Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 624
Filter
1.
Nature ; 599(7883): 147-151, 2021 11.
Article in English | MEDLINE | ID: mdl-34616045

ABSTRACT

Understanding cellular architecture is essential for understanding biology. Electron microscopy (EM) uniquely visualizes cellular structures with nanometre resolution. However, traditional methods, such as thin-section EM or EM tomography, have limitations in that they visualize only a single slice or a relatively small volume of the cell, respectively. Focused ion beam-scanning electron microscopy (FIB-SEM) has demonstrated the ability to image small volumes of cellular samples with 4-nm isotropic voxels1. Owing to advances in the precision and stability of FIB milling, together with enhanced signal detection and faster SEM scanning, we have increased the volume that can be imaged with 4-nm voxels by two orders of magnitude. Here we present a volume EM atlas at such resolution comprising ten three-dimensional datasets for whole cells and tissues, including cancer cells, immune cells, mouse pancreatic islets and Drosophila neural tissues. These open access data (via OpenOrganelle2) represent the foundation of a field of high-resolution whole-cell volume EM and subsequent analyses, and we invite researchers to explore this atlas and pose questions.


Subject(s)
Datasets as Topic , Information Dissemination , Microscopy, Electron, Scanning , Organelles/ultrastructure , Animals , Cell Line , Cells, Cultured , Drosophila melanogaster/cytology , Drosophila melanogaster/ultrastructure , Female , Golgi Apparatus/ultrastructure , Humans , Interphase , Islets of Langerhans/cytology , Male , Mice , Microscopy, Electron, Scanning/methods , Microscopy, Electron, Scanning/standards , Microtubules/ultrastructure , Neuroglia/ultrastructure , Neurons/ultrastructure , Open Access Publishing , Ovarian Neoplasms/immunology , Ovarian Neoplasms/ultrastructure , Ribosomes/ultrastructure , Synaptic Vesicles/ultrastructure , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/ultrastructure
2.
Sci Rep ; 10(1): 11913, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32681052

ABSTRACT

The majority of women with recurrent ovarian cancer (OvCa) develop malignant ascites with volumes that can reach > 2 L. The resulting elevation in intraperitoneal pressure (IPP), from normal values of 5 mmHg to as high as 22 mmHg, causes striking changes in the loading environment in the peritoneal cavity. The effect of ascites-induced changes in IPP on OvCa progression is largely unknown. Herein we model the functional consequences of ascites-induced compression on ovarian tumor cells and components of the peritoneal microenvironment using a panel of in vitro, ex vivo and in vivo assays. Results show that OvCa cell adhesion to the peritoneum was increased under compression. Moreover, compressive loads stimulated remodeling of peritoneal mesothelial cell surface ultrastructure via induction of tunneling nanotubes (TNT). TNT-mediated interaction between peritoneal mesothelial cells and OvCa cells was enhanced under compression and was accompanied by transport of mitochondria from mesothelial cells to OvCa cells. Additionally, peritoneal collagen fibers adopted a more linear anisotropic alignment under compression, a collagen signature commonly correlated with enhanced invasion in solid tumors. Collectively, these findings elucidate a new role for ascites-induced compression in promoting metastatic OvCa progression.


Subject(s)
Ascites/pathology , Ovarian Neoplasms/pathology , Peritoneum/pathology , Tumor Microenvironment , Animals , Anisotropy , Cell Adhesion , Cell Line, Tumor , Collagen/metabolism , Epithelium/pathology , Epithelium/ultrastructure , Female , Humans , Mice, Inbred C57BL , Mitochondria/pathology , Models, Biological , Nanotubes/chemistry , Nanotubes/ultrastructure , Neoplasm Metastasis , Ovarian Neoplasms/ultrastructure , Peritoneum/ultrastructure
3.
Ultrastruct Pathol ; 44(2): 193-202, 2020 Mar 03.
Article in English | MEDLINE | ID: mdl-32183603

ABSTRACT

Ovarian cancer is the seventh most common cancer worldwide in women. Many anticancer drugs are currently used clinically have been isolated from plant species or are based on such substances. Thymol (5-methyl-2-isopropylphenol) and carvacrol are oxygenated aromatic compounds from the monoterpene group. They are the main constituents of thyme essential oil and show antiproliferative, antioxidant, and antiseptic properties. The aim of this study is to compare the antiproliferative and apoptotic effects of thymol and carvacrol on SKOV-3 ovarian cancer cell line. The cancer cells were treated with different concentrations of thymol and carvacrol (100, 200, 400, 600 µM) at 24 h and 48 h durations. The cell viability was investigated by MTT assay and analysis of apoptosis with annexin V assay was determined. The study show that thymol and carvacrol significantly induced apoptosis in all groups as dose and time-dependent (p < .05). The data in the present study demonstrated that thymol and carvacrol have apoptotic and antiproliferative properties in a concentration-dependent manner toward ovarian cancer cells. SKOV-3 cancer cell line was much more sensitive to the toxic effect of thymol than carvacrol.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cymenes/pharmacology , Oils, Volatile/pharmacology , Ovarian Neoplasms , Thymol/pharmacology , Anticarcinogenic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Oils, Volatile/chemistry , Ovarian Neoplasms/ultrastructure
4.
Cell Death Dis ; 10(8): 609, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31406198

ABSTRACT

Paeonol (Pae), a phenolic acid compound isolated from the Moutan Cortex, was previously demonstrated to exert multiple anticancer effects. The rational control of autophagy has been considered a potential treatment strategy for epithelial ovarian cancer. However, whether Pae induces autophagy and the relationship between its antitumour activities and autophagy in epithelial ovarian cancer are still unclear. In this study, we found that Pae induced not only antiproliferation activity and apoptosis but also autophagy, and complete autophagic flux was observed in A2780 and SKOV3 cells. In addition, combination treatment with Pae and an autophagy inhibitor (3-methyladenine and hydroxychloroquine) showed significant synergetic effects on inhibiting cell viability and promoting apoptosis in vitro and in the A2780 xenograft model, without severe side effects, which was often had by cisplatin. These results indicate that autophagy induced by Pae has a cytoprotective role in both A2780 and SKOV3 cells. Mechanistically, we found that Pae inhibited the protein kinase B(Akt)/mammalian target of rapamycin (mTOR) pathway. Furthermore, when combined with the inhibitors MK2206 and rapamycin to inhibit Akt and mTOR kinase activity, Pae-induced autophagy was increased. Taken together, our results demonstrate that Pae induced cytoprotective autophagy by inhibiting the Akt/mTOR pathway in ovarian cancer cells. Thus, the strategy of combining Pae with an autophagy inhibitor to block Akt/mTOR-dependent autophagy could enhance the antitumour activity of Pae and warrants further application for the treatment of ovarian cancer.


Subject(s)
Acetophenones/pharmacology , Autophagy/drug effects , Cytoprotection , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Acetophenones/chemistry , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytoprotection/drug effects , Female , Humans , Hydroxychloroquine/pharmacology , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Ovarian Neoplasms/ultrastructure , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
5.
Angiogenesis ; 22(3): 441-455, 2019 08.
Article in English | MEDLINE | ID: mdl-31161471

ABSTRACT

The origin of blood and lymphatic vessels in high-grade serous adenocarcinoma of ovary (HGSOC) is uncertain. We evaluated the potential of cancer stem cells (CSCs) in HGSOC to contribute to their formation. Using spheroids as an in vitro model for CSCs, we have evaluated their role in primary malignant cells (PMCs) in ascites from previously untreated patients with HGSOC and cell lines. Spheroids from PMCs grown under specific conditions showed significantly higher expression of endothelial, pericyte and lymphatic endothelial markers. These endothelial and lymphatic cells formed tube-like structures, showed uptake of Dil-ac-LDL and expressed endothelial nitric oxide synthase confirming their endothelial phenotype. Electron microscopy demonstrated classical Weibel-Palade bodies in differentiated cells. Genetically, CSCs and the differentiated cells had a similar identity. Lineage tracking using green fluorescent protein transfected cancer cells in nude mice confirmed that spheroids grown in stem cell conditions can give rise to all three cells. Bevacizumab, a monoclonal antibody that targets vascular endothelial growth factor inhibited the differentiation of spheroids to endothelial cells in vitro. These results suggest that CSCs contribute to angiogenesis and lymphangiogenesis in serous adenocarcinoma of the ovary, which can be inhibited.


Subject(s)
Adenocarcinoma/pathology , Lymphangiogenesis , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/pathology , Adenocarcinoma/blood supply , Adenocarcinoma/ultrastructure , Ascites/metabolism , Ascites/pathology , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Biomarkers, Tumor/metabolism , Blood Vessels/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Endothelial Cells/metabolism , Female , Humans , Neoplasm Proteins/metabolism , Neoplasms, Cystic, Mucinous, and Serous/blood supply , Neoplasms, Cystic, Mucinous, and Serous/ultrastructure , Neoplastic Stem Cells/ultrastructure , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/ultrastructure , Pericytes/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor A/metabolism
6.
Mol Biol Rep ; 46(5): 4685-4697, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31025149

ABSTRACT

The current authors previously reported that a carbonyl reductase 1 (CR1) DNA-dendrimer complex could potentially be used in gene therapy for peritoneal metastasis of ovarian cancer. The aims of the current study were to observe the cellular dynamics of peritoneal metastasis of epithelial ovarian cancer cells and to ascertain changes in the dynamics of ovarian cancer cells as a result of transfection of CR1 DNA. (1) Artificial human peritoneal tissue (AHPT) was seeded with serous ovarian cancer cells, and the process leading to development of peritoneal carcinomatosis was observed over time. (2) Peritoneal carcinomatosis was produced in mice and compared to a model using AHPT to determine the appropriateness of AHPT. (3) CR1 DNA was transfected into cancer cells seeded on AHPT, and the dynamics of cancer cells were observed over time. (1) Cancer cells perforated the mesothelium, leaving normal mesothelium intact. However, the cells proliferated between the layers of the mesothelium, forming a mass. After 24 h, cancer cells had invaded the lymphatics, and after 48-72 h cancer cells had invaded deep into the mesothelium, where they formed a mass. (2) Invasion of the peritoneum by cancer cells in a murine model of peritoneal carcinomatosis resembled that in a model using AHPT, and results substantiated the reproducibility of peritoneal carcinomatosis in AHPT. (3) Proliferation of cells transfected with CR1 DNA was significantly inhibited on AHPT, and necrosis was evident. Nevertheless, cancer cell invasion deep into the mesothelium was not inhibited. Use of a new tool, AHPT, in an in vitro model of peritoneal metastasis revealed that CR1 DNA inhibited cancer cell proliferation. CR1 DNA does not play a role in inhibiting invasion of the mesothelium during peritoneal metastasis, but it does affect cancer cell proliferation. Results suggested that CR1 DNA inhibits cancer cell proliferation via necrosis.


Subject(s)
Alcohol Oxidoreductases/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Alcohol Oxidoreductases/genetics , Animals , Cell Culture Techniques , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression , Genes, Reporter , Heterografts , Humans , Immunohistochemistry , Mice , Ovarian Neoplasms/genetics , Ovarian Neoplasms/ultrastructure , Peritoneal Neoplasms/ultrastructure
7.
Dis Model Mech ; 11(9)2018 09 25.
Article in English | MEDLINE | ID: mdl-30254133

ABSTRACT

Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy. EOC dissemination is predominantly via direct extension of cells and multicellular aggregates (MCAs) into the peritoneal cavity, which adhere to and induce retraction of peritoneal mesothelium and proliferate in the submesothelial matrix to generate metastatic lesions. Metastasis is facilitated by the accumulation of malignant ascites (500 ml to >2 l), resulting in physical discomfort and abdominal distension, and leading to poor prognosis. Although intraperitoneal fluid pressure is normally subatmospheric, an average intraperitoneal pressure of 30 cmH2O (22.1 mmHg) has been reported in women with EOC. In this study, to enable experimental evaluation of the impact of high intraperitoneal pressure on EOC progression, two new in vitro model systems were developed. Initial experiments evaluated EOC MCAs in pressure vessels connected to an Instron to apply short-term compressive force. A Flexcell Compression Plus system was then used to enable longer-term compression of MCAs in custom-designed hydrogel carriers. Results show changes in the expression of genes related to epithelial-mesenchymal transition as well as altered dispersal of compressed MCAs on collagen gels. These new model systems have utility for future analyses of compression-induced mechanotransduction and the resulting impact on cellular responses related to intraperitoneal metastatic dissemination.This article has an associated First Person interview with the first authors of the paper.


Subject(s)
Ascites/pathology , Models, Biological , Ovarian Neoplasms/pathology , Cadherins/metabolism , Cell Aggregation , Cell Line, Tumor , Cell Proliferation , Collagen/chemistry , Epithelial-Mesenchymal Transition/genetics , Female , Gels/chemistry , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/ultrastructure
9.
Sci Rep ; 8(1): 7228, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29740072

ABSTRACT

There is growing appreciation of the importance of the mechanical properties of the tumor microenvironment on disease progression. However, the role of extracellular matrix (ECM) stiffness and cellular mechanotransduction in epithelial ovarian cancer (EOC) is largely unknown. Here, we investigated the effect of substrate rigidity on various aspects of SKOV3 human EOC cell morphology and migration. Young's modulus values of normal mouse peritoneum, a principal target tissue for EOC metastasis, were determined by atomic force microscopy (AFM) and hydrogels were fabricated to mimic these values. We find that cell spreading, focal adhesion formation, myosin light chain phosphorylation, and cellular traction forces all increase on stiffer matrices. Substrate rigidity also positively regulates random cell migration and, importantly, directional increases in matrix tension promote SKOV3 cell durotaxis. Matrix rigidity also promotes nuclear translocation of YAP1, an oncogenic transcription factor associated with aggressive metastatic EOC. Furthermore, disaggregation of multicellular EOC spheroids, a behavior associated with dissemination and metastasis, is enhanced by matrix stiffness through a mechanotransduction pathway involving ROCK, actomyosin contractility, and FAK. Finally, this pattern of mechanosensitivity is maintained in highly metastatic SKOV3ip.1 cells. These results establish that the mechanical properties of the tumor microenvironment may play a role in EOC metastasis.


Subject(s)
Carcinoma, Ovarian Epithelial/ultrastructure , Gene Expression Regulation, Neoplastic , Mechanotransduction, Cellular/genetics , Ovarian Neoplasms/ultrastructure , Spheroids, Cellular/ultrastructure , Actomyosin/genetics , Actomyosin/metabolism , Animals , Biomechanical Phenomena , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Cell Cycle Proteins , Cell Line, Tumor , Cell Movement , Elastic Modulus , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Female , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Focal Adhesions/metabolism , Focal Adhesions/pathology , Focal Adhesions/ultrastructure , Humans , Mice , Mice, Inbred C57BL , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peritoneum/metabolism , Peritoneum/ultrastructure , Phosphorylation , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Microenvironment/genetics , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
10.
Nat Commun ; 9(1): 476, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29396402

ABSTRACT

Though used widely in cancer therapy, paclitaxel only elicits a response in a fraction of patients. A strong determinant of paclitaxel tumor response is the state of microtubule dynamic instability. However, whether the manipulation of this physiological process can be controlled to enhance paclitaxel response has not been tested. Here, we show a previously unrecognized role of the microtubule-associated protein CRMP2 in inducing microtubule bundling through its carboxy terminus. This activity is significantly decreased when the FER tyrosine kinase phosphorylates CRMP2 at Y479 and Y499. The crystal structures of wild-type CRMP2 and CRMP2-Y479E reveal how mimicking phosphorylation prevents tetramerization of CRMP2. Depletion of FER or reducing its catalytic activity using sub-therapeutic doses of inhibitors increases paclitaxel-induced microtubule stability and cytotoxicity in ovarian cancer cells and in vivo. This work provides a rationale for inhibiting FER-mediated CRMP2 phosphorylation to enhance paclitaxel on-target activity for cancer therapy.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Microtubules/metabolism , Nerve Tissue Proteins/metabolism , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacology , Protein-Tyrosine Kinases/genetics , RNAi Therapeutics , Tubulin Modulators/pharmacology , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Microscopy, Confocal , Microscopy, Fluorescence , Microtubules/drug effects , Microtubules/ultrastructure , Molecular Dynamics Simulation , Molecular Targeted Therapy , Neoplasm Transplantation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/ultrastructure , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Multimerization/drug effects , Protein Multimerization/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Small Interfering
11.
Int J Gynecol Cancer ; 28(3): 545-552, 2018 03.
Article in English | MEDLINE | ID: mdl-29040188

ABSTRACT

OBJECTIVE: Ovarian cancer is a common gynecological cancer, and parity is negatively associated with the incidence of this disease. This negative association is hypothesized to be due in part to shifting the balance of estrogen and progesterone toward more progesterone and reduced ovulation during pregnancy. However, studies suggested that parity is also associated with estrogen-independent gynecological cancers suggesting balance of hormones may not be the only protective factor. Extracellular vesicles (EVs) play an important role in cell-to-cell communication in physiological and pathological conditions. During pregnancy, large amounts of EVs are extruded from the placenta, and they seem to be involved in the remarkable adaptation of a woman's body to normal pregnancy. We hypothesized that EVs extruded from the placenta play a role in this protective effect. METHODS: Placental EVs were collected from first-trimester placentae, and cancer cell EVs were isolated from ovarian cancer cells. The EVs were exposed to ovarian cancer cells for 48 hours. The proliferation of cancer cells and the cell cycle were measured. In addition, phagocytosis of deported placental EVs by cancer cells was also measured. RESULTS: The proliferation of cancer cells was significantly reduced by treatment with placental EVs (P = 0.001, analysis of variance), but not EVs from monocytes (P = 0.195), compared with untreated cancer cells. Furthermore, placental EVs also prevented the proliferation of cancer cells induced by cancer cell-derived EVs (P = 0.001). This inhibition of proliferation of ovarian cancer cells was partially due to phagocytosis of placental EVs by cancer cells. Phagocytosis of placental EVs delayed progression through the cell cycle. Calreticulin, a phagocytic "eat me" signal carried by placental EVs significantly inhibited ovarian cancer growth (P = 0.001). CONCLUSIONS: Our data demonstrated that EVs extruded from the placenta prevented ovarian cancer cell growth by a mechanism that involved delaying progression of the cell cycle after phagocytosis of the EVs.


Subject(s)
Extracellular Vesicles/transplantation , Ovarian Neoplasms/therapy , Placenta/ultrastructure , Calreticulin/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Extracellular Vesicles/pathology , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/ultrastructure , Phagocytosis , Placenta/transplantation , Pregnancy , Recombinant Proteins/pharmacology , THP-1 Cells
12.
Int J Biochem Cell Biol ; 95: 9-16, 2018 02.
Article in English | MEDLINE | ID: mdl-29247711

ABSTRACT

The high mortality rate of ovarian cancer is connected with the development of acquired resistance to multiple cancer drugs, especially cisplatin. Activation of cytoprotective autophagy has been implicated as a contributing mechanism for acquired cisplatin resistance in ovarian cancer cells. Hexokinase 2 (HK2) phosphorylates glucose to generate glucose-6-phosphate, the rate-limiting step in glycolysis. Higher HK2 expression has been associated with chemoresistance in ovarian cancer. However, whether HK2 functionally contributes to cisplatin resistance in ovarian cancer is unclear. In this study, we investigated the role of HK2 in regulating ovarian cancer cisplatin resistance. Increased HK2 levels were detected in drug-resistant human ovarian cancer cells and tissues. Cisplatin downregulated HK2 in cisplatin-sensitive but not in resistant ovarian cancer cells. HK2 knockdown sensitized resistant ovarian cancer cells to cisplatin-induced cell death and apoptosis. Conversely, HK2 overexpression in cisplatin-sensitive cells induced cisplatin resistance. Mechanistically, cisplatin increased ERK1/2 phosphorylation as well as autophagic activity. Blocking autophagy with the autophagy inhibitor 3-MA sensitized resistant ovarian cancer cells to cisplatin. HK2 overexpression enhanced cisplatin-induced ERK1/2 phosphorylation and autophagy while HK2 knockdown showed the opposite effects. Blocking the MEK/ERK pathway using the MEK inhibitor U0126 prevented cisplatin-induced autophagy enhanced by HK2 overexpression. Furthermore, HK2 knockdown sensitized resistance ovarian tumor xenografts to cisplatin in vivo. In conclusion, our data supported that HK2 promotes cisplatin resistance in ovarian cancer by enhancing drug-induced, ERK-mediated autophagy. Therefore, targeting HK2 may be a new therapeutic strategy to combat chemoresistance in ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Hexokinase/metabolism , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cisplatin/therapeutic use , Female , Hexokinase/antagonists & inhibitors , Hexokinase/genetics , Humans , MAP Kinase Signaling System/drug effects , Mice, Inbred BALB C , Mice, Nude , Microscopy, Electron, Transmission , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/ultrastructure , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Ovary/ultrastructure , Protein Kinase Inhibitors/pharmacology , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Survival Analysis , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Biomed Pharmacother ; 95: 894-903, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28903185

ABSTRACT

Doxorubicin (DOX) is used as a "first-line" antineoplastic drug in ovarian and metastatic breast cancer. However, serious side effects, such as cardiotoxicity have been reported after DOX intravenous administration. Hence, we investigated different micelle-former biomaterials, as Soluplus®, Pluronic F127, Tetronic T1107 and d-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) to develop a potential mixed micellar nanocarrier for DOX delivery. Since DOX hydrochloride is a poor candidate to be encapsulated inside the hydrophobic core of the mixed micelles, we assayed a hydrophobic complex between DOX and sodium deoxycholate (NaDC) as an excellent candidate to be encapsulated within polymeric micelles. The combination of T1107:TPGS (1:3, weight ratio) demonstrated the best physicochemical properties together with a high DL capacity (6.43% w/v). Particularly, DOX in vitro release was higher at acidic tumour microenvironment pH value (5.5) than at physiological counterpart (7.4). The hydrodynamic diameter of the DOX/NaDC-loaded mixed micellar system was 10.7nm (PDI=0.239). The in vitro cytotoxicity of the mixed micellar formulation resulted significantly (p<0.05) higher than Doxil® against ovarian (SKOV-3) and triple-negative breast cancer cells (MDA-MB- 231). Further, the in vitro cellular uptake assays demonstrated a significant increment (p<0.05) of the DOX intracellular content for the mixed micelles versus Doxil® for both, SKOV-3 (at 2, 4 and 6h of incubation) and MDA-MB-231 (at 4h of incubation) cells. These findings suggest that T1107:TPGS (1:3) mixed micelles could be employed as a potential nanotechnological platform for drug delivery of DOX.


Subject(s)
Breast Neoplasms/pathology , Doxorubicin/analogs & derivatives , Doxorubicin/therapeutic use , Micelles , Ovarian Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/ultrastructure , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/pharmacology , Drug Liberation , Endocytosis/drug effects , Female , Humans , Inhibitory Concentration 50 , Ovarian Neoplasms/ultrastructure , Particle Size , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Static Electricity
14.
Br J Cancer ; 117(5): 752-755, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28742796

ABSTRACT

BACKGROUND: Leukocyte telomere length (LTL) is a potential biomarker of cancer prognosis; however, evidence for renal cell carcinoma (RCC) is inconsistent. METHODS: We investigated LTL and RCC-specific survival among 684 cases from the US kidney cancer study (USKC) and 241 cases from the prostate, lung, colorectal, and ovarian cancer screening trial (PLCO). Leukocyte telomere length was measured by quantitative polymerase chain reaction, and hazard ratios (HRs) and 95% confidence intervals (CIs) computed using multivariable Cox models. RESULTS: Short LTL was associated with poorer disease-specific survival in both USKC (lowest vs highest quartile: HR: 2.3, 95% CI: 1.2-4.4; P for trend=0.02) and PLCO (HR: 2.4, 95% CI: 1.0-5.4; P=0.04). Among USKC cases, the association was strongest for stage-I RCC (HR: 5.5, 95% CI: 1.6-19.0; P=0.006). CONCLUSIONS: Our findings suggest that shorter LTL is an independent marker of poor RCC prognosis, particularly for stage-I disease.


Subject(s)
Carcinoma, Renal Cell/ultrastructure , Colorectal Neoplasms/ultrastructure , Kidney Neoplasms/ultrastructure , Leukocytes/ultrastructure , Lung Neoplasms/ultrastructure , Ovarian Neoplasms/ultrastructure , Prostatic Neoplasms/ultrastructure , Telomere Shortening , Telomere/ultrastructure , Aged , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/mortality , Case-Control Studies , Colorectal Neoplasms/blood , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Female , Humans , Kidney Neoplasms/blood , Kidney Neoplasms/genetics , Kidney Neoplasms/mortality , Lung Neoplasms/blood , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Male , Middle Aged , Ovarian Neoplasms/blood , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , Prognosis , Prostatic Neoplasms/blood , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Survival Rate
15.
J Proteome Res ; 16(8): 3083-3091, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28675934

ABSTRACT

The extracellular matrix (ECM) is a complex meshwork of insoluble fibrillar proteins and signaling factors interacting together to provide architectural and instructional cues to the surrounding cells. Alterations in ECM organization or composition and excessive ECM deposition have been observed in diseases such as fibrosis, cardiovascular diseases, and cancer. We provide here optimized protocols to solubilize ECM proteins from normal or tumor tissues, digest the proteins into peptides, analyze ECM peptides by mass spectrometry, and interpret the mass spectrometric data. In addition, we present here two novel R-script-based web tools allowing rapid annotation and relative quantification of ECM proteins, peptides, and intensity/abundance in mass spectrometric data output files. We illustrate this protocol with ECMs obtained from two pairs of tissues, which differ in ECM content and cellularity: triple-negative breast cancer and adjacent mammary tissue, and omental metastasis from high-grade serous ovarian cancer and normal omentum. The complete proteomics data set generated in this study has been deposited to the public repository ProteomeXchange with the data set identifier: PXD005554.


Subject(s)
Extracellular Matrix/chemistry , Ovarian Neoplasms/chemistry , Proteomics/methods , Triple Negative Breast Neoplasms/chemistry , Breast/cytology , Extracellular Matrix/pathology , Extracellular Matrix Proteins/analysis , Female , Humans , Mass Spectrometry , Molecular Sequence Annotation , Omentum/cytology , Ovarian Neoplasms/secondary , Ovarian Neoplasms/ultrastructure , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/ultrastructure
16.
J Nippon Med Sch ; 84(3): 139-143, 2017.
Article in English | MEDLINE | ID: mdl-28724848

ABSTRACT

Adenomatoid tumors (ATs) are rare, benign neoplasms occurring mainly in reproductive organs such as the uterus, ovaries, fallopian tubes, and testes. Uterine adenomatoid tumors (UATs) are generally incidentally diagnosed during histopathological examination of excisional biopsies performed for other indications, most commonly uterine leiomyomas. We herein present a 38-year-old woman who underwent laparoscopic excision of a uterine leiomyoma and a right ovarian teratoma. Microscopic examination of the excisional biopsy revealed that the enucleated uterine tumor was composed of proliferating glandular tissue covered with single-layered cells that were surrounded by proliferating smooth muscle cells, corresponding exactly to the features of UATs. The excised ovarian cyst was confirmed to be a typical mature cystic teratoma. According to these histopathological findings, the patient was finally diagnosed with a UAT and coexisting teratoma. No recurrence was detected up to 6 months after excision. To the best of our knowledge, this is the eighth case report on laparoscopically enucleated UATs. Although recurrence risk may be low in UATs, further case reports are necessary to elucidate the safety and validity of laparoscopic excision for UATs.


Subject(s)
Adenomatoid Tumor/complications , Adenomatoid Tumor/surgery , Laparoscopy/methods , Ovarian Neoplasms/complications , Ovarian Neoplasms/surgery , Teratoma/complications , Teratoma/surgery , Uterine Neoplasms/complications , Uterine Neoplasms/surgery , Adenomatoid Tumor/diagnosis , Adenomatoid Tumor/ultrastructure , Adult , Female , Humans , Microscopy , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/ultrastructure , Teratoma/diagnosis , Teratoma/ultrastructure , Treatment Outcome , Uterine Neoplasms/diagnosis , Uterine Neoplasms/ultrastructure
17.
Neoplasia ; 19(7): 549-563, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28601643

ABSTRACT

Epithelial ovarian carcinoma spreads via shedding of cells and multicellular aggregates (MCAs) from the primary tumor into peritoneal cavity, with subsequent intraperitoneal tumor cell:mesothelial cell adhesion as a key early event in metastatic seeding. Evaluation of human tumor extracts and tissues confirms that well-differentiated ovarian tumors express abundant E-cadherin (Ecad), whereas advanced lesions exhibit upregulated N-cadherin (Ncad). Two expression patterns are observed: "mixed cadherin," in which distinct cells within the same tumor express either E- or Ncad, and "hybrid cadherin," wherein single tumor cell(s) simultaneously expresses both cadherins. We demonstrate striking cadherin-dependent differences in cell-cell interactions, MCA formation, and aggregate ultrastructure. Mesenchymal-type Ncad+ cells formed stable, highly cohesive solid spheroids, whereas Ecad+ epithelial-type cells generated loosely adhesive cell clusters covered by uniform microvilli. Generation of "mixed cadherin" MCAs using fluorescently tagged cell populations revealed preferential sorting into cadherin-dependent clusters, whereas mixing of cell lines with common cadherin profiles generated homogeneous aggregates. Recapitulation of the "hybrid cadherin" Ecad+/Ncad+ phenotype, via insertion of the CDH2 gene into Ecad+ cells, resulted in the ability to form heterogeneous clusters with Ncad+ cells, significantly enhanced adhesion to organotypic mesomimetic cultures and peritoneal explants, and increased both migration and matrix invasion. Alternatively, insertion of CDH1 gene into Ncad+ cells greatly reduced cell-to-collagen, cell-to-mesothelium, and cell-to-peritoneum adhesion. Acquisition of the hybrid cadherin phenotype resulted in altered MCA surface morphology with increased surface projections and increased cell proliferation. Overall, these findings support the hypothesis that MCA cadherin composition impacts intraperitoneal cell and MCA dynamics and thereby affects ultimate metastatic success.


Subject(s)
Cadherins/genetics , Gene Expression , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Cadherins/metabolism , Carcinoma, Ovarian Epithelial , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Neoplasm Staging , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/ultrastructure , Phenotype , Tissue Array Analysis
18.
J Biol Chem ; 292(32): 13111-13121, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28655772

ABSTRACT

Membrane type 1 matrix metalloproteinase (MT1-MMP, MMP-14) is a transmembrane collagenase highly expressed in metastatic ovarian cancer and correlates with poor survival. Accumulating evidence shows that the cytoplasmic tail of MT1-MMP is subjected to phosphorylation, and this post-translational modification regulates enzymatic activity at the cell surface. To investigate the potential role of MT1-MMP cytoplasmic residue Thr567 phosphorylation in regulation of metastasis-associated behaviors, ovarian cancer cells that express low endogenous levels of MT1-MMP were engineered to express wild-type MT1-MMP, a phosphomimetic mutant (T567E), or a phosphodeficient mutant (T567A). Results show that Thr567 modulation influences behavior of both individual cells and multicellular aggregates (MCAs). The acquisition of either wild-type or mutant MT1-MMP expression results in altered cohesion of epithelial sheets and the formation of more compact MCAs relative to parental cells. Cells expressing MT1-MMP-T567E phosphomimetic mutants exhibit enhanced cell migration. Furthermore, MCAs formed from MT1-MMP-T567E-expressing cells adhere avidly to both intact ex vivo peritoneal explants and three-dimensional collagen gels. Interaction of these MCAs with peritoneal mesothelium disrupts mesothelial integrity, exposing the submesothelial collagen matrix on which MT1-MMP-T567E MCAs rapidly disperse. Together, these findings suggest that post-translational regulation of the Thr567 in the MT1-MMP cytoplasmic tail may function as a regulatory mechanism to impact ovarian cancer metastatic success.


Subject(s)
Matrix Metalloproteinase 14/metabolism , Ovarian Neoplasms/metabolism , Peritoneum/pathology , Protein Processing, Post-Translational , Amino Acid Substitution , Animals , Cell Adhesion , Cell Aggregation , Cell Line, Tumor , Cell Movement , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Matrix Metalloproteinase 14/chemistry , Matrix Metalloproteinase 14/genetics , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/ultrastructure , Peritoneum/ultrastructure , Phosphorylation , Point Mutation , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Threonine/chemistry , Tissue Culture Techniques
20.
Ultrastruct Pathol ; 41(1): 62-66, 2017.
Article in English | MEDLINE | ID: mdl-28029275

ABSTRACT

Ovarian mature cystic teratoma (OMCT) is an ovarian benign neoplasm with excellent prognosis presenting components of the three germinal layers. However, transformation into a malignant neoplasm is a rare event (so-called somatic transformation). In most of the cases, the malignant component expresses as epidermoid carcinoma, but occasionally central nervous system tumors occur. Some of the previously reported tumors are astrocytoma, glioblastoma, and ependymoma. Somatic transformation of OMCT into an oligodendroglioma is exceptional. We report a 19-year-old female with a left OMCT with an area of oligonedroglial cells proliferation characterized by immunohistochemical studies with positivity for GFAP and S100, with a low Ki67 index (5%). Additionally, electron microscopy revealed oligodendrocytes with parallel bundles of cytoplasmic intermediate filaments, confirming the oligodendroglial nature of the proliferation. The patient was treated only with left oophorectomy, and three and half years after surgery, there is no evidence of disease.


Subject(s)
Biomarkers, Tumor/analysis , Cell Proliferation , Immunohistochemistry , Microscopy, Electron , Neoplasms, Cystic, Mucinous, and Serous/diagnosis , Oligodendroglioma/diagnosis , Ovarian Neoplasms/diagnosis , Teratoma/diagnosis , Female , Humans , Neoplasms, Cystic, Mucinous, and Serous/chemistry , Neoplasms, Cystic, Mucinous, and Serous/surgery , Neoplasms, Cystic, Mucinous, and Serous/ultrastructure , Oligodendroglioma/chemistry , Oligodendroglioma/surgery , Oligodendroglioma/ultrastructure , Ovarian Neoplasms/chemistry , Ovarian Neoplasms/surgery , Ovarian Neoplasms/ultrastructure , Ovariectomy , Predictive Value of Tests , Salpingectomy , Teratoma/chemistry , Teratoma/surgery , Teratoma/ultrastructure , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...