Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(12)2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38928452

RESUMEN

Bone marrow mesenchymal stem cells (BMSCs) are key players in promoting ovarian cancer cell proliferation, orchestrated by the dynamic interplay between cytokines and their interactions with immune cells; however, the intricate crosstalk among BMSCs and cytokines has not yet been elucidated. Here, we aimed to investigate interactions between BMSCs and ovarian cancer cells. We established BMSCs with a characterized morphology, surface marker expression, and tri-lineage differentiation potential. Ovarian cancer cells (SKOV3) cultured with conditioned medium from BMSCs showed increased migration, invasion, and colony formation, indicating the role of the tumor microenvironment in influencing cancer cell behavior. BMSCs promoted SKOV3 tumorigenesis in nonobese diabetic/severe combined immunodeficiency mice, increasing tumor growth. The co-injection of BMSCs increased the phosphorylation of p38 MAPK and GSK-3ß in SKOV3 tumors. Co-culturing SKOV3 cells with BMSCs led to an increase in the expression of cytokines, especially MCP-1 and IL-6. These findings highlight the influence of BMSCs on ovarian cancer cell behavior and the potential involvement of specific cytokines in mediating these effects. Understanding these mechanisms will highlight potential therapeutic avenues that may halt ovarian cancer progression.


Asunto(s)
Proliferación Celular , Citocinas , Células Madre Mesenquimatosas , Neoplasias Ováricas , Células Madre Mesenquimatosas/metabolismo , Femenino , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Humanos , Animales , Citocinas/metabolismo , Ratones , Línea Celular Tumoral , Técnicas de Cocultivo , Microambiente Tumoral , Movimiento Celular , Medios de Cultivo Condicionados/farmacología , Células de la Médula Ósea/metabolismo , Ratones SCID , Ratones Endogámicos NOD , Diferenciación Celular
2.
J Biomed Sci ; 30(1): 35, 2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37259079

RESUMEN

BACKGROUND: Cancer-specific adoptive T cell therapy has achieved successful milestones in multiple clinical treatments. However, the commercial production of cancer-specific T cells is often hampered by laborious cell culture procedures, the concern of retrovirus-based gene transfection, or insufficient T cell purity. METHODS: In this study, we developed a non-genetic engineering technology for rapidly manufacturing a large amount of cancer-specific T cells by utilizing a unique anti-cancer/anti-CD3 bispecific antibody (BsAb) to directly culture human peripheral blood mononuclear cells (PBMCs). The anti-CD3 moiety of the BsAb bound to the T cell surface and stimulated the differentiation and proliferation of T cells in PBMCs. The anti-cancer moiety of the BsAb provided these BsAb-armed T cells with the cancer-targeting ability, which transformed the naïve T cells into cancer-specific BsAb-armed T cells. RESULTS: With this technology, a large amount of cancer-specific BsAb-armed T cells can be rapidly generated with a purity of over 90% in 7 days. These BsAb-armed T cells efficiently accumulated at the tumor site both in vitro and in vivo. Cytotoxins (perforin and granzyme) and cytokines (TNF-α and IFN-γ) were dramatically released from the BsAb-armed T cells after engaging cancer cells, resulting in a remarkable anti-cancer efficacy. Notably, the BsAb-armed T cells did not cause obvious cytokine release syndrome or tissue toxicity in SCID mice bearing human tumors. CONCLUSIONS: Collectively, the BsAb-armed T cell technology represents a simple, time-saving, and highly safe method to generate highly pure cancer-specific effector T cells, thereby providing an affordable T cell immunotherapy to patients.


Asunto(s)
Anticuerpos Biespecíficos , Antineoplásicos , Neoplasias , Ratones , Animales , Humanos , Linfocitos T , Leucocitos Mononucleares , Ratones SCID , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/uso terapéutico , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Antineoplásicos/metabolismo
3.
Int J Mol Sci ; 24(14)2023 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-37511582

RESUMEN

This study investigated the effects of progesterone receptors A (PRA) and B (PRB) on proliferation, migration, invasion, anchorage-independent growth (AIG), and apoptosis of FE25 cells, a precancer p53- and retinoblastoma-defective human fallopian tube epithelial cell line. We observed that the transfection of PRA (FE25-PRA) or PRB (FE25-PRB) into FE25 cells significantly increased the expression of PRA or PRB at both RNA and protein levels without affecting cell morphology. The FE25-PRA cells exhibited slower proliferation, whereas FE25-PRB showed faster cell proliferation than the control cells. In contrast, the FE25-PRA cells showed the highest migration and invasion abilities, whereas the FE25-PRB cells showed the lowest migration and invasion abilities. After treatment with progesterone, all cell types showed decreased AIG levels, increased apoptotic rates in Terminal deoxynucleotidyl transferase (TdT) dUTP nick end labeling assay (TUNEL) staining, and increased levels of apoptotic proteins ascertained based on cleaved caspase-3 levels. The half-maximal inhibitory concentration of carboplatin increased in FE25-PRB cells, but that of paclitaxel remained unchanged. Overall, this study suggests that PRA and PRB have distinct roles in regulating the behavior of FE25 cells, and targeting these receptors could be a potential therapeutic strategy for ovarian cancer treatment. If PRA or PRB overexpression is observed in high-grade serous carcinoma, progesterone could be considered as an adjuvant therapy for these specific cancer patients. However, further research is needed to confirm these findings and investigate the mechanisms underlying these effects.


Asunto(s)
Progesterona , Receptores de Progesterona , Femenino , Humanos , Línea Celular Tumoral , Células Epiteliales/metabolismo , Trompas Uterinas/metabolismo , Progesterona/farmacología , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína de Retinoblastoma
4.
J Biomed Sci ; 29(1): 5, 2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35062934

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is the sixth most common type of cancer and has a high mortality rate worldwide. Sorafenib is the only systemic treatment demonstrating a statistically significant but modest overall survival benefit. We previously have identified the aurora kinases (AURKs) and FMS-like tyrosine kinase 3 (FLT3) multikinase inhibitor DBPR114 exhibiting broad spectrum anti-tumor effects in both leukemia and solid tumors. The purpose of this study was to evaluate the therapeutic potential of DBPR114 in the treatment of advanced HCC. METHODS: Human HCC cell lines with histopathology/genetic background similar to human HCC tumors were used for in vitro and in vivo studies. Human umbilical vein endothelial cells (HUVEC) were used to evaluate the drug effect on endothelial tube formation. Western blotting, immunohistochemical staining, and mRNA sequencing were employed to investigate the mechanisms of drug action. Xenograft models of sorafenib-refractory and sorafenib-acquired resistant HCC were used to evaluate the tumor response to DBPR114. RESULTS: DBPR114 was active against HCC tumor cell proliferation independent of p53 alteration status and tumor grade in vitro. DBPR114-mediated growth inhibition in HCC cells was associated with apoptosis induction, cell cycle arrest, and polyploidy formation. Further analysis indicated that DBPR114 reduced the phosphorylation levels of AURKs and its substrate histone H3. Moreover, the levels of several active-state receptor tyrosine kinases were downregulated by DBPR114, verifying the mechanisms of DBPR114 action as a multikinase inhibitor in HCC cells. DBPR114 also exhibited anti-angiogenic effect, as demonstrated by inhibiting tumor formation in HUVEC cells. In vivo, DBPR114 induced statistically significant tumor growth inhibition compared with the vehicle control in multiple HCC tumor xenograft models. Histologic analysis revealed that the DBPR114 treatment reduced cell proliferation, and induced apoptotic cell death and multinucleated cell formation. Consistent with the histological findings, gene expression analysis revealed that DBPR114-modulated genes were mostly related to the G2/M checkpoint and mitotic spindle assembly. DBPR114 was efficacious against sorafenib-intrinsic and -acquired resistant HCC tumors. Notably, DBPR114 significantly delayed posttreatment tumor regrowth and prolonged survival compared with the regorafenib group. CONCLUSION: Our results indicated that targeting AURK signaling could be a new effective molecular-targeted agent in the treatment of patients with HCC.


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos/genética , Células Endoteliales , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Sorafenib/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa 3 Similar a fms/genética
5.
Int J Mol Sci ; 23(22)2022 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-36430409

RESUMEN

Discovering new drugs is an expensive and time-consuming process, including target identification, bioavailability, pharmacokinetic (PK) tests, pharmacodynamic (PD) tests, toxicity profiles, recommended dosage test, and observation of the side effects, etc. Repurposed drugs could bypass some steps, starting from phase II trials, and shorten the processes. Statins, also known as HMG-CoA inhibitors (HMGCR), are commonly used to manage and prevent various cardiovascular diseases and have been shown to improve the morbidity and mortality of patients. In addition to the inhibitory effects on the production of cholesterol, the beneficial effects of statins on the prognosis and risk of various cancers are also shown. Statins not only inhibited cell proliferation, metastasis, and chemoresistance but affected the tumor microenvironment (TME). Thus, statins have great potential to be repurposed in oncology. Hence, we review the meta-analysis, cohort, and case-control studies of statins in gynecological cancers, and elucidate how statins regulate cell proliferation, apoptosis, tumor growth, and metastasis. Although the results in gynecological cancers remain controversial and the effects of different statins in different histotypes of gynecological cancers and TME are needed to elucidate further, statins are excellent candidates and worthy of being repurposed drugs in treating gynecological cancers.


Asunto(s)
Enfermedades Cardiovasculares , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Neoplasias , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Neoplasias/tratamiento farmacológico , Enfermedades Cardiovasculares/tratamiento farmacológico , Microambiente Tumoral , Apoptosis
6.
Biotechnol Appl Biochem ; 68(3): 676-682, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32610363

RESUMEN

Gap junctional intercellular communication (GJIC) is the transfer of ions, metabolites, and second messengers between neighboring cells through intercellular junctions. Connexin 43 (Cx43) was found to be the type of gap junction protein responsible for human granulosa cells (GCs) and oocyte communication, which is required for folliculogenesis and oocyte maturation. Bisphenol A (BPA), an estrogenic-like endocrine-disrupting chemical, is one of the most widely produced chemicals around the world. There are reports that the chemical might cause endometrial tumorigenesis and several female reproductive disorders. This study demonstrated that cell culture medium, containing antioxidants (N-acetyl-l-cysteine and l-ascorbic acid-2-phosphate), was able to enhance the survival and self-renewal of GCs. In addition, we found that BPA at environmentally relevant concentration (10-7  M) reduced Cx43 expression and GJIC in GCs through estrogen receptor and mitogen-activated protein kinase pathways. The results of this study not only reveal the reproductive toxicity of BPA but also provide possible mechanisms by which BPA inhibited GJIC in GCs.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Comunicación Celular/efectos de los fármacos , Conexina 43/antagonistas & inhibidores , Regulación hacia Abajo , Uniones Comunicantes/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Fenoles/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Conexina 43/genética , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Femenino , Uniones Comunicantes/metabolismo , Células de la Granulosa/metabolismo , Humanos
7.
Phys Chem Chem Phys ; 17(35): 23140-6, 2015 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-26279125

RESUMEN

We have performed a comprehensive study on the spectral relaxation dynamics of silver nanoclusters (AgNCs) synthesized in poly(methacrylic acid) (PMAA). In different polymer conformations and solvents, the spectral relaxation dynamics of PMAA-AgNCs can be globally fitted by a bi-exponential decay, the short component is about 0.2-0.3 ns, whereas the long component is in the range 1-3 ns. The spectral relaxation is associated with the energy transfer dynamics and the excitation of multiple emissive AgNCs. In this study, we have demonstrated the feasibility of using AgNCs as a fluorescent probe for fluorescence anisotropy studies. Meanwhile, the molecular crowding effects of the PMAA-AgNCs were addressed using the Triton X-100 reverse micelles. The results indicate that the fluorescence quantum yield of the AgNCs will be significantly increased under crowded conditions, which is beneficial for their usage in intracellular imaging studies.


Asunto(s)
Nanopartículas del Metal/química , Ácidos Polimetacrílicos/química , Plata/química , Micelas , Octoxinol/química
8.
Biotechnol Appl Biochem ; 62(5): 591-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25385323

RESUMEN

Octamer-binding transcription factor 4 (Oct-4), an important gene regulating stem cell pluripotency, is well-known for its ability to reprogram somatic cells in vitro, either alone or in concert with other factors. The aim of this study was to assess the effect of ectopic expression of Oct human amniotic fluid stem cells. We developed a novel method for isolation of putative human amniotic fluid-derived multipotent stem cells. These cells showing mesenchymal stem cell phenotypes (human amniotic fluid-derived mesenchymal stem cells, hAFMSCs) were transfected with a plasmid carrying genes for Oct-4 and the green fluorescent protein (GFP). The stably transfected cells, hAFMSCs-Oct4/GFP, were selected by using G418 and found to express the GFP reporter gene under the control of Oct-4 promoter. We found that hAFMSCs developed by our method possess very high self-renewal ability (about 78 cumulative population doublings) and multilineage differentiation potency. Significantly, the hAFMSCs-Oct4/GFP cells showed enhanced expression of the three major pluripotency genes Oct-4, Nanog, and Sox-2, and increased colony-forming ability and growth rate compared with the parental hAFMSCs. We demonstrated that the ectopic expression of Oct-4 gene in hAFMSCs with high self-renewal ability could upregulate Nanog and Sox-2 gene expression and enhance cell growth rate and colony-forming efficiency. Therefore, the ectopic expression of Oct-4 could be a strategy to develop pluripotency in hAFMSCs for clinical applications.


Asunto(s)
Líquido Amniótico/citología , Expresión Génica Ectópica , Proteínas de Homeodominio/genética , Células Madre Mesenquimatosas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factores de Transcripción SOXB1/genética , Regulación hacia Arriba/genética , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Femenino , Humanos , Células Madre Mesenquimatosas/citología , Proteína Homeótica Nanog , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Embarazo
9.
Taiwan J Obstet Gynecol ; 63(2): 178-185, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38485312

RESUMEN

OBJECTIVE: Endometriosis is an estrogen-dependent chronic inflammatory disease in women of reproductive age. A review of the literature revealed that cytokines and inflammatory factors are associated with endometriosis-associated infertility. Interleukin 33 (IL-33) is a strong inducer of other pro-inflammatory cytokines. Vascular cell adhesion molecule-1 (VCAM-1) plays a central role in recruiting inflammatory cells, whose expression facilitates leukocyte adhesion and is rapidly induced by pro-inflammatory cytokines. Many studies have indicated that VCAM-1 expression is high in endometriosis; however, whether the expression of VCAM-1 is related to IL-33 is unclear. MATERIALS AND METHODS: Human ovarian endometriotic stromal cells (hOVEN-SCs) were treated with IL-33 to enable investigation of cell characterization, gene and protein expression, and signal pathways. Proliferation potential was measured using an MTT assay. Gene expression was analyzed using reverse transcription-polymerase chain reaction. Protein expression assay was performed using western blot analysis. RESULTS: This study investigated the effects of IL-33 on VCAM-1 and COX-2 expression in hOVEN-SCs. First, the results revealed that the IL-33/ST2/mitogen-activated protein kinase (MAPK) signaling pathway could increase the expression of VCAM-1 and COX-2 in hOVEN-SCs. Second, we discovered that COX-2 expression was essential for IL-33-induced VCAM-1 expression because the effects could be negated through NS398, a selective COX-2 inhibitor. Finally, treatment of IL-33-treated hOVEN-SCs with celecoxib significantly and dose-responsively decreased VCAM-1 expression. CONCLUSION: Taken together, these results indicate that IL-33 can upregulate VCAM-1 expression in hOVEN-SCs through the IL-33/ST2/MAPK/COX-2 signaling pathway and thereby contribute to endometriosis.


Asunto(s)
Endometriosis , Molécula 1 de Adhesión Celular Vascular , Humanos , Femenino , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo , Molécula 1 de Adhesión Celular Vascular/farmacología , Celecoxib/metabolismo , Celecoxib/farmacología , Interleucina-33/metabolismo , Ciclooxigenasa 2/metabolismo , Endometriosis/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Células del Estroma/metabolismo , Células Cultivadas
10.
JMIR Res Protoc ; 13: e55662, 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38466979

RESUMEN

BACKGROUND: In recent years, advancements in cancer treatment have enabled cancer cell inhibition, leading to improved patient outcomes. However, the side effects of chemotherapy, especially leukopenia, impact patients' ability to tolerate their treatments and affect their quality of life. Traditional Chinese medicine is thought to provide complementary cancer treatment to improve the quality of life and prolong survival time among patients with cancer. OBJECTIVE: This study aims to evaluate the effectiveness of Chinese herbal medicine (CHM) as a complementary treatment for neutropenia prevention and immunity modulation during chemotherapy in patients with breast cancer. METHODS: We will conduct a real-world pragmatic clinical trial to evaluate the effectiveness of CHM as a supplementary therapy to prevent neutropenia in patients with breast cancer undergoing chemotherapy. Patients will be classified into CHM or non-CHM groups based on whether they received CHM during chemotherapy. Using generalized estimating equations or repeated measures ANOVA, we will assess differences in white blood cell counts, absolute neutrophil counts, immune cells, and programmed cell death protein 1 (PD-1) expression levels between the 2 groups. RESULTS: This study was approved by the research ethics committee of Hualien Tzu Chi Hospital (IRB 110-168-A). The enrollment process began in September 2021 and will stop in December 2024. A total of 140 patients will be recruited. Data cleaning and analysis are expected to finish in the middle of 2025. CONCLUSIONS: Traditional Chinese medicine is the most commonly used complementary medicine, and it has been reported to significantly alleviate chemotherapy-related side effects. This study's findings may contribute to developing effective interventions targeting chemotherapy-related neutropenia among patients with breast cancer in clinical practice. TRIAL REGISTRATION: International Traditional Medicine Clinical Trial Registry ITMCTR2023000054; https://tinyurl.com/yc353hes. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID): DERR1-10.2196/55662.

11.
Tzu Chi Med J ; 35(1): 24-30, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36866340

RESUMEN

Mesenchymal stem cells (MSCs) were applied to the therapy for degenerative diseases, immune, and inflammation. In tumor microenvironments (TME), different sources of MSCs showed that tumor-promoting and -inhibiting effects were mediated by different signaling pathways. Cancer-associated MSCs (CaMSCs) could be recruited from bone marrow or local tissues and mainly showed tumor-promoting and immunosuppressive effects. The transformed CaMSCs preserve the characteristics of stem cells, but the properties of regulating TME are different. Hence, we specifically focus on CaMSCs and discuss the detailed mechanisms of regulating the development of cancer cells and immune cells. CaMSCs could be a potential therapeutic target in various types of cancer. However, the detailed mechanisms of CaMSCs in the TME are relatively less known and need further study.

12.
Cell Transplant ; 32: 9636897231195240, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37632354

RESUMEN

Exosomes are phospholipid bilayer vesicles that are released by all types of cells, containing proteins, lipids, and nucleic acids such as DNAs and RNAs. Exosomes can be transferred between cells and play a variety of physiological and pathological regulatory functions. Noncoding RNAs, including micro RNAs, long noncoding RNAs, and circular RNAs, are the most studied biomolecules from exosomes and more and more studies found that noncoding RNAs play an important role in the diagnosis, prognosis, and treatment of diseases, including various types of cancer. Gynecological malignancies such as ovarian, endometrial, and cervical cancer seriously threaten women's life. Therefore, this article reviews the roles and applications of exosomes in gynecological malignancies, including the promotion or inhibition of tumor progression and regulation of tumor microenvironments, and as potential therapeutic targets for treating gynecological cancers.


Asunto(s)
Exosomas , Neoplasias de los Genitales Femeninos , MicroARNs , Neoplasias , ARN Largo no Codificante , Humanos , Femenino , Exosomas/metabolismo , Neoplasias de los Genitales Femeninos/metabolismo , Neoplasias de los Genitales Femeninos/patología , Neoplasias/metabolismo , MicroARNs/metabolismo , ARN no Traducido/metabolismo , ARN Largo no Codificante/metabolismo , Microambiente Tumoral
13.
Cells ; 12(22)2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37998370

RESUMEN

Fallopian tube epithelial cells (FTECs) play a significant role in the development of high-grade serous ovarian cancer (HGSOC), but their utilization in in vitro experiments presents challenges. To address these limitations, induced pluripotent stem cells (iPSCs) have been employed as a potential solution, driven by the hypothesis that orthologous iPSCs may offer superior differentiation capabilities compared with their non-orthologous counterparts. Our objective was to generate iPSCs from FTECs, referred to as FTEC-iPSCs, and compare their differentiation potential with iPSCs derived from skin keratinocytes (NHEK). By introducing a four-factor Sendai virus transduction system, we successfully derived iPSCs from FTECs. To assess the differentiation capacity of iPSCs, we utilized embryoid body formation, revealing positive immunohistochemical staining for markers representing the three germ layers. In vivo tumorigenesis evaluation further validated the pluripotency of iPSCs, as evidenced by the formation of tumors in immunodeficient mice, with histological analysis confirming the presence of tissues from all three germ layers. Quantitative polymerase chain reaction (qPCR) analysis illuminated a sequential shift in gene expression, encompassing pluripotent, mesodermal, and intermediate mesoderm-related genes, during the iPSC differentiation process into FTECs. Notably, the introduction of WNT3A following intermediate mesoderm differentiation steered the cells toward a FTEC phenotype, supported by the expression of FTEC-related markers and the formation of tubule-like structures. In specific culture conditions, the expression of FTEC-related genes was comparable in FTECs derived from FTEC-iPSCs compared with those derived from NHEK-iPSCs. To conclude, our study successfully generated iPSCs from FTECs, demonstrating their capacity for FTEC differentiation. Furthermore, iPSCs originating from orthologous cell sources exhibited comparable differentiation capabilities. These findings hold promise for using iPSCs in modeling and investigating diseases associated with these specific cell types.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Femenino , Animales , Ratones , Células Madre Pluripotentes Inducidas/metabolismo , Trompas Uterinas/metabolismo , Epitelio , Piel , Diferenciación Celular
14.
Am J Cancer Res ; 13(8): 3324-3341, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37693155

RESUMEN

This study explored the role of leucine-rich repeat neuronal 4 (LRRN4) in ovarian carcinogenesis using the p53- and Rb-defective human fallopian tube epithelial cell line FE25. We evaluated the expression of LRRN4 in FE25 cells with and without LRRN4 knockdown by short hairpin RNA (shRNA) and studied its effects on cell proliferation, cell cycle, migration, invasion, chemotherapeutic sensitivity, apoptosis, and xenograft formation. The results showed that FE25 shRNA-LRRN4 cells exhibited more aggressive malignant behaviors than FE25 cells, including faster proliferation and increased cell distribution in the G2/M phase, Akt pathway activation, cell migration, and cell invasion, as well as decreased sensitivity to chemotherapeutic drugs. FE25 shRNA-LRRN4 cells exhibited reduced levels of apoptosis and decreased expression of cleaved caspase 3, 7, 8, and 9, indicating reduced apoptotic activity. Additionally, FE25 shRNA-LRRN4 cells showed decreased LRRN4 and CK7 expression and increased WT1 expression, suggesting a potential role for LRRN4 in ovarian carcinogenesis. FE25 shRNA-LRRN4 generated a xenograft in mice with increased levels of WT1 and TP53 expression compared to their levels in cells. Overall, this study suggests that LRRN4 may play a role in ovarian carcinogenesis by promoting aggressive malignant behavior in FE25 cells through the activation of the Akt pathway. These findings provide insights into the potential molecular mechanisms underlying ovarian cancer and may have implications for the development of new therapeutic targets for this disease.

15.
Taiwan J Obstet Gynecol ; 62(1): 16-21, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36720532

RESUMEN

OBJECTIVE: Research has suggested that tumor-initiating tumor stem cells are derived from normal stem cells and that tumor cells undergo progressive de-differentiation to achieve a stem cell-like state. Tumor stem cells are characterized by high proliferation ability, high plasticity, expression of multi-drug resistance proteins, and the ability to seed new tumors. Octamer-binding transcription factor 4 (Oct-4) and its activation targets are overexpressed in the tumor stem cells of various types of tumors, and this expression is associated with the pathogenesis, development, and poor prognosis of tumors. The primary objective of this study was to test if a stably transfected with Oct-4 gene cell line, RL95-2/Oct-4, has the characteristics of tumor stem cells. MATERIALS AND METHODS: Human endometrial carcinoma cells (RL95-2) were transfected with a plasmid carrying genes for Oct-4 and green fluorescent protein (GFP). The stably transfected cells, RL95-2/Oct-4, were selected using G418 and observed to express the GFP reporter gene under the control of the Oct-4 promoter. GFP expression levels of RL95-2/Oct-4 cells were measured using flow cytometry. The proliferation potential of cells was determined according to cumulative population doubling and colony-formation efficiency. Gene expression was analyzed using reverse transcription-polymerase chain reaction. RESULTS: RL95-2/Oct-4 cells not only exhibited increased expression of the three most important stem cell genes, Oct-4, Nanog, and Sox2, but also had increased expression of the endometrial tumor stem cell genes CD133 and ALDH1. Furthermore, enhanced expression of these genes in the RL95-2/Oct-4 cells was associated with higher colony-forming ability and growth rate than in parental RL95-2 cells. We also observed that cisplatin induced less cell death in RL95-2/Oct-4 cells than in RL95-2 cells, indicating that RL95-2/Oct-4 cells were more resistant to chemotherapeutic agents. CONCLUSION: The study findings contribute to investigate the effects of Oct-4 on tumor stem cell origins.


Asunto(s)
Cisplatino , Neoplasias Endometriales , Factor 3 de Transcripción de Unión a Octámeros , Femenino , Humanos , Línea Celular Tumoral , Proliferación Celular , Cisplatino/farmacología , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Resistencia a Antineoplásicos
16.
Stem Cells Transl Med ; 12(1): 39-53, 2023 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-36610716

RESUMEN

Current mesenchymal stem cell (MSC) research is based on xenotransplantation of human MSCs (hMSCs) in immunodeficient mice and cannot comprehensively predict MSC repair mechanisms and immunomodulatory effects in damaged tissue. This study compared the therapeutic efficacy, mechanisms, and immune response of hMSCs and mouse MSCs (mMSCs) in immunocompetent mice with CCl4-induced acute liver failure. mMSCs maintained F4/80+ hepatic macrophage recruitment into the damaged liver region, increased IL-6-dependent hepatocyte proliferation, and reduced inflammatory TNF-α cytokine secretion. Moreover, mMSCs reduced α-SMA+ myofibroblast activation by lowering TGF-ß1 accumulation in damaged liver tissue. In contrast, hMSCs lowered TNF-α and TGF-ß1 by reducing the recruitment of F4/80+ hepatic macrophages, which lost the ability to remove debris and induce IL-6 liver regeneration. Finally, hMSCs, but not mMSCs, caused a significant antibody response in immunocompetent mice; therefore, hMSCs are unsuitable for long-term MSC studies. This comparative study provides reference information for further MSC studies of immunocompetent mice.


Asunto(s)
Fallo Hepático Agudo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Animales , Humanos , Ratones , Inmunidad , Interleucina-6/farmacología , Fallo Hepático Agudo/terapia , Factor de Crecimiento Transformador beta1/farmacología , Factor de Necrosis Tumoral alfa/farmacología
17.
Biotechnol Appl Biochem ; 59(3): 262-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23586837

RESUMEN

A tumorigenic cell line with estrogen receptor and HER2 expression (ER/HER2⁺), R2N1d, was developed from a human breast epithelial cell type with stem cell characteristics in a growth factor/hormone-deprived cell culture condition. This study was undertaken to test whether tumor growth and other biological effects could be induced by estrogen in this cell line. The results clearly show that estrogen treatment greatly promoted the tumor growth of R2N1d cells in immune-deficient mice. Estrogen treatment of R2N1d cells in vitro was also found to induce other phenotypic changes related to breast carcinogenesis, that is, 1) the induction of epithelial-mesenchymal transition (EMT) shown by molecular and functional marker changes; 2) a significant increase of the CD44(high)/CD24(-/low) stem cell population; 3) the enhancement of cell growth rate and colony-forming ability; and 4) the acquisition of metastatic ability, that is, increased cell migration and invasiveness. From these results, we conclude that 1) estrogen could induce EMT and cancer stem cells and promote tumor growth in ER⁺/HER2⁺ cells known to be derived from human breast epithelial stem cells, and 2) normal stem cells could give rise to cancer stem cells.


Asunto(s)
Células Epiteliales/citología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Estradiol/farmacología , Receptor ErbB-2/genética , Receptores de Estrógenos/genética , Células Madre/citología , Animales , Neoplasias de la Mama/patología , Adhesión Celular , Línea Celular Tumoral , Femenino , Citometría de Flujo , Humanos , Ratones , Ratones Desnudos
18.
Cell Transplant ; 31: 9636897221104452, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35712817

RESUMEN

Endometrial cancer is the most common gynecologic cancer with high heterogeneity. However, there are limited treatment options for advanced endometrial carcinoma. In recent years, immunotherapy has broadly been used for the treatment of various cancers. However, the efficacy of immunotherapy against endometrial cancer is limited. The tumor microenvironment, including mesenchymal stem cells (MSCs), may contribute to tumor progression through cancer cells themselves and through cells of the immune system. We successfully isolated endometrial cancer-derived MSCs (EmCaMSCs) from patients and found that the population of MSCs in tumor tissues was approximately 1%-5%. The population of MSCs correlated with the stage of the disease. EmCaMSCs expressed MSC markers and exhibited trilineage differentiation ability. The programmed death ligands PD-L1 and PD-L2 were highly expressed in EmCaMSCs; their expression could be further enhanced by tumor necrosis factor-α and interferon-γ. When cocultured with peripheral blood mononuclear cells (PBMCs), anti-CD3, and anti-CD28, EmCaMSCs inhibited the proliferation of PBMCs, which were partially rescued by treatment with anti-PD-L1 antibodies. From the profile of conditioned medium of EmCaMSCs, we discovered that interleukin (IL)-8 and insulin-like growth factor-binding protein 6 could also rescue the proliferation of PBMCs. Furthermore, EmCaMSCs cocultured with IL-2-induced PBMCs exhibited decreased expression of CD56, CD4, and CD8 and showed decreased cytotoxicity toward K562 cells and endometrial cancer cells. Overall, EmCaMSCs were isolated successfully from endometrial cancer tissues and exhibited immunosuppressive effects and may be targeted for the treatment of endometrial cancer by anti-cytokine and immune checkpoint inhibitors. The percentage of MSCs in tumor stroma might predict the prognosis of endometrial cancer.


Asunto(s)
Neoplasias Endometriales , Células Madre Mesenquimatosas , Técnicas de Cocultivo , Neoplasias Endometriales/metabolismo , Femenino , Humanos , Tolerancia Inmunológica , Leucocitos Mononucleares , Células Madre Mesenquimatosas/metabolismo , Microambiente Tumoral
19.
Am J Obstet Gynecol ; 204(1): 21.e1-7, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20833385

RESUMEN

OBJECTIVE: Management of equivocal Papanicolaou smear result remains to be challenging even with the aid of human papillomavirus test. Recently, 3 novel methylation-silenced genes, PAX1, WT1, and PCDH10, have been found to be specifically associated with cervical cancer. We compared the performances of methylation test of these genes with human papillomavirus tests in triage of equivocal Papanicolaou smear result. STUDY DESIGN: Two hundred twenty-two women with Papanicolaou smear results of atypical cells of undetermined significance nested to a multicenter, nation-wide cohort (the T1899 cohort) were studied. Status of cervical neoplasm was diagnosed with colposcopic biopsy. Status of gene methylation was determined by methylation-specific polymerase chain reaction. High-risk human papillomavirus DNA was detected by polymerase chain reaction-reverse line blot hybridization and Hybrid Capture 2. RESULTS: Cervical intraepithelial neoplasm 1, cervical intraepithelial neoplasm 2, cervical intraepithelial neoplasm 3, carcinoma in situ, carcinoma, and normal cervix were diagnosed in 58, 17, 14, 10, 1, and 120 women, respectively. Methylation of PCDH10, WT1, and PAX1 was highly associated with the severity of cervical neoplasm (P < 10⁻9, < 10⁻7, and < 10⁻5, respectively). In comparison with a negative test result, the odds ratio (95% confidence intervals) for cervical intraepithelial neoplasm 3 or more severe neoplasms for women tested positive for methylation of these 3 genes were 26.4 (9.0-77.3), 18.1 (6.9-47.2), and 10.3 (4.1-25.9), respectively; whereas those positive for human papillomavirus polymerase chain reaction and Hybrid Capture 2 were 10.5 (3.5-31.9) and 5.6 (2.3-21.4). In triage for atypical cells of undetermined significance, each methylation test had less colposcopy referral and false-positive rates, but higher false-negative rate than the human papillomavirus tests. With a combination test of PCDH10 or WT1 methylation, a comparable false-negative rate (P = .62) but much less false-positive rate (P = .002) and colposcopy referral rate (P < 10⁻6) were achieved. CONCLUSION: In triage of atypical cells of undetermined significance Papanicolaou smear results, methylation test of WT1 and PCDH10 is superior to human papillomavirus test in this multicenter cohort. Comparing to current human papillomavirus triage, the new test has only one third of false positivity and half of colposcopy referral, with no compromise of the sensitivity in diagnosis of cervical intraepithelial neoplasm 3 or more severe neoplasms.


Asunto(s)
Cadherinas/análisis , Carcinoma in Situ/diagnóstico , Proteínas de Neoplasias/análisis , Factores de Transcripción Paired Box/análisis , Displasia del Cuello del Útero/diagnóstico , Neoplasias del Cuello Uterino/diagnóstico , Proteínas WT1/análisis , Cadherinas/genética , Carcinoma in Situ/patología , Carcinoma in Situ/virología , Cuello del Útero/patología , Estudios de Cohortes , Intervalos de Confianza , Metilación de ADN , ADN Viral/análisis , Femenino , Humanos , Proteínas de Neoplasias/genética , Oportunidad Relativa , Factores de Transcripción Paired Box/genética , Prueba de Papanicolaou , Papillomaviridae/genética , Infecciones por Papillomavirus/diagnóstico , Infecciones por Papillomavirus/patología , Reacción en Cadena de la Polimerasa/métodos , Protocadherinas , Taiwán , Displasia del Cuello del Útero/patología , Displasia del Cuello del Útero/virología , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/virología , Frotis Vaginal/métodos , Proteínas WT1/genética
20.
Biol Cell ; 102(3): 159-72, 2010 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-19895368

RESUMEN

BACKGROUND INFORMATION: The common phenotypes of cancer and stem cells suggest that cancers arise from stem cells. Oestrogen is one of the few most important determinants of breast cancer, as shown by several lines of convincing evidence. We have previously reported a human breast epithelial cell type (Type 1 HBEC) with stem cell characteristics and ER alpha (oestrogen receptor alpha) expression. A tumorigenic cell line, M13SV1R2, was developed from this cell type after SV40 (simian virus 40) large T-antigen transfection and X-ray irradiation. The cell line, however, was not responsive to oestrogen for cell growth or tumour development. In the present study, we tested the hypothesis that deprivation of growth factors and hormones may change the tumorigenicity and oestrogen response of this cell line. RESULTS: The M13SV1R2 cells lost their tumorigenicity after culturing in a growth factor/hormone-deprived medium for >10 passages (referred to as R2d cells) concomitant with the expression of two tumour suppressor genes, namely those coding for maspin and alpha 6 integrin. However, these cells acquired oestrogen responsiveness in cell growth and tumour development. By immunocytochemistry, Western blotting and flow cytometry analysis, oestrogen treatment of R2d cells was found to induce many important effects related to breast carcinogenesis, namely: (i) the emergence of a subpopulation of cells expressing CD44+/high/CD24-/low breast tumour stem cell markers; (ii) the induction of EMT (epithelial-to-mesenchymal transition); (iii) the acquisition of metastatic ability; and (iv) the expression of COX-2 (cyclo-oxygenase-2) through a CD44-mediated mechanism. CONCLUSION: An oestrogen-responsive cell line with ER alpha and CD44+/CD24-/low expression can be derived from breast epithelial stem cells. The tumorigenicity and oestrogen response of these cells could depend on the cell culture conditions. The findings of this study have implications in regard to the origins of (1) ER alpha-positive breast cancers, (2) CD44+/CD24-/low breast tumour stem cells and (3) the metastatic ability of breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/metabolismo , Receptor alfa de Estrógeno/metabolismo , Células Madre Neoplásicas/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Pruebas de Carcinogenicidad , Carcinoma/genética , Técnicas de Cultivo de Célula , Desdiferenciación Celular/efectos de los fármacos , Desdiferenciación Celular/fisiología , Línea Celular , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Medios de Cultivo/química , Medios de Cultivo/farmacología , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Receptor alfa de Estrógeno/genética , Estrógenos/deficiencia , Estrógenos/farmacología , Femenino , Genes Supresores de Tumor/efectos de los fármacos , Humanos , Receptores de Hialuranos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Metástasis de la Neoplasia/fisiopatología , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA