1, but not leptin, adiponectin, or resistin, promoted and inhibited the growth and apoptosis of ESCC cells, respectively. In turn, ESCC cells decreased the production of these adipokines in AT and the number of preadipocytes and mesenchymal stem cell-like cells, which developed from AT. These results suggest that i) AT may influence the progression of ESCC with increased growth or invasion and decreased apoptosis through insulin-like growth factor-1/insulin-like growth factor-1 receptor signaling, ii) AT may affect human epidermal growth factor receptor 2-targeted therapy; and iii) the cancer cells may affect adipokine production in AT.


Subject(s)
Adipose Tissue/physiology , Carcinoma, Squamous Cell/physiopathology , Esophageal Neoplasms/physiopathology , Adiponectin/pharmacology , Animals , Apoptosis/physiology , Biomarkers, Tumor/metabolism , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic , Esophageal Squamous Cell Carcinoma , Filamins/metabolism , Humans , Hypertrophy/physiopathology , Insulin-Like Growth Factor I , Lipid Metabolism/physiology , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 9/metabolism , Microscopy, Electron , Protein Precursors/metabolism , Rats, Wistar , Receptor, ErbB-2/metabolism , Resistin/pharmacology , Stromal Cells/physiology , Tumor Cells, Cultured , Kalinin
6.
Pathol Int ; 66(2): 75-82, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26753834

ABSTRACT

Tumor budding is a major risk factor for T1 colorectal cancer. Quality control of the pathological diagnosis of budding is crucial, irrespective of the pathologist's experience. This study examines the interobserver variability according to pathologists' experience and evaluates the influence of cytokeratin (CK) immunostaining in the assessment of budding. Hematoxylin-eosin (HE) and CK-immunostained slides of 40 cases with T1 primary colorectal cancer were examined. Budding grades were individually evaluated by 12 pathologists who we categorized into three groups by their experience (expert, with >10 years of experience (n = 4), senior, with 5-10 years (n = 4), and junior, < 5 years (n = 4)). The results revealed a tendency for the more experienced pathologists to assign higher budding grades compared to the less-experienced pathologists. In the junior group, the interobserver variability obtained with HE slides was poor, but it was markedly improved in the evaluation using CK-immunostained slides. The benefit of CK immunostaining was only slight in the expert group. CK immunostaining would be useful when a pathologist is not experienced enough or does not have enough confidence in the assessment of budding.


Subject(s)
Colorectal Neoplasms/pathology , Keratins/metabolism , Colorectal Neoplasms/metabolism , Humans , Immunohistochemistry , Lymphatic Metastasis , Neoplasm Grading , Observer Variation , Reproducibility of Results
7.
PLoS One ; 10(9): e0137257, 2015.
Article in English | MEDLINE | ID: mdl-26339797

ABSTRACT

Gastric cancer grows under a hypoxic environment. HIF-1α is known to play an important role in controlling the production of reactive oxygen species (ROS) in the mitochondria under hypoxic conditions. We previously established HIF-1α knockdown (KD) cells and control (SC) cells in the 58As9 gastric cancer cell line. In this study, we revealed that KD cells, but not SC cells, induced apoptosis under conditions of hypoxia (1% O2) due to excessive production of ROS. A quantitative RT-PCR analysis demonstrated that the expressions of ten genes, which are involved in the control mechanisms of ROS (including the Warburg effect, mitophagy, electron transport chain [ETC] modification and ROS scavenging), were regulated by HIF-1α. Moreover, the promotion of glucose uptake by glucose plus insulin (GI) treatment enhanced the apoptotic effect, which was accompanied by further ROS production in hypoxic KD cells. A Western blot analysis showed that the membranous expression of GLUT1 in KD cells was elevated by glucose and/or insulin treatments, indicating that the GI-induced glucose uptake is mediated by the increased translocation of GLUT1 on the cell membrane. Finally, the anti-tumor effect of HIF-1α knockdown (KD) plus GI was evaluated using a tumor xenograft model, where a hypoxic environment naturally exists. As a result, the GI treatment strongly inhibited the growth of the KD tumors whereby cell apoptosis was highly induced in comparison to the control treatment. In contrast, the growth of the SC tumors expressing HIF-1α was not affected by the GI treatment. Taken together, the results suggest that HIF-1α inhibition plus GI may be an ideal therapy, because the apoptosis due to the destruction of ROS homeostasis is specifically induced in gastric cancer that grows under a hypoxic environment, but not in the normal tissue under the aerobic conditions.


Subject(s)
Gene Expression Regulation, Neoplastic , Glucose/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia/drug therapy , Insulin/pharmacology , Stomach Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Hypoxia , Cell Line, Tumor , Female , Gene Knockdown Techniques , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oxygen/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
8.
Pathol Int ; 64(6): 276-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24965110

ABSTRACT

Stenosing flexor tenosynovitis, trigger finger, is a common clinical disorder causing painful locking or contracture of the involved digits, and most instances are idiopathic. This problem is generally caused by a size mismatch between the swollen flexor tendon and the thickened first annular pulley. Although hypertrophic pulleys have been histologically and ultrasonographically detected, little is known about the histopathology of the tenosynovium covering the tendons of trigger fingers. We identified chondrocytoid cells that produced hyaluronic acid in 23 (61%) fingers and hypocellular collagen matrix in 32 (84%) fingers around the tenosynovium among 38 specimens of tenosynovium from patients with trigger fingers. These chondrocytoid cells expressed the synovial B cell marker CD44, but not the chondrocyte marker S-100 protein. The incidence of these findings was much higher than that of conventional findings of synovitis, such as inflammatory infiltrate (37%), increased vascularity (37%), hyperplasia of synovial lining cells (21%), or fibrin exudation (5%). We discovered the following distinctive histopathological features of trigger finger: hyaluronic acid-producing chondrocytoid cells originated from fibroblastic synovial B cells, and a hypocellular collagen matrix surrounding the tenosynovium. Thus, an edematous extracellular matrix with active hyaluronic acid synthesis might increase pressure under the pulley and contribute to the progression of stenosis.


Subject(s)
Synovial Membrane/pathology , Tendons/pathology , Trigger Finger Disorder/pathology , Adult , Aged , Female , Humans , Hyaluronic Acid/metabolism , Male , Middle Aged , S100 Proteins/metabolism , Synovial Membrane/metabolism , Tendons/metabolism , Trigger Finger Disorder/metabolism
9.
Hum Pathol ; 44(11): 2614-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23954138

ABSTRACT

Phosphaturic mesenchymal tumors of the mixed connective tissue type (PMT-MCTs) are rare neoplasms, most of which are benign and cause tumor-induced osteomalacia because of overproduction of a phosphaturic hormone, fibroblast growth factor 23 (FGF23). This entity may have been unrecognized or misdiagnosed as other mesenchymal tumors, such as giant cell tumor, hemangiopericytoma, and osteosarcoma. Ten percent of these tumors, without phosphaturia, were diagnosed only by their histologic features. We report here the first case of malignant PMT-MCT, nonphosphaturic variant, resulting in fatal multiple lung metastases. Chondromyxoid matrix with "grungy" calcification, multinucleated giant cell proliferation, and expression of FGF23 mRNA (reverse transcription-polymerase chain reaction) and fibroblast growth factor 23 protein (immunohistochemistry) were seen in the primary and recurrent tumors of the right foot. The lung metastases showed flocculent calcification and FGF23 protein expression as well as giant cell proliferation. This unique case highlights the need for careful histologic assessment of PMT-MCTs, especially the nonphosphaturic variant, and the need for recognition of its rare malignant behavior.


Subject(s)
Foot/pathology , Lung Neoplasms/secondary , Mesenchymoma/pathology , Neoplasm Recurrence, Local/pathology , Neoplasms, Connective Tissue/pathology , Soft Tissue Neoplasms/pathology , Adult , Calcinosis , Cell Proliferation , Diagnosis, Differential , Fatal Outcome , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Giant Cells/pathology , Humans , Hypophosphatemia, Familial , Immunohistochemistry , Lung/pathology , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Mesenchymoma/metabolism , Mesenchymoma/secondary , Neoplasm Recurrence, Local/metabolism , Neoplasms, Connective Tissue/metabolism , Neoplasms, Connective Tissue/secondary , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/surgery
10.
J Artif Organs ; 16(3): 322-31, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23526130

ABSTRACT

Adipose tissue, together with the mesothelial layer and microvessels, is a major component of the mesenteric peritoneum, and the mesenterium is a target site for peritoneal fibrosis. Adipose tissue has been speculated to play a role in peritoneal dialysis (PD)-related fibrosis, but the precise cellular kinetics of adipose tissue during this process remain to be determined. To clarify this critical issue, we analyzed the kinetics of adipose tissue using a novel peritoneal reconstruction model in which the effects of mesothelial cells or endothelial cells could be identified. Adipose tissue was co-cultured with mesothelial cells or endothelial cells in a combined organ culture and fluid flow stress culture system. Spindle mesenchymal cells and immature adipocytes derived from adipose tissue were characterized by immunohistochemistry. Adipose tissue fragments cultured in this system yielded many spindle mesenchymal cells in non-co-culture conditions. However, the number of spindle mesenchymal cells emerging from adipose tissue was reduced in co-culture conditions with a covering layer of mesothelial cells. Mesothelial cells co-cultured in the separated condition did not inhibit the emergence of spindle mesenchymal cells from adipose tissue. Interestingly, endothelial cells promoted the emergence of lipid-laden immature adipocytes from adipose tissue under fluid flow stress. We have demonstrated that adipose tissue behavior is not only regulated by mesothelial cells and endothelial cells under fluid flow stress, but is also involved in fibrosis and fat mass production in the peritoneum. Our findings suggest that adipose tissue is a potential source of cells for peritoneal fibrosis caused by PD therapy.


Subject(s)
Adipose Tissue/pathology , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/etiology , Peritoneum/pathology , Animals , Coculture Techniques , Endothelial Cells/pathology , Epithelial Cells/pathology , Humans , Peritoneal Fibrosis/pathology , Rats , Rats, Sprague-Dawley
11.
Cell Tissue Res ; 352(3): 611-21, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23512139

ABSTRACT

Systemic adipose tissue is involved in the pathophysiology of obesity-associated liver diseases. However, a method has not been established for analyzing the direct interaction between adipose tissue and hepatocytes. We describe a useful three-dimensional model comprising a collagen gel coculture system in which HepG2 hepatocytes are cultured on a gel layer with visceral adipose tissue fragments (VAT) or subcutaneous tissue samples (SAT). Male adipose tissues were obtained from 5-week-old Wistar rats and human autopsy cases. Cellular behavior was analyzed by electron microscopy, immunohistochemistry, Western blot, real-time reverse transcription plus the polymerase chain reaction and enzyme-linked immunosorbent assay. VAT significantly promoted lipid accumulation and apoptosis in HepG2 cells and suppressed their growth and differentiation compared with SAT. VAT produced higher concentrations of fatty acids (palmitate, oleate, linoleate) than SAT. HepG2 cells significantly decreased the production of these fatty acids in VAT. Only HepG2 cells treated with 250 µM palmitate replicated VAT-induced apoptosis. Neither VAT nor SAT affected lipotoxicity-associated signals of nuclear factor kappa B, c-Jun N-terminal kinase and inositol requiring enzyme-1α in HepG2 cells. HepG2 cells never affected adiponectin, leptin, or resistin production in VAT and SAT. The data indicate that our model actively creates adipose tissue and HepG2 hepatocyte interactions, suggesting that (1) VAT plays more critical roles in hepatocyte lipotoxicity than SAT; (2) palmitate but not adipokines, is partly involved in the mechanisms of VAT-induced lipotoxicity; (3) HepG2 cells might inhibit fatty acid production in VAT to protect themselves against lipotoxicity. Our model should serve in studies of interactions between adipose tissue and hepatocytes and of the mechanisms in obesity-related lipotoxicity and liver diseases.


Subject(s)
Adipose Tissue/cytology , Cell Communication , Cell Culture Techniques/methods , Hepatocytes/cytology , Adipokines/biosynthesis , Animals , Apoptosis/drug effects , Cell Communication/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Fatty Acids/biosynthesis , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Humans , Intra-Abdominal Fat/cytology , Lipid Metabolism/drug effects , Lipids/toxicity , Male , Mice , Models, Biological , NIH 3T3 Cells , Rats , Rats, Wistar , Subcutaneous Fat/cytology
12.
Bone ; 52(1): 102-10, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22985890

ABSTRACT

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 ß-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.


Subject(s)
Calcification, Physiologic , Cell Movement , Collagen Type I/metabolism , Extracellular Matrix/metabolism , Osteoblasts/cytology , Osteoclasts/cytology , Animals , Base Sequence , DNA Primers , Immunohistochemistry , Mice , Real-Time Polymerase Chain Reaction
13.
Biochem Biophys Res Commun ; 416(3-4): 391-6, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22120632

ABSTRACT

Encapsulating peritoneal sclerosis (EPS) often develops after transfer to hemodialysis and transplantation. Both termination of peritoneal dialysis (PD) and transplantation-related factors are risks implicated in post-PD development of EPS, but the precise mechanism of this late-onset peritoneal fibrosis remains to be elucidated. We previously demonstrated that fluid flow stress induced mesothelial proliferation and epithelial-mesenchymal transition via mitogen-activated protein kinase (MAPK) signaling. Therefore, we speculated that the prolonged bioactive effect of fluid flow stress may affect mesothelial cell kinetics after cessation of fluid streaming. To investigate how long mesothelial cells stay under the bioactive effect brought on by fluid flow stress after removal of the stress, we initially cultured mesothelial cells under fluid flow stress and then cultured the cells under static conditions. Mesothelial cells exposed to fluid flow stress for a certain time showed significantly high proliferative activity compared with static conditions after stoppage of fluid streaming. The expression levels of protein phosphatase 2A, which dephosphorylates MAPK, in mesothelial cells changed with time and showed a biphasic pattern that was dependent on the duration of exposure to fluid flow stress. There were no differences in the fluid flow stress-related bioactive effects on mesothelial cells once a certain time had passed. The present findings show that fluid flow stress exerts a prolonged bioactive effect on mesothelial cells after termination of fluid streaming. These findings support the hypothesis that a history of PD for a certain period could serve as a trigger of EPS after stoppage of PD.


Subject(s)
Cell Proliferation , Epithelial Cells/pathology , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/etiology , Stress, Physiological , Animals , Epithelial Cells/enzymology , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Peritoneal Fibrosis/pathology , Phosphorylation , Rats , Rats, Wistar
14.
Perit Dial Int ; 31(4): 466-76, 2011.
Article in English | MEDLINE | ID: mdl-21532005

ABSTRACT

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.


Subject(s)
Dialysis Solutions , Epithelial Cells , Peritoneal Dialysis , Peritoneal Fibrosis/etiology , Peritoneum/cytology , Cells, Cultured , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Humans , Hyperplasia , Mitogen-Activated Protein Kinases/physiology , Rheology , Stress, Mechanical
15.
Cell Tissue Res ; 344(2): 287-98, 2011 May.
Article in English | MEDLINE | ID: mdl-21384185

ABSTRACT

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.


Subject(s)
Adipose Tissue/cytology , Cell Communication/physiology , Stomach Neoplasms/pathology , Stromal Cells/cytology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adipose Tissue/metabolism , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2 Inhibitors/pharmacology , Humans , Immunohistochemistry , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 3/biosynthesis , Mucins/biosynthesis , Neoplasm Invasiveness , Phenotype , Rats , Rats, Wistar , Receptor, ErbB-2/biosynthesis , Stomach Neoplasms/metabolism , Stromal Cells/metabolism
16.
Endocrinology ; 152(4): 1599-605, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21303960

ABSTRACT

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.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/metabolism , Coculture Techniques/methods , Myocytes, Cardiac/cytology , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Enzyme-Linked Immunosorbent Assay , Fatty Acid Transport Proteins/genetics , Fatty Acid Transport Proteins/metabolism , Humans , Immunohistochemistry , Mice , Microscopy, Electron , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Myosins/genetics , Myosins/metabolism , NIH 3T3 Cells , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Troponin T/genetics , Troponin T/metabolism
17.
ISRN Endocrinol ; 2011: 275782, 2011.
Article in English | MEDLINE | ID: mdl-22363871

ABSTRACT

The thyroid is composed of thyroid follicles supported by extracellular matrix, capillary network, and stromal cell types such as fibroblasts. The follicles consist of thyrocytes and C cells. In this microenvironment, thyrocytes are highly integrated in their specific structural and functional polarization, but monolayer and floating cultures cannot allow thyrocytes to organize the follicles with such polarity. In contrast, three-dimensional (3-D) collagen gel culture enables thyrocytes to form 3-D follicles with normal polarity. However, these systems never reconstruct the follicles consisting of both thyrocytes and C cells. Thyroid tissue-organotypic culture retains 3-D follicles with both thyrocytes and C cells. To create more appropriate experimental models, we here characterize four culture systems above and then introduce the models for studying thyroid biology and disorders. Finally, we propose a new approach to the cell type-specific culture systems on the basis of in vivo microenvironments of various cell types.

18.
Acta Histochem Cytochem ; 43(1): 1-7, 2010 Feb 27.
Article in English | MEDLINE | ID: mdl-20300218

ABSTRACT

The air-liquid interface (ALI) is a common microenvironment of the skin, but it is unknown whether the ALI affects melanoma cell behaviors. Using a collagen gel invasion assay, immunohistochemistry, and Western blots, here we show that melanoma cell proliferation in cultures with an ALI is higher than melanoma cell proliferation in submerged cultures. Bromodeoxyuridine (BrdU) uptake, an indicator of cell proliferation, of melanoma cells at the ALI was about 3 times that of submerged cells, while ALI and submerged melanoma cells had similar levels of single-stranded DNA (a marker of apoptosis). The ALI enhanced the expression of Raf-1, MEK-1 and pERK-1/2 components of the mitogen-activated protein kinase (MAPK) cascade, in cells more than the submerged condition did. The increases in BrdU uptake and pERK-1/2 expression promoted by ALI was abolished by the MEK inhibitor, PD-98059. ALI-treated and submerged melanoma cells did not infiltrate into the collagen gel, and they showed no significant difference in the expression of the invasion- and motility-related molecules, matrix metalloproteinase-1 and -9, laminin 5, and filamin A. Our data indicate that the ALI, a skin microenvironment, accelerates the growth, but not the apoptosis or invasion, of melanoma cells through MAPK activation.

19.
Kidney Int ; 78(1): 60-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20336056

ABSTRACT

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.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/metabolism , 3T3 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipokines , Adiponectin/genetics , Adiponectin/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Differentiation/genetics , Cell Line , Coculture Techniques , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Dogs , Leptin/genetics , Mesenchymal Stem Cells/metabolism , Mice , Morphogenesis/genetics , Rats , Rats, Wistar , Regeneration/genetics
20.
J Gastroenterol ; 44(4): 261-70, 2009.
Article in English | MEDLINE | ID: mdl-19280112

ABSTRACT

PURPOSE: The purpose of this study was to compare the efficacy of proton pump inhibitor (PPI) with H(2) receptor antagonist (H(2)RA) in treatment of upper abdominal symptoms. METHODS: This was a multi-center, open study conducted at 102 hospitals in Japan. Patients with reflux esophagitis received famotidine 10 mg twice daily for 2 weeks, then omeprazole 10 mg once daily for 2 weeks. Thereafter, patients were switched to famotidine 10 mg twice daily for a third 2-weekperiod, provided those with a medical condition agreed to continue the study. Patients evaluated the treatment response to each gastrointestinal symptom using a predefined patient questionnaire and gastrointestinal symptom rating scale (GSRS). RESULTS: 161 patients entered the study, of whom 8 were excluded from all analyses due to lack of participation following entry. Overall symptom improvement rate (n = 130) at week 4, after the 2-week omeprazole treatment, was 75.4% and this was significantly higher than that after the first 2-week famotidine treatment (41.5%) at week 2. In patients (n = 36) who completed 6 weeks of treatment, 2-week omeprazole treatment at week 4 showed a significantly higher overall symptom improvement rate compared with both the first 2-week and third 2-week famotidine treatments. CONCLUSIONS: Omeprazole was superior to famotidine for treatment of upper abdominal symptoms in patients with reflux esophagitis, which suggested that gastric acid might be a cause not only of reflux symptoms, but also of ulcer symptoms and dysmotility symptoms such as epigastric pain and feeling of fullness in reflux esophagitis.


Subject(s)
Anti-Ulcer Agents/therapeutic use , Esophagitis, Peptic/drug therapy , Famotidine/therapeutic use , Omeprazole/therapeutic use , Adult , Aged , Female , Histamine H2 Antagonists/therapeutic use , Humans , Male , Middle Aged , Proton Pump Inhibitors/therapeutic use , Severity of Illness Index , Surveys and Questionnaires , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...