Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Molecules ; 28(23)2023 Nov 23.
Article in English | MEDLINE | ID: mdl-38067455

ABSTRACT

Mosla chinensis Maxim is an annual herb with many potential purposes in agricultural, industrial, and pharmaceutical fields. At present, the extract of the whole plant from M. chinensis has been proven to demonstrate antifungal, antioxidant, and anti-inflammatory activities. Previous studies focused on the enzyme pretreatment in hydrodistillation from M. chinensis. However, organic solvent or supercritical fluid carbon dioxide extraction (SFE-CO2) methods, which are commonly utilized in industry, have seldom been studied and cannot provide multiple evaluations of yield. In this work, we analysed compounds from M. chinensis by HPLC-DAD, discussed n-hexane extraction, and conducted further investigations on SFE-CO2 through the design of response surface methodology (RSM). The sample obtained from pilot-scale SFE-CO2 was also tested against nine kinds of microorganisms. Single-factor results revealed that the extraction rates from M. chinensis by steam distillation, n-hexane extraction, and SFE-CO2 were 1%, 2.09%, and 3.26%, respectively. RSM results showed a significant improvement in extraction rate through optimising pressure and time, and the interaction of both factors was more important than that of temperature-pressure and temperature-time. A pilot-scale test with an extraction rate of 3.34% indicated that the predicted RSM condition was operable. In addition, samples from the pilot-scale SFE-CO2 showed antibacterial effects against three previously unreported bacteria (Gardnerella vaginalis, methicillin-resistant Staphylococcus aureus, and Propionibacterium acnes). These results fill the gap in previous research and provide more information for the application and development of M. chinensis in the future.


Subject(s)
Chromatography, Supercritical Fluid , Lamiaceae , Methicillin-Resistant Staphylococcus aureus , Oils, Volatile , Oils, Volatile/analysis , Carbon Dioxide , Chromatography, High Pressure Liquid , Chromatography, Supercritical Fluid/methods
2.
ACS Omega ; 8(3): 3176-3183, 2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36713698

ABSTRACT

As novel nanomaterials developed gradually with nanotechnology, carbon dots have been widely applied in medical applications, including disease treatment, drug delivery, antibacterial applications, and phototherapy. Based on the similar process between Chinese medicinal materials for hemostasis and modern carbon dots, this paper reports the preparation of four luminescent carbon dots with Chinese medicinal materials (plants and animals) as carbon sources and the investigation on their hemostatic effects in vitro and in rat bleeding models. It is found that the four studied carbon dots exhibit similar hemostatic effects and hemostatic mechanisms through impacting both endogenous and exogenous coagulation pathways. In addition, these carbon dots all exhibit anti-inflammatory effects and good biocompatibility, ensuring their potential in pretraumatic fields. This work provides a new perspective for hemostatic carbon dots prepared using carbonized natural plants and animals and new ideas for the research of new hemostatic materials.

3.
Int J Biol Sci ; 18(10): 4203-4218, 2022.
Article in English | MEDLINE | ID: mdl-35844787

ABSTRACT

Rationale: Triple-negative breast cancer (TNBC) does not respond to anti-estrogen and anti-HER2 therapies and is commonly treated by chemotherapy. TNBC has a high recurrence rate, particularly within the first 3 years. Thus, there is an urgent clinical need to develop more effective therapies for TNBC. Topoisomerase I (TOP1) inhibitors cause DNA damage, making these drugs desirable for TNBC treatment since DNA repair machinery is defective in this subtype of breast cancer. Among the main molecular subtypes of breast cancer, the TNBC cell lines exhibited the highest TOP1 inhibition sensitivity. However, clinically used TOP1 inhibitors, such as topotecan and irinotecan, have shown limited clinical applications and the reasons remain unclear. Understanding the mechanism of differential responses to TOP1 blockade and identifying the predictive markers for cancer cell sensitivity will help further TOP1-targeted therapy for TNBC treatment and improve the clinical use of TOP1 inhibitors. Methods: Viability assays were used to evaluate breast cancer cell sensitivity to topotecan and other TOP1 inhibitors as well as TOP2 inhibitors. An in vitro-derived topotecan-resistant TNBC cell model and TNBC xenograft models were employed to confirm cancer cell response to TOP1 blockade. RNA-seq was used to identify potential predictive markers for TNBC cell response to TOP1 blockade. Western blotting and qRT-PCR were performed to measure the protein levels and RNA expression. ATAC-seq and luciferase reporter assays were used to examine MYC transcriptional regulations. The effects of MYC and JNK in cancer cell response to TOP1 inhibition were validated via loss-of-function and gain-of-function experiments. Results: We observed two distinct and diverging cancer cell responses - sensitive versus resistant to TOP1 inhibition, which was confirmed by TNBC xenograft mouse models treated by topotecan. TNBC cells exhibited bifurcated temporal patterns of ATR pathway activation upon TOP1 inhibitor treatment. The sensitive TNBC cells showed an "up then down" dynamic pattern of ATR/Chk1 signaling, while the resistant TNBC cells exhibited a "persistently up" profile. On the contrary, opposite temporal patterns of induced expression of MYC, a key regulator and effector of DNA damage, were found in TNBC cells treated by TOP1 inhibitors. Mechanistically, we showed that TOP1-induced JNK signaling upregulated MYC expression. Furthermore, pharmacological inhibition of ATR reversed TNBC cell resistance to topotecan, whereas MYC knockdown and JNK inhibition reduced cancer cell sensitivity. Conclusions: Dynamic temporal profiles of induced ATR/Chk1 and JNK activation as well as MYC expression, may predict cancer cell response to TOP1 inhibitors. JNK activation-mediated constitutive elevation of MYC expression may represent a novel mechanism governing cancer cell sensitivity to TOP1-targeting therapy. Our results may provide implications for identifying TNBC patients who might benefit from the treatment with TOP1 inhibitors.


Subject(s)
DNA Topoisomerases, Type I , Triple Negative Breast Neoplasms , Animals , Cell Line, Tumor , Cell Proliferation , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type I/pharmacology , DNA Topoisomerases, Type I/therapeutic use , Humans , Mice , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction/genetics , Topotecan/pharmacology , Topotecan/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
4.
RSC Adv ; 12(19): 11640-11648, 2022 Apr 13.
Article in English | MEDLINE | ID: mdl-35432945

ABSTRACT

Carbon dots (CDs) have attracted much attention in theoretical researches and their practical applications due to their excellent optical properties, and many researchers discovered that flurophores play a very important role in synthesis process of CDs and the luminescence of prepared CDs. In this study, two CDs were pyrolysis with citric acid, N-acetyl-l-cysteine and glutathione derivatives as carbon sources. Four intermediate small molecules were separated from the prepared CDs through ultrafiltration and chromatography, and their chemical structures were determined. The formation process of CDs was monitored through identified small molecule intermediates and HPLC. It is speculated that the two CDs have the same formation pathway, including TPA (5-oxo-2,3-dihydro-5H-[1,3]thiazolo[3,2-a]pyridine-3,7-dicarboxylic acid) synthesis, fluorophore polymerization, carbon chain extension, and carbonization. It was also discovered that these two CDs have the same fluorescence properties, thiazolopyridone structure, and nitrogen-sulfur co-doped functional groups are important reasons for the mixed excitation dependence of CDs. This study would provide valuable theoretical basis for the studies on preparation of excellent CDs, raw material selection, and CDs formation mechanism.

5.
Mol Ther ; 30(2): 672-687, 2022 02 02.
Article in English | MEDLINE | ID: mdl-34274535

ABSTRACT

Triple-negative breast cancer (TNBC) has a high propensity for organ-specific metastasis. However, the underlying mechanisms are not well understood. Here we show that the primary TNBC tumor-derived C-X-C motif chemokines 1/2/8 (CXCL1/2/8) stimulate lung-resident fibroblasts to produce the C-C motif chemokines 2/7 (CCL2/7), which, in turn, activate cholesterol synthesis in lung-colonizing TNBC cells and induce angiogenesis at lung metastatic sites. Inhibiting cholesterol synthesis in lung-colonizing breast tumor cells by pulmonary administration of simvastatin-carrying HER3-targeting nanoparticles reduces angiogenesis and growth of lung metastases in a syngeneic TNBC mouse model. Our findings reveal a novel, chemokine-regulated mechanism for the cholesterol synthesis pathway and a critical role of metastatic site-specific cholesterol synthesis in the pulmonary tropism of TNBC metastasis. The study has implications for the unresolved epidemiological observation that use of cholesterol-lowering drugs has no effect on breast cancer incidence but can unexpectedly reduce breast cancer mortality, suggesting interventions of cholesterol synthesis in lung metastases as an effective treatment to improve survival in individuals with TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Animals , Cell Line, Tumor , Chemokines , Humans , Lung/metabolism , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics
6.
Sci Rep ; 11(1): 2408, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33510281

ABSTRACT

Triple-negative breast cancer (TNBC) is associated with a high rate of early recurrence and distant metastasis, frequent development of therapeutic resistance, and a poor prognosis. There is a lack of targeted therapies for this aggressive subtype of breast cancer. Identifying novel effective treatment modalities for TNBC remains an urgent and unmet clinical need. In this study, we investigated the anti-cancer effect of triptonide, a natural compound derived from the traditional Chinese medicinal herb Tripterygium wilfordii Hook F, in TNBC. We found that triptonide inhibits human TNBC cell growth in vitro and growth of TNBC xenograft mammary tumors. It induces apoptosis and suppresses stem-like properties as indicated by reduced mammosphere formation and aldehyde dehydrogenase activity in TNBC cells. We show that triptonide downregulates multiple cancer stem cell-associated genes but upregulates SNAI1 gene expression. In support of SNAI1 induction as a negative feedback response to triptonide treatment, in vitro-derived triptonide-resistant HCC1806 cells display a markedly higher expression of SNAI1 compared with parental cells. Mechanistically, the increase of SNAI1 expression is mediated by the activation of JNK signaling, but not by ERK and AKT, two well-established SNAI1 regulators. Furthermore, knockdown of SNAI1 in the triptonide-resistant HCC1806 cells increases sensitivity to triptonide and reduces mammosphere formation. These results indicate that triptonide holds promise as a novel anti-tumor agent for TNBC treatment. Our study also reveals a SNAI1-associated feedback mechanism which may lead to acquired resistance to triptonide.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Discovery , Triterpenes/pharmacology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Gene Expression , Gene Knockdown Techniques , Humans , Mice , Molecular Structure , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triterpenes/chemistry , Triterpenes/therapeutic use , Xenograft Model Antitumor Assays
7.
PLoS One ; 15(6): e0232068, 2020.
Article in English | MEDLINE | ID: mdl-32559187

ABSTRACT

Cyclin Dependent Kinase 9 (CDK9) associates with Bromodomain and Extra-Terminal Domain (BET) proteins to promote transcriptional elongation by phosphorylation of serine 2 of RNAP II C-terminal domain. We examined the therapeutic potential of selective CDK9 inhibitors (AZD 4573 and MC180295) against human multiple myeloma cells in vitro. Short-hairpin RNA silencing of CDK9 in Multiple Myeloma (MM) cell lines reduced cell viability compared to control cells showing the dependency of MM cells on CDK9. In order to explore synergy with the CDK9 inhibitor, proteolysis targeting chimeric molecule (PROTAC) ARV 825 was added. This latter drug causes ubiquitination of BET proteins resulting in their rapid and efficient degradation. Combination treatment of MM cells with ARV 825 and AZD 4573 markedly reduced their protein expression of BRD 2, BRD 4, MYC and phosphorylated RNA pol II as compared to each single agent alone. Combination treatment synergistically inhibited multiple myeloma cells both in vitro and in vivo with insignificant weight loss. The combination also resulted in marked increase of apoptotic cells at low dose compared to single agent alone. Taken together, our studies show for the first time that the combination of a BET PROTAC (ARV 825) plus AZD 4573 (CDK9 inhibitor) is effective against MM cells.


Subject(s)
Cyclin-Dependent Kinase 9/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proteins/metabolism , Proteolysis/drug effects , Animals , Azepines/pharmacology , Azepines/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Down-Regulation/drug effects , Drug Synergism , Humans , Mice , Mice, SCID , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Protein Kinase Inhibitors/therapeutic use , Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA Polymerase II/metabolism , RNA, Small Interfering/metabolism , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Thalidomide/therapeutic use , Transplantation, Heterologous
8.
Oncotarget ; 9(14): 11503-11514, 2018 Feb 20.
Article in English | MEDLINE | ID: mdl-29545915

ABSTRACT

PURPOSE: Conditional reprogramming methods allow for the inexhaustible in vitro proliferation of primary epithelial cells from human tissue specimens. This methodology has the potential to enhance the utility of primary cell culture as a model for mammary gland research. However, few studies have systematically characterized this method in generating in vitro normal human mammary epithelial cell models. RESULTS: We show that cells derived from fresh normal breast tissues can be propagated and exhibit heterogeneous morphologic features. The cultures are composed of CK18, desmoglein 3, and CK19-positive luminal cells and vimentin, p63, and CK14-positive myoepithelial cells, suggesting the maintenance of in vivo heterogeneity. In addition, the cultures contain subpopulations with different CD49f and EpCAM expression profiles. When grown in 3D conditions, cells self-organize into distinct structures that express either luminal or basal cell markers. Among these structures, CK8-positive cells enclosing a lumen are capable of differentiation into milk-producing cells in the presence of lactogenic stimulus. Furthermore, our short-term cultures retain the expression of ERα, as well as its ability to respond to estrogen stimulation. MATERIALS AND METHODS: We have investigated conditionally reprogrammed normal epithelial cells in terms of cell type heterogeneity, cellular marker expression, and structural arrangement in two-dimensional (2D) and three-dimensional (3D) systems. CONCLUSIONS: The conditional reprogramming methodology allows generation of a heterogeneous culture from normal human mammary tissue in vitro. We believe that this cell culture model will provide a valuable tool to study mammary cell function and malignant transformation.

9.
Cancer Biol Ther ; 19(10): 858-868, 2018.
Article in English | MEDLINE | ID: mdl-29580128

ABSTRACT

Distant metastasis accounts for the vast majority of deaths in patients with cancer. Breast cancer exhibits a distinct metastatic pattern commonly involving bone, liver, lung, and brain. Breast cancer can be divided into different subtypes based on gene expression profiles, and different breast cancer subtypes show preference to distinct organ sites of metastasis. Luminal breast tumors tend to metastasize to bone while basal-like breast cancer (BLBC) displays a lung tropism of metastasis. However, the mechanisms underlying this organ-specific pattern of metastasis still remain to be elucidated. In this review, we will summarize the recent advances regarding the molecular signaling pathways as well as the therapeutic strategies for treating breast cancer lung metastasis.


Subject(s)
Breast Neoplasms/pathology , Lung Neoplasms/secondary , Biomarkers , Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Cytokines/metabolism , Female , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/therapy , Neoplasm Grading , Neoplasm Staging , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction , Tumor Microenvironment
10.
J Control Release ; 271: 127-138, 2018 02 10.
Article in English | MEDLINE | ID: mdl-29288681

ABSTRACT

Resistance to anti-tumor therapeutics is an important clinical problem. Tumor-targeted therapies currently used in the clinic are derived from antibodies or small molecules that mitigate growth factor activity. These have improved therapeutic efficacy and safety compared to traditional treatment modalities but resistance arises in the majority of clinical cases. Targeting such resistance could improve tumor abatement and patient survival. A growing number of such tumors are characterized by prominent expression of the human epidermal growth factor receptor 3 (HER3) on the cell surface. This study presents a "Trojan-Horse" approach to combating these tumors by using a receptor-targeted biocarrier that exploits the HER3 cell surface protein as a portal to sneak therapeutics into tumor cells by mimicking an essential ligand. The biocarrier used here combines several functions within a single fusion protein for mediating targeted cell penetration and non-covalent self-assembly with therapeutic cargo, forming HER3-homing nanobiologics. Importantly, we demonstrate here that these nanobiologics are therapeutically effective in several scenarios of resistance to clinically approved targeted inhibitors of the human EGF receptor family. We also show that such inhibitors heighten efficacy of our nanobiologics on naïve tumors by augmenting HER3 expression. This approach takes advantage of a current clinical problem (i.e. resistance to growth factor inhibition) and uses it to make tumors more susceptible to HER3 nanobiologic treatment. Moreover, we demonstrate a novel approach in addressing drug resistance by taking inhibitors against which resistance arises and re-introducing these as adjuvants, sensitizing tumors to the HER3 nanobiologics described here.


Subject(s)
Antineoplastic Agents/administration & dosage , Biological Products/administration & dosage , Drug Carriers/administration & dosage , Drug Resistance, Neoplasm/drug effects , Nanoparticles/administration & dosage , Peptides/administration & dosage , Receptor, ErbB-3/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Female , Humans , Mice , Neoplasms/drug therapy
11.
Oncogene ; 37(10): 1399-1408, 2018 03.
Article in English | MEDLINE | ID: mdl-29249801

ABSTRACT

Triple-negative breast cancer (TNBC) has high rates of local recurrence and distant metastasis, partially due to its high invasiveness. The Forkhead box C1 (FOXC1) transcription factor has been shown to be specifically overexpressed in TNBC and associated with poor clinical outcome. How TNBC's high invasiveness is driven by FOXC1 and its downstream targets remains poorly understood. In the present study, pathway-specific PCR array assays revealed that WNT5A and matrix metalloproteinase-7 (MMP7) were upregulated by FOXC1 in TNBC cells. Interestingly, WNT5A mediates the upregulation of MMP7 by FOXC1 and the WNT5A-MMP7 axis is essential for FOXC1-induced invasiveness of TNBC cells in vitro. Xenograft models showed that the lung extravasation and metastasis of FOXC1-overexpressing TNBC cells were attenuated by knocking out WNT5A, but could be restored by MMP7 overexpression. Mechanistically, FOXC1 can bind directly to the WNT5A promoter region to activate its expression. Engineered DNA-binding molecule-mediated chromatin immunoprecipitation (enChIP), coupled with mass spectrometry, identified FOXC1-interacting proteins including a group of heterogeneous nuclear ribonucleoproteins involved in WNT5A transcription induction. Finally, we found that WNT5A activates NF-κB signaling to induce MMP7 expression. Collectively, these data demonstrate a FOXC1-elicited non-canonical WNT5A signaling mechanism comprising NF-κB and MMP7 that is essential for TNBC cell invasiveness, thereby providing implications toward developing an effective therapy for TNBC.


Subject(s)
Forkhead Transcription Factors/physiology , Matrix Metalloproteinase 7/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Wnt-5a Protein/genetics , Animals , Cell Line, Tumor , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Matrix Metalloproteinase 7/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Signal Transduction/genetics , Transcriptional Activation , Wnt-5a Protein/metabolism
12.
Sci Rep ; 7(1): 14017, 2017 10 25.
Article in English | MEDLINE | ID: mdl-29070831

ABSTRACT

The forkhead box transcription factor FOXC1 plays a critical role in embryogenesis and the development of many organs. Its mutations and high expression are associated with many human diseases including breast cancer. Although FOXC1 knockout mouse studies showed that it is not required for mammary gland development during puberty, it is not clear whether its overexpression alters normal mammary development in vivo. To address this question, we generated transgenic mice with mammary-specific FOXC1 overexpression. We report that transgenic FOXC1 overexpression suppresses lobuloalveologenesis and lactation in mice. This phenotype is associated with higher percentages of estrogen receptor-, progesterone receptor-, or ki67-positive mammary epithelial cells in the transgenic mice at the lactation stage. We also show that expression of the Elf5 transcription factor, a master regulator of mammary alveologenesis and luminal cell differentiation, is markedly reduced in mammary epithelial cells of transgenic mice. Likewise, levels of activated Stat5, another inducer of alveolar expansion and a known mediator of the Elf5 effect, are also lowered in those cells. In contrast, the cytokeratin 8-positive mammary cell population with progenitor properties is elevated in the transgenic mice at the lactation stage, suggesting inhibition of mammary cell differentiation. These results may implicate FOXC1 as a new important regulator of mammary gland development.


Subject(s)
Forkhead Transcription Factors/physiology , Mammary Glands, Animal/metabolism , Animals , Cell Differentiation , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Keratin-8/metabolism , Ki-67 Antigen/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/physiology , Mice , Mice, Transgenic , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , STAT5 Transcription Factor/metabolism , Transcription Factors/metabolism
13.
Cell Commun Signal ; 15(1): 22, 2017 06 19.
Article in English | MEDLINE | ID: mdl-28629477

ABSTRACT

BACKGROUND: The pathogenesis of human basal-like breast cancer (BLBC) is not well understood and patients with BLBC have a poor prognosis. Expression of the epidermal growth factor receptor (EGFR) and nuclear factor-κB (NF-κB) is well-known to be upregulated in BLBC. The forkhead box C1 (FOXC1) transcription factor, an important prognostic biomarker specific for BLBC, has been shown to be induced by EGF and is critical for EGF effects in breast cancer cells. How FOXC1 is transcriptionally activated in BLBC is not clear. METHODS: Luciferase reporter assays were performed to show that NF-κB-p65 enhances FOXC1 promoter activity in BLBC cells (MDA-MB-468). Electrophoretic mobility shift assay, biotinylated oligonucleotide precipitation assay, and chromatin immunoprecipitation assay were used to show that NF-κB interacts and binds to the promoter region of FOXC1. RESULTS: In this study, we demonstrate that NF-κB is a pivotal mediator of the EGF/EGFR regulation of FOXC1 expression by binding to the FOXC1 promoter to activate FOXC1 transcription. Loss or inhibition of NF-κB diminished FOXC1 expression. CONCLUSION: Collectively, our findings reveal a novel EGFR-NF-κB-FOXC1 signaling axis that is critical for BLBC cell function, supporting the notion that intervention in the FOXC1 pathway may provide potential modalities for BLBC treatment.


Subject(s)
Breast Neoplasms/pathology , Epidermal Growth Factor/metabolism , Forkhead Transcription Factors/metabolism , NF-kappa B/metabolism , Signal Transduction , Base Sequence , Cell Line, Tumor , Forkhead Transcription Factors/genetics , Humans , Promoter Regions, Genetic/genetics
14.
Stem Cell Reports ; 8(2): 205-215, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28132888

ABSTRACT

Human induced pluripotent stem cells (iPSCs) can give rise to multiple cell types and hold great promise in regenerative medicine and disease-modeling applications. We have developed a reliable two-step protocol to generate human mammary-like organoids from iPSCs. Non-neural ectoderm-cell-containing spheres, referred to as mEBs, were first differentiated and enriched from iPSCs using MammoCult medium. Gene expression profile analysis suggested that mammary gland function-associated signaling pathways were hallmarks of 10-day differentiated mEBs. We then generated mammary-like organoids from 10-day mEBs using 3D floating mixed gel culture and a three-stage differentiation procedure. These organoids expressed common breast tissue, luminal, and basal markers, including estrogen receptor, and could be induced to produce milk protein. These results demonstrate that human iPSCs can be directed in vitro toward mammary lineage differentiation. Our findings provide an iPSC-based model for studying regulation of normal mammary cell fate and function as well as breast disease development.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Organoids , Biomarkers , Cell Culture Techniques , Cell Differentiation/genetics , Ectoderm/cytology , Ectoderm/embryology , Embryoid Bodies/cytology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/metabolism , Organoids/cytology , Regenerative Medicine , Transcriptome
15.
Oncotarget ; 7(46): 75729-75738, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27708239

ABSTRACT

Breast cancers arising in the setting of the hereditary breast cancer genes BRCA1 and BRCA2 are most commonly classified as basal-like breast cancer (BLBC) or luminal breast cancer, respectively. BLBC is an aggressive subtype of breast cancer associated with liver and lung metastases and poorer prognosis than other subtypes and for which chemotherapy is the only systemic therapy. Multiple immunohistochemical markers are used to identify the basal-like subtype, including the absence of estrogen receptor alpha, progesterone receptor, and human epidermal growth factor receptor 2. Forkhead box C1 (FOXC1) has been identified as a specific marker expressed in BLBC in general breast cancer cohorts. We examined an institutional cohort of breast cancer patients with germline BRCA1 (n=46) and BRCA2 (n=35) mutations and found that FOXC1 expression on immunohistochemical staining is associated with BRCA1 vs BRCA2 mutations [30/46 vs. 6/35]. In BRCA1 mutant tumors, FOXC1 was expressed in 28/31 BLBC tumors and 2/13 non-BLBC tumors, In BRCA2 mutant tumors, FOXC1 was expressed in 5/5 BLBC tumors and 1/30 non-BLBC tumors. In cell culture models of BRCA1-mutant breast cancer, FOXC1 is associated with increased proliferation and may serve as a marker for sensitivity to PARP-inhibitor therapy with olaparib.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms/genetics , Forkhead Transcription Factors/genetics , Neoplasms, Basal Cell/genetics , Adult , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Gene Knockout Techniques , Genes, BRCA1 , Genes, BRCA2 , Humans , Immunohistochemistry , Middle Aged , Mutation , Neoplasm Grading , Neoplasm Staging , Neoplasms, Basal Cell/metabolism , Neoplasms, Basal Cell/pathology , Phthalazines/pharmacology , Piperazines/pharmacology
16.
Sci Rep ; 6: 32007, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27550649

ABSTRACT

Surface ectoderm (SE) cells give rise to structures including the epidermis and ectodermal associated appendages such as hair, eye, and the mammary gland. In this study, we validate a protocol that utilizes BMP4 and the γ-secretase inhibitor DAPT to induce SE differentiation from human induced pluripotent stem cells (hiPSCs). hiPSC-differentiated SE cells expressed markers suggesting their commitment to the SE lineage. Computational analyses using integrated quantitative transcriptomic and proteomic profiling reveal that TGFß superfamily signaling pathways are preferentially activated in SE cells compared with hiPSCs. SE differentiation can be enhanced by selectively blocking TGFß-RI signaling. We also show that SE cells and neural ectoderm cells possess distinct gene expression patterns and signaling networks as indicated by functional Ingenuity Pathway Analysis. Our findings advance current understanding of early human SE cell development and pave the way for modeling of SE-derived tissue development, studying disease pathogenesis, and development of regenerative medicine approaches.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Diamines/pharmacology , Ectoderm/cytology , Gene Expression Profiling/methods , Induced Pluripotent Stem Cells/cytology , Proteomics/methods , Thiazoles/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Ectoderm/drug effects , Ectoderm/metabolism , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
17.
Mol Cell Oncol ; 3(3): e1131668, 2016 May.
Article in English | MEDLINE | ID: mdl-27314088

ABSTRACT

The Forkhead box C1 (FOXC1) transcriptional factor is a critical biomarker for basal-like breast cancer (BLBC). We recently reported that FOXC1 promotes cancer stem cell properties in BLBC by activating Smoothened (SMO)-independent Hedgehog (Hh) signaling, suggesting a FOXC1-mediated mechanism for BLBC cell function and anti-Hh therapy resistance.

18.
Cell Rep ; 13(5): 1046-58, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26565916

ABSTRACT

The mesoderm- and epithelial-mesenchymal transition-associated transcription factor FOXC1 is specifically overexpressed in basal-like breast cancer (BLBC), but its biochemical function is not understood. Here, we demonstrate that FOXC1 controls cancer stem cell (CSC) properties enriched in BLBC cells via activation of Smoothened (SMO)-independent Hedgehog (Hh) signaling. This non-canonical activation of Hh is specifically mediated by Gli2. Furthermore, we show that the N-terminal domain of FOXC1 (aa 1-68) binds directly to an internal region (aa 898-1168) of Gli2, enhancing the DNA-binding and transcription-activating capacity of Gli2. FOXC1 expression correlates with that of Gli2 and its targets in human breast cancers. Moreover, FOXC1 overexpression reduces sensitivity to anti-Hedgehog (Hh) inhibitors in BLBC cells and xenograft tumors. Together, these findings reveal FOXC1-mediated non-canonical Hh signaling that determines the BLBC stem-like phenotype and anti-Hh sensitivity, supporting inhibition of FOXC1 pathways as potential approaches for improving BLBC treatment.


Subject(s)
Breast Neoplasms/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/metabolism , Nuclear Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Forkhead Transcription Factors/chemistry , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/chemistry , Nuclear Proteins/chemistry , Protein Binding , Signal Transduction , Smoothened Receptor , Zinc Finger Protein Gli2
19.
Clin Breast Cancer ; 15(6): 448-457.e2, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26248960

ABSTRACT

BACKGROUND: We investigated the effect of basal protein expression on trastuzamab response in patients with HER2-positive (HER2(+)) breast cancer who received trastuzamab (T) and in HER2(+) breast cancer cell lines. PATIENTS AND METHODS: Expression of cytokeratin (CK) 5/6, CK14, and epidermal growth factor receptor (EGFR) was evaluated after immunohistochemical staining in paraffin-embedded tissue of 97 patients with stage I to III HER2(+) breast cancer treated with chemotherapy/T. Groups with and without basal protein expression were compared with respect to clinicopathologic parameters and survival. We treated 4 cell lines (2 basal-HER2 [HCC1569, HCC1954] and 2 nonbasal HER2 [BT474, SKBR3]) each with vehicle, T 20 µg/mL, paclitaxel 0.01 µM (P), and T with P (T + P). Cell viability was assessed and HER2 pathway suppression was compared between groups using immunoblot analysis. Mammosphere formation was used to assess breast cancer stem cell properties. RESULTS: EGFR expression was significantly associated with cancer-specific survival (CSS) (P = .05). CK5/6 expression strongly correlated with overall and disease-free survival, and CSS (P = .03, P = .04, and P = .03, respectively). Statistical significance was maintained for EGFR and CK5/6 after adjustment for covariates. CK14 was not associated with survival. All cell lines expressed similar levels of HER2. T and P alone inhibited proliferation of nonbasal cell lines; T + P had an additive cytotoxic effect. Basal cells were resistant to T, P inhibited proliferation, but T + P had no additive cytotoxic effect on cell growth in basal cells. Immunoblot analysis showed a significant decrease in phosphorylated Akt levels after treatment with T or T + P in nonbasal cells but not in basal cells. Akt blockade suppressed growth of basal and nonbasal HER2(+) cells. Furthermore, basal HER2 cell lines had increased mammosphere formation, which suggests increased stem cell properties compared with nonbasal HER2 cell lines. CONCLUSION: CK5/6 and EGFR expression are predictive of worse prognosis in HER2(+) breast cancer patients treated with T. Basal HER2 breast cancer cell lines are resistant to trastuzamab, which is mediated through the Akt pathway; AKT inhibition abrogates this resistance. Basal HER2 cell lines also have increased stem cell properties, which might play a role in the resistance pathway.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/physiology , ErbB Receptors/biosynthesis , Keratins/biosynthesis , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Disease-Free Survival , ErbB Receptors/analysis , Female , Humans , Immunoblotting , Immunohistochemistry , Kaplan-Meier Estimate , Keratins/analysis , Middle Aged , Neoplasm Invasiveness , Phenotype , Proportional Hazards Models , Receptor, ErbB-2/antagonists & inhibitors , Trastuzumab/therapeutic use , Treatment Outcome
20.
PLoS One ; 10(7): e0131285, 2015.
Article in English | MEDLINE | ID: mdl-26147507

ABSTRACT

Breast cancer is the most common cancer in women and a leading cause of cancer-related deaths for women worldwide. Various cell models have been developed to study breast cancer tumorigenesis, metastasis, and drug sensitivity. The MCF10A human mammary epithelial cell line is a widely used in vitro model for studying normal breast cell function and transformation. However, there is limited knowledge about whether MCF10A cells reliably represent normal human mammary cells. MCF10A cells were grown in monolayer, suspension (mammosphere culture), three-dimensional (3D) "on-top" Matrigel, 3D "cell-embedded" Matrigel, or mixed Matrigel/collagen I gel. Suspension culture was performed with the MammoCult medium and low-attachment culture plates. Cells grown in 3D culture were fixed and subjected to either immunofluorescence staining or embedding and sectioning followed by immunohistochemistry and immunofluorescence staining. Cells or slides were stained for protein markers commonly used to identify mammary progenitor and epithelial cells. MCF10A cells expressed markers representing luminal, basal, and progenitor phenotypes in two-dimensional (2D) culture. When grown in suspension culture, MCF10A cells showed low mammosphere-forming ability. Cells in mammospheres and 3D culture expressed both luminal and basal markers. Surprisingly, the acinar structure formed by MCF10A cells in 3D culture was positive for both basal markers and the milk proteins ß-casein and α-lactalbumin. MCF10A cells exhibit a unique differentiated phenotype in 3D culture which may not exist or be rare in normal human breast tissue. Our results raise a question as to whether the commonly used MCF10A cell line is a suitable model for human mammary cell studies.


Subject(s)
Breast/cytology , Cell Line/cytology , Epithelial Cells/cytology , Animals , Biomarkers/analysis , Cell Adhesion , Cell Culture Techniques , Cell Line/chemistry , Cell Line/drug effects , Cell Shape , Collagen , Collagen Type I , Culture Media/pharmacology , Drug Combinations , Epithelial Cells/chemistry , Epithelial Cells/drug effects , Female , Intercellular Signaling Peptides and Proteins/pharmacology , Keratins/analysis , Laminin , Membrane Proteins/analysis , Milk Proteins/analysis , Phenotype , Proteoglycans , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Stem Cells/chemistry , Stem Cells/cytology , Vimentin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...