Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cytotherapy ; 26(7): 769-777, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38556961

ABSTRACT

BACKGROUND AIMS: The administration of human cell-processed therapeutic products (hCTPs) is associated with a risk of tumorigenesis due to the transformed cellular contaminants. To mitigate this risk, these impurities should be detected using sensitive and validated assays. The digital soft agar colony formation (D-SAC) assay is an ultrasensitive in vitro test for detecting tumorigenic transformed cells in hCTPs. METHODS: In this study, we first evaluated the colony formation efficiency (CFE) precision of tumorigenic reference cells in positive control samples according to a previously reported D-SAC assay protocol (Protocol I) from multiple laboratories. However, the CFE varied widely among laboratories. Thus, we improved and optimized the test protocol as Protocol II to reduce variability in the CFE of tumorigenic reference cells. Subsequently, the improved protocol was validated at multiple sites. Human mesenchymal stromal cells (hMSCs) were used as model cells, and positive control samples were prepared by spiking them with HeLa cells. RESULTS: Based on the previously reported protocol, the CFE was estimated using an ultra-low concentration (0.0001%) of positive control samples in multiple plates. Next, we improved the protocol to reduce the CFE variability. Based on the CFE results, we estimated the sample size as the number of wells (Protocol II) and assessed the detectability of 0.0001% HeLa cells in hMSCs to validate the protocol at multiple sites. Using Protocol I yielded low CFEs (mean: 30%) and high variability between laboratories (reproducibility coefficient of variance [CV]: 72%). In contrast, Protocol II, which incorporated a relatively high concentration (0.002%) of HeLa cells in the positive control samples, resulted in higher CFE values (mean: 63%) and lower variability (reproducibility CV: 18%). Moreover, the sample sizes for testing were estimated as the number of wells per laboratory (314-570 wells) based on the laboratory-specific CFE (42-76%). Under these conditions, all laboratories achieved a detection limit of 0.0001% HeLa cells in hMSCs in a predetermined number of wells. Moreover, colony formation was not observed in the wells seeded with hMSCs alone. CONCLUSIONS: The D-SAC assay is a highly sensitive and robust test for detecting malignant cells as impurities in hCTPs. In addition, optimal assay conditions were established to test tumorigenic impurities in hCTPs with high sensitivity and an arbitrary false negative rate.


Subject(s)
Cell- and Tissue-Based Therapy , Mesenchymal Stem Cells , Humans , HeLa Cells , Cell- and Tissue-Based Therapy/methods , Mesenchymal Stem Cells/cytology , Cell Transformation, Neoplastic
2.
J Toxicol Sci ; 47(9): 359-373, 2022.
Article in English | MEDLINE | ID: mdl-36047110

ABSTRACT

Hepatotoxicity is one of the most common toxicities observed in non-clinical safety studies of drug candidates, and it is important to understand the hepatotoxicity mechanism to assess the risk of drug-induced liver injury in humans. In this study, we investigated the mechanism of hepatotoxicity caused by 2-[2-Methyl-1-(oxan-4-yl)-1H-benzimidazol-5-yl]-1,3-benzoxazole (DSP-0640), a drug candidate that showed hepatotoxicity characterized by centrilobular hypertrophy and vacuolation of hepatocytes in a 4-week oral repeated-dose toxicity study in male rats. In the liver of rats treated with DSP-0640, the expression of aryl hydrocarbon receptor (AHR) target genes, including Cyp1a1, was upregulated. In in vitro reporter assays, however, DSP-0640 showed only minimal AHR-activating potency. Therefore, we investigated the possibility that DSP-0640 indirectly activated AHR by inhibiting the CYP1 enzyme-dependent clearance of endogenous AHR agonists. In in vitro assays, DSP-0640 showed inhibitory effects on both rat and human CYP1A1 and enhanced rat and human AHR-mediated reporter gene expression induced by 6-formylindolo[3,2-b]carbazole, a well-known endogenous AHR agonist. The possible involvement of CYP1A1 inhibition in AHR activation was also demonstrated with other hepatotoxic compounds tacrine and albendazole. These results suggest that CYP1A1 inhibition-mediated AHR activation is involved in the hepatotoxicity caused by DSP-0640 and that DSP-0640 might induce hepatotoxicity in humans as well. We propose that CYP1A1 inhibition-mediated AHR activation is a novel mechanism for drug-induced hepatotoxicity.


Subject(s)
Chemical and Drug Induced Liver Injury , Cytochrome P-450 CYP1A1 , Receptors, Aryl Hydrocarbon , Animals , Basic Helix-Loop-Helix Transcription Factors , Chemical and Drug Induced Liver Injury/genetics , Cytochrome P-450 CYP1A1/metabolism , Genes, Reporter , Hepatocytes/metabolism , Humans , Male , Rats , Receptors, Aryl Hydrocarbon/metabolism
3.
Breast Cancer Res Treat ; 156(1): 45-55, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26943913

ABSTRACT

The great majority of invasive lobular carcinoma (ILC) is estrogen-dependent luminal A type carcinoma but the details of estrogen actions and its intratumoral metabolism have not been well studied compared to invasive ductal carcinoma (IDC). We first immunolocalized estrogen-related enzymes including estrogen sulfotransferase (EST), estrogen sulfatase (STS), 17ß-hydroxysteroid dehydrogenase (HSD) 1/2, and aromatase. We then evaluated the tissue concentrations of estrogens in ILC and IDC and subsequently estrogen-responsive gene profiles in these tumors in order to explore the possible differences and/or similarity of intratumoral estrogen environment of these two breast cancer subtypes. The status of STS and 17ßHSD1 was significantly lower in ILCs than IDCs (p = 0.022 and p < 0.0001), but that of EST and 17ßHSD2 vice versa (p < 0.0001 and p = 0.0106). In ILCs, tissue concentrations of estrone and estradiol were lower than those in IDCs (p = 0.0709 and 0.069). In addition, the great majority of estrogen response genes tended to be lower in ILCs. Among those genes above, FOXP1 was significantly higher in ILCs than in IDCs (p = 0.002). FOXP1 expression was reported to be significantly higher in relapse-free IDC patients treated with tamoxifen. Therefore, tamoxifen may be considered an option of endocrine therapy for luminal A type ILC patients. This is the first study to demonstrate the detailed and comprehensive status of intratumoral production and metabolism of estrogens and the status of estrogen response genes in luminal A-like ILC with comparison to those in luminal A-like IDCs.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Lobular/metabolism , Estrogens/biosynthesis , 17-Hydroxysteroid Dehydrogenases/metabolism , Adult , Aged , Aged, 80 and over , Aromatase/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/genetics , Carcinoma, Lobular/pathology , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Repressor Proteins/genetics , Sulfatases/metabolism , Sulfotransferases/metabolism
4.
Mol Cell Endocrinol ; 413: 236-47, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26170067

ABSTRACT

Prostaglandins are a group of lipid compounds involved in inflammation and cancer. We focused on PGF2α and its stereoisomer 11ß-PGF2α and examined the expression and functions of their cognate receptor (FP receptor) and metabolizing enzymes (AKR1B1 and AKR1C3 respectively) in breast cancer. In immunohistochemical analysis FP receptor status associated with adverse clinical outcome only in the AKR1C3 positive cases. Therefore, we studied FP receptor-mediated functions of 11ß-PGF2α using FP receptor expressed MCF-7 cell line (MCF-FP). 11ß-PGF2α treatment phosphorylated ERK and CREB and induced Slug expression through FP receptor in MCF-FP, and MCF-FP cells demonstrated decreased chemosensitivity compared to parental controls. Finally, the correlation between FP receptor and Slug was also confirmed immunohistochemically in breast cancer cases. Overall these results indicated that the actions of AKR1C3 can produce FP receptor ligands whose activation results in carcinoma cell survival in breast cancer.


Subject(s)
3-Hydroxysteroid Dehydrogenases/metabolism , Breast Neoplasms/metabolism , Dinoprost/metabolism , Gene Expression Regulation, Neoplastic , Hydroxyprostaglandin Dehydrogenases/metabolism , Neoplasm Proteins/metabolism , Receptors, Prostaglandin/metabolism , Transcription Factors/biosynthesis , 3-Hydroxysteroid Dehydrogenases/genetics , Adult , Aged , Aged, 80 and over , Aldo-Keto Reductase Family 1 Member C3 , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Catalysis , Dinoprost/genetics , Female , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Middle Aged , Neoplasm Proteins/genetics , Receptors, Prostaglandin/genetics , Snail Family Transcription Factors , Transcription Factors/genetics
5.
Cell Oncol (Dordr) ; 38(3): 227-35, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25869021

ABSTRACT

PURPOSE: Krüppel-like factor 15 (KLF15) is a transcription factor that is involved in various biological processes, including cellular proliferation, differentiation and death. In addition, KLF15 has recently been implicated in the development of several human malignancies, including breast cancer. In vitro breast cancer studies have pointed at a putative role in the regulation of cell proliferation. As yet, however, KLF15 expression analyses in primary human breast cancers have not been reported. Here, we set out to investigate the clinical and biological significance of KLF15 expression in human breast cancers. METHODS: KLF15 expression was evaluated by immunohistochemistry in 54 primary invasive ductal breast carcinomas, and its status was correlated with various clinicopathological parameters. We also assessed KLF15 expression in vitro in 4 breast cancer-derived cell lines using Western blotting, and examined the effects of exogenous KLF15 expression on cell cycle progression using flow cytometry. Concomitant (changes in) p21, p27 and TOPO2A expression levels were examined using real-time RT-PCR and immunocytochemistry, respectively. RESULTS: In ~90% of the primary breast carcinoma tissues tested, KLF15 was found to be expressed and localized in either the cytoplasm, the nucleus or both. Predominant nuclear immunoreactivity was found to be associated with clinicopathological factors predicting a better clinical outcome (i.e., ER positive, HER2 negative, low grade, low Ki-67 expression). The breast cancer-derived cell lines tested showed a low KLF15 expression with a predominant cytoplasmic localization. Subsequent exogenous KLF15 over-expression resulted in a predominant nuclear localization and a concomitant decreased cellular proliferation and an arrest at the G0/G1 phase of the cell cycle. In addition, we found that nuclear KLF15 expression results in up-regulation of p21, a pivotal suppressor of the G1 to S phase transition of the cell cycle. CONCLUSIONS: Our results indicate that nuclear KLF15 expression suppresses breast cancer cell proliferation at least partially through p21 up-regulation and subsequent cell cycle arrest. This is a first study addressing the role of KLF15 in breast cancer development.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Genes, Tumor Suppressor , Kruppel-Like Transcription Factors/genetics , Nuclear Proteins/genetics , Adult , Aged , Aged, 80 and over , Blotting, Western , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Cell Cycle/genetics , Cell Line, Tumor , Female , Flow Cytometry , Humans , Immunohistochemistry , Kruppel-Like Transcription Factors/biosynthesis , Middle Aged , Nuclear Proteins/biosynthesis , Real-Time Polymerase Chain Reaction , Transfection
6.
Int J Biol Markers ; 30(2): e184-9, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25588857

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) is characterized by the absence of estrogen receptor, progesterone receptor and HER2. TNBCs are a diverse subgroup, but one promising marker and therapeutic target of this breast cancer is the androgen receptor (AR). Previously we demonstrated that AR and cognate intracrine pathways are associated with decreased proliferation in invasive ductal carcinoma with their decrease also detected between organ-confined and invasive diseases. Therefore, in this study, we examined the status of AR and androgen-producing enzymes during the process of metastasis to lymph nodes and cancer recurrence. MATERIALS AND METHODS: We studied 2 series of patients with TNBC, one from Kumamoto University Hospital composed of 16 matched cases of primary and locally or distal recurrences and the other from Tohoku University Hospital examining 46 lymph node metastasis from 23 patients. In addition to studying concordance in AR expression, we also examined the interactions between AR and Ki-67 labeling index and AR and site of distal metastasis. RESULTS: In both series, AR status was concordant between primary and recurrent/metastatic disease, but coordinated expression of AR and androgenic enzymes was lost during the process. The inverse association between AR and Ki-67, previously reported in invasive ductal carcinoma (IDC), was markedly potentiated in both lymph node and recurrent cancers. In addition, AR expression appeared to have little effect on visceral metastasis but was associated directly with bone metastasis and inversely with brain metastasis. CONCLUSIONS: The results of our present study demonstrated that AR remained in the majority of metastatic samples from AR-positive primary TNBCs and that AR manipulation could be exploited in the metastatic settings of TNBC.


Subject(s)
Receptors, Androgen/metabolism , Triple Negative Breast Neoplasms/blood , Female , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Triple Negative Breast Neoplasms/pathology
7.
Cancer Sci ; 105(11): 1503-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25230018

ABSTRACT

Invasive lobular carcinoma (ILC) accounts for approximately 10% of all breast carcinomas and is characterized by higher levels of androgen receptor (AR) compared to invasive ductal carcinoma (IDC). Despite this potentially androgen-responsive environment, the combined importance of AR and androgen metabolism in non-neoplastic lobules and lobular carcinoma remains unknown. Therefore, in this study, we evaluated the status of pivotal androgen-producing enzymes 17ß-hydroxysteroid dehydrogenase type 5 (17ßHSD5) and 5α-reductase type 1 (5αRed1) in 178 cases of ILC and surrounding histologically non-neoplastic lobular tissue using immunohistochemistry. Androgen receptor prevalence was higher but androgenic enzymes lower in ILC than non-neoplastic lobules. In ILC cases the status of 5αRed1 and 17ßHSD5 was inversely correlated with tumor size (P = 0.0053) and nuclear grade (P = 0.0290), and significantly associated with better overall survival of the patients (P = 0.0059). Based on these findings, we hypothesized that androgen signaling could act as a tumor suppressor. As previous studies suggested that androgens might partially act by increasing levels of the estrogen inactivating enzyme 17ß-hydroxysteroid dehydrogenase type 2 (17ßHSD2) in IDC tissues, this was reasonably considered a potential mechanism of androgen actions. Significantly positive correlation was detected between the status of androgenic enzymes and 17ßHSD2 (P < 0.0001) and intratumoral 17ßHSD2 was inversely correlated with tumor size in ILC (P = 0.0075). These correlations suggest one protective mode of androgen action could be through modulation of estrogen metabolism. Results of our present study indicated that androgen-producing enzymes could play pivotal protective roles in AR-enriched ILC cases.


Subject(s)
Androgens/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Carcinoma, Lobular/genetics , Estradiol Dehydrogenases/genetics , Estradiol Dehydrogenases/metabolism , Female , Humans , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Tumor Burden
8.
Breast Cancer Res Treat ; 145(2): 281-93, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24715382

ABSTRACT

One of the active intracellular pathways/networks in triple-negative breast carcinoma (TNBC) is that of the androgen receptor (AR). In this study, we examined AR and androgen-metabolising enzyme immunoreactivity in subcategories of TNBC to further elucidate the roles of androgenic pathways in TNBC. We utilised formalin-fixed paraffin-embedded breast cancer samples from ductal carcinoma in situ (DCIS) and invasive ductal carcinoma patient cohorts. We then used immunohistochemistry to classify these samples into basal-like and non-basal samples and to assess interactions between AR, androgen-metabolising enzymes and proliferation. To further substantiate our hypothesis and provide mechanistic insights, we also looked at the expression and regulation of these factors in publically available microarray data and in a panel of TNBC AR-positive cell lines. DCIS was associated with higher levels of AR and enzymes (p < 0.02), although a similar difference was not noticed in basal and non-basal samples. AR and enzymes were correlated in all states. In TNBC cell lines (MDA-MD-453, MFM-223 and SUM185-PE), we found that DHT treatment up-regulated 5αR1 and 17ßHSD5 suggesting a mechanistic explanation for the correlations observed in the histological samples. Publicly available microarray data in TNBC cases suggested similar patterns to those observed in histological samples. In the majority of settings, including publically available microarray data, an inverse association between AR and proliferation was detected. These findings suggest that decreases in AR and androgen-metabolising enzymes may be involved in the increased biological aggressiveness in TNBC development.


Subject(s)
Androgens/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Aldo-Keto Reductase Family 1 Member C3 , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Cell Proliferation , Cholestenone 5 alpha-Reductase/genetics , Cholestenone 5 alpha-Reductase/metabolism , Dihydrotestosterone/pharmacology , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Keratin-5/metabolism , Keratin-6/metabolism , Receptors, Androgen/metabolism , Triple Negative Breast Neoplasms/drug therapy
9.
Hum Pathol ; 44(10): 2338-45, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23953348

ABSTRACT

Androgen receptor and androgen metabolizing enzymes, 17ß-hydroxysteroid dehydrogenase type 5 (17ßHSD5) and 5α-reductase1 (5α1), are frequently detected in primary tumor of breast cancer, but their status in metastatic lymph nodes has not been examined. The biological role of androgen in breast cancer and its metastatic process also remain unknown. In this study, we used immunohistochemistry to localize the expression of androgen receptor, 17ßHSD5, and 5α1 in primary tumors and paired metastatic lymph nodes and correlated the findings with clinicopathologic factors of individual patients. Approximately 70% of primary tumors and paired metastatic lymph nodes expressed androgen receptor, with significant correlation between both lesions. However, 17ßHSD5 and 5α1 immunoreactivity was decreased in metastatic lymph nodes. Alone or in tandem with androgen receptor, 5α1 was associated with significantly lower Ki-67 index, lower pathologic grade, and higher estrogen receptor positivity, but androgen receptor/5α1 double positivity in lymph nodes was associated with larger lymph node metastasis and higher TNM stage. In conclusion, androgen receptor immunoreactivity remained stable during the process of metastasis, whereas androgen-metabolizing enzymes decreased. Although results of our study and previous reports imply additional roles of androgen metabolism in the metastasis process, especially conversion by 5α1, there may be divergence between its effects on primary tumor and those in metastatic lymph nodes.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Androgens/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Lymph Nodes/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Cell Proliferation , Female , Humans , Ki-67 Antigen/metabolism , Lymph Nodes/metabolism , Lymphatic Metastasis , Middle Aged , Neoplasm Staging , Receptors, Androgen/metabolism
10.
Cancer Sci ; 104(5): 639-46, 2013 May.
Article in English | MEDLINE | ID: mdl-23373898

ABSTRACT

Triple negative breast cancer (TNBC) is defined by estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 negativity. Patients with TNBC frequently undergo an aggressive clinical course due to the unavailability of specific targeted therapies. Androgen receptor (AR) was reported to be expressed in up to 60% of TNBC cases but there have been controversies as to the roles of androgen signaling through AR in TNBC. Therefore, in this study, we analyzed the status of AR in combination with androgen synthesizing enzymes (5α-reductase type 1 (5αR1) and 17ß-hydroxysteroid dehydrogenase type 5 (17ßHSD5)] in order to further understand androgenic actions in TNBC. Androgen receptor, 5αR1, and 17ßHSD5 were immunolocalized in a cohort of 203 TNBC patients from Thailand and Japan. We then correlated the findings with clinicopathological characteristics (age, stage, tumor diameter, lymph node invasion, metastatic spread, Ki-67 labeling index, disease-free survival, and overall survival) of the patients. Univariate analysis revealed that AR+/enzyme+ cases were associated with a significantly lower Ki-67 labeling index than AR-/enzyme- samples. Multivariate analysis indicated the presence of significant positive correlations between AR and enzyme status in tumor cells, and between tumor diameter, lymph node invasion, and distant metastasis. Significant negative correlations were also detected between Ki-67 labeling index and AR status (P = 0.04) or 5αR1 (P < 0.001). Cox proportional hazards analysis showed that Ki-67 labeling index and stage were the only factors predicting disease-free and overall survival of the patients, although univariate Kaplan-Meier analysis revealed AR/5αR1 negativity suggested a more adverse clinical course up to 80 months after surgery. These results suggest that the presence of androgen synthesizing pathways in addition to AR expression in tumor cells could confer a better clinical outcome through suppression of cell proliferation.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Receptors, Androgen/metabolism , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Cell Growth Processes/physiology , Disease-Free Survival , Female , Humans , Japan , Kaplan-Meier Estimate , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , Middle Aged , Receptors, Androgen/genetics , Thailand
SELECTION OF CITATIONS
SEARCH DETAIL
...