Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Exp Cell Res ; 422(1): 113416, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36375513

RESUMEN

In the previous study, we originally developed cancer stem cells (CSCs) models from mouse induced pluripotent stem cells (miPSCs) by culturing miPSCs in the conditioned medium of cancer cell lines, which mimiced as carcinoma microenvironment. However, the molecular mechanism of conversion in detail remains to be uncovered. Microarray analysis of the CSCs models in this study revealed Dsg2, one of the members of the desmosomal cadherin family, was up-regulated when compared with the original miPSCs. Moreover, the expression of key factors in Wnt/ß-catenin signaling pathway were also found up-regulated in one of the CSCs models, named miPS-LLCcm. An autocrine loop was implied between Dsg2 and Wnt/ß-catenin signaling pathway when miPSCs were treated with Wnt/ß-catenin signaling pathway activators, Wnt3a and CHIR99021, and when the CSCs model were treated with inhibitors, IWR-1 and IWP-2. Furthermore, the ability of proliferation and self-renewal in the CSCs model was markedly decreased in vitro and in vivo when Dsg2 gene was knocked down by shRNA. Our results showed that the Wnt/ß-catenin signaling pathway is activated by the up-regulation of Dsg2 expresssion during the conversion of miPSCs into CSCs implying a potential mechanism of the tranformation of stem cells into malignant phenotype.


Asunto(s)
Desmogleína 2 , Células Madre Pluripotentes Inducidas , Células Madre Neoplásicas , Vía de Señalización Wnt , Animales , Ratones , beta Catenina/genética , beta Catenina/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Células Madre Neoplásicas/metabolismo , Regulación hacia Arriba/genética , Vía de Señalización Wnt/genética , Desmogleína 2/genética , Desmogleína 2/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo
2.
J Cell Biochem ; 123(7): 1183-1196, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35578735

RESUMEN

Human Cripto-1 is a member of the epidermal growth factor (EGF)-Cripto-FRL-1-Cryptic (CFC) family family and performs critical roles in cancer and various pathological and developmental processes. Recently we demonstrated that a soluble form of Cripto-1 suppresses the self-renewal and enhances the differentiation of cancer stem cells (CSCs). A functional form of soluble Cripto-1 was found to be difficult to obtain because of the 12 cysteine residues in the protein which impairs the folding process. Here, we optimized the protocol for a T7 expression system, purification from inclusion bodies under denatured conditions refolding of a His-tagged Cripto-1 protein. A concentrations of 0.2-0.4 mM isopropyl ß-D-1-thiogalactopyranoside (IPTG) at 37°C was found to be the optimal concentration for Cripto-1 expression while imidazole at 0.5 M was the optimum concentration to elute the Cripto-1 protein from a Ni-column in the smallest volume. Cation exchange column chromatography of the Cripto-1 protein in the presence of 8 M urea exhibited sufficient elution profile at pH 5, which was more efficient at recovery. The recovery of the protein reached to more than 26.6% after refolding with arginine. The purified Cripto-1 exhibited high affinity to the anti-ALK-4 antibody and suppressed sphere forming ability of CSCs at high dose and induced cell differentiation.


Asunto(s)
Neoplasias , Células Madre Neoplásicas , Diferenciación Celular , Factor de Crecimiento Epidérmico/química , Factor de Crecimiento Epidérmico/farmacología , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo
3.
Br J Cancer ; 127(2): 193-201, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35292758

RESUMEN

The growth of cancer tissue is thought to be considered driven by a small subpopulation of cells, so-called cancer stem cells (CSCs). CSCs are located at the apex of a hierarchy in a cancer tissue with self-renewal, differentiation and tumorigenic potential that produce the progeny in the tissue. Although CSCs are generally believed to play a critical role in the growth, metastasis, and recurrence of cancers, the origin of CSCs remains to be reconsidered. We hypothesise that, chronic diseases, including obesity and diabetes, establish the cancer-inducing niche (CIN) that drives the undifferentiated/progenitor cells into CSCs, which then develop malignant tumours in vivo. In this context, a CIN could be traced to chronic inflammation that involves long-lasting tissue damage and repair after being exposed to factors such as cytokines and growth factors. This must be distinguished from the cancer microenvironment, which is responsible for cancer maintenance. The concept of a CIN is most important for cancer prevention as well as cancer therapy.


Asunto(s)
Neoplasias , Diferenciación Celular , Humanos , Inflamación/patología , Neoplasias/patología , Células Madre Neoplásicas/patología , Microambiente Tumoral
4.
Cell Biochem Funct ; 40(3): 310-320, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35285948

RESUMEN

Diphenyleneiodonium (DPI) has long been evaluated as an anticancer drug inhibiting NADPH oxidase, the IC50 in several cancer cell lines was reported 10 µM, which is too high for efficacy. In this study, we employed miPS-Huh7cmP cells, which we previously established as a cancer stem cell (CSC) model from induced pluripotent stem cells, to reevaluate the efficacy of DPI because CSCs are currently one of the main foci of therapeutic strategy to treat cancer, but generally considered resistant to chemotherapy. As a result, the conventional assay for the cell growth inhibition by DPI accounted for an IC50 at 712 nM that was not enough to define the effectiveness as an anticancer drug. Simultaneously, the wound-healing assay revealed an IC50 of approximately 500 nM. Comparatively, the IC50 values shown on sphere formation, colony formation, and tube formation assays were 5.52, 12, and 8.7 nM, respectively. However, these inhibitory effects were not observed by VAS2780, also a reputed NADPH oxidase inhibitor. It is noteworthy that these three assays are evaluating the characteristic of CSCs and are designed in the three-dimensional (3D) culture methods. We concluded that DPI could be a suitable candidate to target mitochondrial respiration in CSCs. We propose that the 3D culture assays are more efficient to screen anti-CSC drug candidates and better mimic tumor microenvironment when compared to the adherent monolayer of 2D culture system used for a conventional assay, such as cell growth inhibition and wound-healing assays.


Asunto(s)
Antineoplásicos , Células Madre Pluripotentes Inducidas , Neoplasias , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular , NADPH Oxidasas/metabolismo , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Compuestos Onio
5.
Adv Exp Med Biol ; 1393: 125-139, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36587305

RESUMEN

Many tumors are resistant to conventional cancer therapies because a tumor is composed of heterogeneous cell population. Especially, subpopulation of cancer stem cells, which have self-renewal and differentiation properties and responsible for the tumor initiation, is generally considered resistant to chemo-, radio-, and immune therapy. Understanding the mechanism of drug resistance in cancer stem cells should lead to establish more effective therapeutic strategies. Actually, different molecular mechanisms are conceivable for cancer stem cells acquiring drug resistance. These mechanisms include not only cytoplasmic signaling pathways but also the intercellular communications in the tumor microenvironment. Recently, a great deal of successful reports challenged to elucidate the mechanisms of drug resistance and to develop novel treatments targeting cancer stem cells.


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias , Humanos , Neoplasias/patología , Transformación Celular Neoplásica/patología , Diferenciación Celular , Células Madre Neoplásicas/patología , Microambiente Tumoral
6.
Adv Exp Med Biol ; 1393: 83-101, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36587303

RESUMEN

Cancer stem cells (CSCs) are small subpopulation sharing similar properties like normal stem such as self-renewal and differentiation potential to direct tumor growth. Last few years, scientists considered CSCs as the cause of phenotypic heterogeneity in diverse cancer types. Also, CSCs contribute to cancer metastasis and recurrence. The cellular and molecular regulators influence on the CSCs' phenotype changing their behaviors in different stages of cancer progression. CSC markers play significance roles in cancer diagnosis and characterization. We delineate the cross-talks between CSCs and the tumor microenvironment that supports their intrinsic properties including survival, stemness, quiescence and their cellular and molecular adaptation. An insight into the markers of CSCs specific to organs is described.


Asunto(s)
Neoplasias , Humanos , Neoplasias/genética , Neoplasias/patología , Células Madre Neoplásicas/patología , Diferenciación Celular , Fenotipo , Microambiente Tumoral/genética
7.
Biochem Biophys Res Commun ; 583: 49-55, 2021 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-34735879

RESUMEN

Cancer stem cells (CSCs) are responsible for cancer initiation, drug resistance, and aggressive tumor phenotypes. Our lab has established a novel method to induce CSCs from induced pluripotent stem (iPS) cells in a microenvironment mimicking chronic inflammation. The converted cells acquired CSC characteristics and developed malignant tumors. Recently, we demonstrated that nonmutagenic chemical inhibitors accelerated the conversion of mouse iPS (miPS) cells into CSCs. Here, we investigated the effects of AZD-6244, a MEK1/2-specific inhibitor, on the conversion of iPS cells into CSCs. The miPS cells were cultured for one week in the presence of the conditioned medium (CM) of Lewis lung carcinoma (LLC) cells and AZD-6244, PD0325901, a pan-MEK inhibitor, or GDC-0879, a B-Raf inhibitor. As a result, AZD-6244 enhanced the conversion of iPS cells into CSCs and upregulated AKT phosphorylation as same as GDC-0879 and PD0325901. The converted cells maintained their self-renewal ability and stemness gene expression. The expression of the CSC markers CD24, CD44 and CD133 was higher in the cells cultured with MAPK inhibitors than in those cultured without MAPK inhibitors. Moreover, converted cells gained migration and invasion abilities assessed by in vitro assays. Therefore, the inhibition of MEK1/2 was found to be critical for the conversion of normal stem cells into CSCs in the tumor-inducing microenvironment.

8.
Cell Biol Int ; 45(4): 749-756, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33274828

RESUMEN

Angiogenesis is generally involved in tumor growth and metastasis. Cancer stem cells (CSCs) are considered to facilitate the angiogenesis. Therefore, CSCs could be the effective targets to stop angiogenesis. Recently, our group successfully generated CSC models from induced pluripotent stem cells (iPSCs) in the presence of conditioned medium derived from cancer derived cells. These novel model CSCs has been characterized by highly tumorigenic, angiogenic and metastatic potentials in vivo. The angiogenic potential of CSCs has been explained by the expression of both angiogenic factors and their receptors implying the angiogenesis in autocrine manner. In this protocol we optimized the method to evaluate tumor angiogenesis with the CSC model, which was described effective to assess sorafenib as an antiangiogenic drug, on chick chorioallantoic membrane (CAM) assay. Our results demonstrate that CSCs developed from iPSCs and CAM assay are a robust and cost-effective tool to evaluate tumor angiogenesis with CSCs. Collectively, CSCs in CAM assay could serve as a very useful model for the screening of potential therapeutic agents targeting tumor angiogenesis.


Asunto(s)
Neoplasias , Neovascularización Patológica , Sorafenib/farmacología , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Embrión de Pollo , Membrana Corioalantoides/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Células Madre Neoplásicas
9.
Cell Biochem Funct ; 39(7): 896-907, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34268768

RESUMEN

Metformin exhibits anti-cancer activities in various types of tumours while it is prescribed as the first-line drug for type 2 diabetes. Since new evidence has recently suggested that metformin could target cancer stem cells (CSCs) and prevent their recurrence, repositioning of metformin could be considered as a candidate for anti-CSC agent. In this study, we assessed the effect of metformin on the cancer stem cells developed from induced pluripotent stem cells. As the result, metformin significantly suppressed the self-renewal ability of CSCs when assessed by 3-(4,5-dimethythiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and cell counting methods exhibiting the IC50 as approximately 20 mM, which suppressed tube formation by CSCs on Matrigel reducing the angiogenic potential of CSCs. Cell cycle analysis showed that metformin reduced the percentage of cells in the S phase increasing the percentage of cells in G0/G1 phase. Moreover, the tumorigenicity of CSCs was found to be attenuated when the cells were injected with metformin. From these results, we concluded that metformin could be promising for targeted therapy by repositioning the widely available drugs with safety. SIGNIFICANCE OF THE STUDY: Metformin could target CSCs and prevent their recurrence, repositioning of metformin could be considered as a candidate for the anti-CSC agent. In this paper, we assessed the effect of metformin on the CSCs developed from induced pluripotent stem cells. Here, we show that metformin suppresses the self-renewal and tube formation abilities of CSCs. We also show that metformin reduces the percentage of cells in the S phase increasing the percentage of cells in G0/G1 phase. Moreover, the tumorigenicity of CSCs was found to be attenuated when grafted in vivo after treatment with metformin.


Asunto(s)
Antineoplásicos/farmacología , Autorrenovación de las Células/efectos de los fármacos , Metformina/farmacología , Modelos Biológicos , Células Madre Pluripotentes/citología , Animales , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Tumorales Cultivadas
10.
Br J Cancer ; 122(9): 1378-1390, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32203212

RESUMEN

BACKGROUND: Liver cancer is the second most common cause of cancer-related death. Every type of tumours including liver cancer contains cancer stem cells (CSCs). To date, the molecular mechanism regulating the development of liver CSCs remains unknown. METHODS: In this study, we tried to generate a new model of liver CSCs by converting mouse induced pluripotent stem cells (miPSCs) with hepatocellular carcinoma (HCC) cell line Huh7 cells conditioned medium (CM). miPSCs treated with CM were injected into the liver of BALB/c nude mice. The developed tumours were then excised and analysed. RESULTS: The primary cultured cells from the malignant tumour possessed self-renewal capacity, differentiation potential and tumorigenicity in vivo, which were found rich in liver cancer-associated markers as well as CSC markers. CONCLUSIONS: We established a model of liver CSCs converting from miPS and showed different stages of stemness during conversion process. Our CSC model will be important to assess the molecular mechanisms necessary to develop liver CSCs and could help in defeating liver cancer.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Carcinoma Hepatocelular/genética , Medios de Cultivo Condicionados/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Neoplasias Hepáticas/genética , Animales , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/patología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Medios de Cultivo Condicionados/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/patología , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos BALB C , Células Madre Neoplásicas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
11.
Int J Mol Sci ; 20(5)2019 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-30818864

RESUMEN

Paclitaxel (PTX) is one of the front-line drugs approved for the treatment of ovarian cancer. However, the application of PTX is limited due to the significant hydrophobicity and poor pharmacokinetics. We previously reported target-directed liposomes carrying tumor-selective conjugated antibody and encapsulated glycosylated PTX (gPTX-L) which successfully overcome the PTX limitation. The tubulin stabilizing activity of gPTX was equivalent to that of PTX while the cytotoxic activity of gPTX was reduced. In human ovarian cancer cell lines, SK-OV-3 and OVK18, the concentration at which cell growth was inhibited by 50% (IC50) for gPTX range from 15⁻20 nM, which was sensitive enough to address gPTX-L with tumor-selective antibody coupling for ovarian cancer therapy. The cell membrane receptor CD44 is associated with cancer progression and has been recognized as a cancer stem cell marker including ovarian cancer, becoming a suitable candidate to be targeted by gPTX-L therapy. In this study, gPTX-loading liposomes conjugated with anti-CD44 antibody (gPTX-IL) were assessed for the efficacy of targeting CD44-positive ovarian cancer cells. We successfully encapsulated gPTX into liposomes with the loading efficiency (LE) more than 80% in both of gPTX-L and gPTX-IL with a diameter of approximately 100 nm with efficacy of enhanced cytotoxicity in vitro and of convenient treatment in vivo. As the result, gPTX-IL efficiently suppressed tumor growth in vivo. Therefore gPTX-IL could be a promising formulation for effective ovarian cancer therapies.


Asunto(s)
Receptores de Hialuranos/metabolismo , Terapia Molecular Dirigida , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Paclitaxel/uso terapéutico , Anticuerpos Monoclonales/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Glicosilación , Humanos , Liposomas/ultraestructura , Neoplasias Ováricas/patología , Paclitaxel/farmacología
12.
Clin Lab ; 64(10): 1685-1693, 2018 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-30336529

RESUMEN

BACKGROUND: Liver fibrosis is a dynamic procedure that results from an irregularity between fibrogenesis and fibrolysis. After time this procedure can lead to cirrhosis of the liver. Liver fibrosis and cirrhosis assessment is very important for both therapeutic decisions and prognostic evaluations. In this study, we tried to use serum ferritin (SF) together with five fibrosis tests (Age-Platelet index (API), aspartate aminotransferase to alanine aminotransferase ratio (AAR), AST to platelet ratio index (APRI), Fibrosis 4 score (FIB-4), and fibro-quotient (Fibro-Q)) to assess liver fibrosis and cirrhosis and estimate possible correlation between inflammation and SF. METHODS: This study was carried out on eighty-eight patients infected with HCV and twenty healthy subjects as a control. Complete blood count (CBC), aspartate aminotransferase (AST), alanine aminotransferase (ALT), antiHCV antibody, detection of HCV RNA by real-time PCR, and serum ferritin (SF) were assessed. Then API, ARR, APRI, FIB-4, and Fibro-Q were calculated. Different fibrosis stages (mild fibrosis stage (F1), moderate fibrosis stage (F2), severe fibrosis stage (F3), cirrhotic stage (F4)) were assessed using transient elastography by Fibro Scan®. RESULTS: FIB-4 index was significantly elevated (p < 0.01) with the progression of liver fibrosis at F1, F2, F3, and F4 when compared to healthy control group. The APRI score elevation between F0 and F3 and between F0 and F4 was significant (p < 0.01). SF was elevated in all fibrosis stages and significantly (p < 0.01) at F3 and F4 compared to controls. CONCLUSIONS: APRI coupled with SF should be the best reliable biomarkers for liver cirrhosis. Simultaneously, from our data SF involved in all stages of inflammation. Therefore, down regulation of ferritin in the early stage of fibrosis should be helpful in decreasing the inflammatory effect of ferritin.


Asunto(s)
Biomarcadores/sangre , Ferritinas/sangre , Anticuerpos contra la Hepatitis C/sangre , Cirrosis Hepática/sangre , ARN Viral/sangre , Adulto , Alanina Transaminasa/sangre , Aspartato Aminotransferasas/sangre , Plaquetas/metabolismo , Fibrosis , Hepacivirus/genética , Hepacivirus/inmunología , Hepacivirus/fisiología , Anticuerpos contra la Hepatitis C/inmunología , Humanos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/virología , Persona de Mediana Edad , Recuento de Plaquetas , ARN Viral/genética , Curva ROC
13.
Int J Mol Sci ; 19(11)2018 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-30373174

RESUMEN

Cripto-1 is a glycophosphatidylinositol (GPI) anchored signaling protein of epidermal growth factor (EGF)-Cripto-1-FRL1-Cryptic (CFC) family and plays a significant role in the early developmental stages and in the different types of cancer cells, epithelial to mesenchymal transition and tumor angiogenesis. Previously, we have developed cancer stem cells (miPS-LLCcm) from mouse iPSCs by culturing them in the presence of conditioned medium of Lewis Lung Carcinoma (LLC) cells for four weeks. Nodal and Cripto-1 were confirmed to be expressed in miPS-LLCcm cells by quantitative reverse transcription PCR (rt-qPCR) implying that Cr-1 was required in maintaining stemness. To investigate the biological effect of adding exogenous soluble CR-1 to the cancer stem cells, we have prepared a C-terminally truncated soluble form of recombinant human CR-1 protein (rhsfCR-1), in which the GPI anchored moiety was removed by substitution of a stop codon through site-directed mutagenesis. rhsfCR-1 effectively suppressed the proliferation and sphere forming ability of miPS-LLCcm cells in a dose-dependent manner in the range of 0 to 5 µg/mL, due to the suppression of Nodal-Cripto-1/ALK4/Smad2 signaling pathway. Frequency of sphere-forming cells was dropped from 1/40 to 1/69 by rhsfCR-1 at 1 µg/mL. Moreover, rhsfCR-1 in the range of 0 to 1 µg/mL also limited the differentiation of miPS-LLCcm cells into vascular endothelial cells probably due to the suppression of self-renewal, which should reduce the number of cells with stemness property. As demonstrated by a soluble form of exogenous Cripto-1 in this study, the efficient blockade would be an attractive way to study Cripto-1 dependent cancer stem cell properties for therapeutic application.


Asunto(s)
Autorrenovación de las Células , Proteínas Ligadas a GPI/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/citología , Animales , Diferenciación Celular , Línea Celular , Humanos , Ratones , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal , Proteína Smad2/metabolismo
14.
Cytotechnology ; 75(3): 243-253, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37181678

RESUMEN

Induced pluripotent stem cells (iPSCs) are useful tools for modeling diseases and developing personalized medicine. We have been developing cancer stem cells (CSCs) from iPSCs with conditioned medium (CM) of cancer-derived cells as the mimicry of the microenvironment of tumor initiation. However, the conversion of human iPSCs has not always been efficient with only CM. In this study, human iPSCs reprogrammed from monocytes of healthy volunteers were cultured in a media containing 50% of the CM from human pancreatic cancer derived BxPC3 cells supplemented with a MEK inhibitor (AZD6244) and a GSK-3α/ß inhibitor (CHIR99021). The survived cells were assessed for the characteristics of CSCs in vitro and in vivo. As a result, they exhibited CSC phenotypes of self-renewal, differentiation, and malignant tumorigenicity. Primary culture of the malignant tumors of the converted cells exhibited the elevated expression of CSC related genes CD44, CD24 and EPCAM maintaining the expression of stemness genes. In conclusion, the inhibition of GSK-3α/ß and MEK and the microenvironment of tumor initiation mimicked by the CM can convert human normal stem cells into CSCs. This study could provide insights into establishing potentially novel personalized cancer models which could help investigate the tumor initiation and screening of personalized therapies on CSCs. Supplementary Information: The online version contains supplementary material available at 10.1007/s10616-023-00575-1.

15.
Biomaterials ; 301: 122249, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37506511

RESUMEN

The heterogeneous cell population in the stromal microenvironment is considered to be attributed to the multiple sources from which the cells originate. Tumor associated myoepithelial cells (TAMEs) are one of the most important populations in the tumor microenvironment (TME) especially in breast cancer. On the other hand, cancer stem cells (CSCs) have previously been described to be the origin of tumor-associated cellular components in the TME. We prepared a cancer stem cell model converting mouse-induced pluripotent stem cells (miPSCs) in the presence of conditioned medium of breast cancer cell line MDA-MB-231 cells. The converted cells developed tumors progressing into invasive carcinoma with ductal carcinoma in situ (DCIS) like structure when transplanted into mouse mammary fat pads. The primary cultured cells from the tumor further exhibited markers of CSC such as Sox2, Oct3/4, - CD133 and EpCAM, and mammary gland-related TAME markers such as α-smooth muscle actin, cytokeratin 8, whey acidic protein, prolactin receptor and progesterone receptor as well. These results indicated that the CSCs could be an origin of TAMEs contributing to mammary gland epithelial cell differentiation and the progression to invasive carcinoma during tumor development. The gene expression profiles confirmed the enhanced signaling pathways of PI3K/AKT and MAPK, which have been demonstrated to be enriched in the CSC models, together with the estrogen receptor signaling which was peculiar to mammary gland-derived character.


Asunto(s)
Carcinoma Intraductal no Infiltrante , Ratones , Animales , Carcinoma Intraductal no Infiltrante/patología , Microambiente Tumoral , Fosfatidilinositol 3-Quinasas , Biomarcadores de Tumor , Células Madre Neoplásicas/patología
16.
Methods Mol Biol ; 2549: 399-407, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33755909

RESUMEN

Cancer stem cells (CSCs) are capable of continuous proliferation and self-renewal and are proposed to play significant roles in oncogenesis, tumor growth, metastasis, and cancer recurrence. CSCs are considered derived from normal stem cells affected by the inflammatory microenvironment. Stem cells, are considered to be induced into progenitor cells, which differentiate into various normal phenotypes depending on the normal niche. We hypothesized that CSCs could be derived from stem cells in the cancer-inducing niche, which is a condition of chronic inflammation rich in growth factors, interleukins, chemokines, etc. Exosomes are considered to be the key mediators responsible for the cell-to-cell communications carrying proteins, nucleic acids, metabolites, etc., to shuttle between cells. If these cells are in the environment of chronic inflammation, the exosomes should be reflecting the conditions. In this chapter, we detail the method of CSC initiation using extracellular vesicles (EVs) derived from cancer cell. The stem cells treated with the EVs acquired characteristics of CSCs showing spheroids expressing stemness markers in the suspension culture and high tumorigenicity in Balb/c nude mice. EVs might perform as suitable inducer for initiating CSCs from stem cells or progenitor cells. The model of CSCs and the procedure of their establishment with EVs will help study the exact effect of EVs in the cancer-inducing niche and tumor microenvironment.


Asunto(s)
Vesículas Extracelulares , Recurrencia Local de Neoplasia , Animales , Vesículas Extracelulares/metabolismo , Inflamación/metabolismo , Ratones , Ratones Desnudos , Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/patología , Microambiente Tumoral
17.
Sci Rep ; 12(1): 15628, 2022 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-36115905

RESUMEN

Previously, our group has demonstrated establishment of Cancer Stem Cell (CSC) models from stem cells in the presence of conditioned medium of cancer cell lines. In this study, we tried to identify the factors responsible for the induction of CSCs. Since we found the lipid composition could be traced to arachidonic acid cascade in the CSC model, we assessed prostaglandin E2 (PGE2) as a candidate for the ability to induce CSCs from induced pluripotent stem cells (iPSCs). Mouse iPSCs acquired the characteristics of CSCs in the presence of 10 ng/mL of PGE2 after 4 weeks. Since constitutive Akt activation and pik3cg overexpression were found in the resultant CSCs, of which growth was found independent of PGE2, chronic stimulation of the receptors EP-2/4 by PGE2 was supposed to induce CSCs from iPSCs through epigenetic effect. The bioinformatics analysis of the next generation sequence data of the obtained CSCs proposed not only receptor tyrosine kinase activation by growth factors but also extracellular matrix and focal adhesion enhanced PI3K pathway. Collectively, chronic stimulation of stem cells with PGE2 was implied responsible for cancer initiation enhancing PI3K/Akt axis.


Asunto(s)
Dinoprostona , Neoplasias , Animales , Ácido Araquidónico/metabolismo , Medios de Cultivo Condicionados/farmacología , Dinoprostona/metabolismo , Dinoprostona/farmacología , Ratones , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
18.
Sci Rep ; 12(1): 347, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013447

RESUMEN

Cancer stem cells (CSCs) are capable of continuous proliferation, self-renewal and are proposed to play significant roles in oncogenesis, tumor growth, metastasis and cancer recurrence. We have established a model of CSCs that was originally developed from mouse induced pluripotent stem cells (miPSCs) by proposing miPSCs to the conditioned medium (CM) of cancer derived cells, which is a mimic of carcinoma microenvironment. Further research found that not only PI3K-Akt but also EGFR signaling pathway was activated during converting miPSCs into CSCs. In this study, we tried to observe both of PI3Kγ inhibitor Eganelisib and EGFR inhibitor Gefitinib antitumor effects on the models of CSCs derived from miPSCs (miPS-CSC) in vitro and in vivo. As the results, targeting these two pathways exhibited significant inhibition of cell proliferation, self-renewal, migration and invasion abilities in vitro. Both Eganelisib and Gefitinib showed antitumor effects in vivo while Eganelisib displayed more significant therapeutic efficacy and less side effects than Gefitinib on all miPS-CSC models. Thus, these data suggest that the inhibitiors of PI3K and EGFR, especially PI3Kγ, might be a promising therapeutic strategy against CSCs defeating cancer in the near future.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Receptores ErbB/antagonistas & inhibidores , Gefitinib/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Receptores ErbB/metabolismo , Femenino , Células Madre Pluripotentes Inducidas/enzimología , Células Madre Pluripotentes Inducidas/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Transducción de Señal , Carga Tumoral/efectos de los fármacos
19.
J Exp Clin Cancer Res ; 41(1): 29, 2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35063003

RESUMEN

BACKGROUND: Cancer stem cells (CSCs) are generated under irregular microenvironment in vivo, of which mimic is quite difficult due to the lack of enough information of the factors responsible for cancer initiation. Here, we demonstrated that mouse induced pluripotent cells (miPSCs) reprogrammed from normal embryonic fibroblasts were susceptible to the microenvironment affected by cancer cells to convert into CSCs in vivo. METHODS: Three different pancreatic cancer line cells, BxPC3, PANC1, and PK8 cells were mixed with miPSCs and subcutaneously injected into immunodeficient mice. Tumors were evaluated by histological analysis and cells derived from iPSCs were isolated and selected from tumors. The isolated cells were characterized for cancer stem cell characters in vitro and in vivo as well as their responses to anticancer drugs. The impact of co-injection of iPSCs with cancer cells on transcriptome and signaling pathways of iPSCs was investigated. RESULTS: The injection of miPSCs mixed with human pancreatic cancer cells into immunodeficient mice maintained the stemness of miPSCs and changed their phenotype. The miPSCs acquired CSC characteristics of tumorigenicity and self-renewal. The drug responses and the metastatic ability of CSCs converted from miPSCs varied depending on the microenvironment of cancer cells. Interestingly, transcriptome profiles of these cells indicated that the pathways related with aggressiveness and energy production were upregulated from the levels of miPSCs. CONCLUSIONS: Our result suggests that cancer-inducing microenvironment in vivo could rewire the cell signaling and metabolic pathways to convert normal stem cells into CSCs.


Asunto(s)
Expresión Génica/genética , Células Madre Pluripotentes Inducidas/metabolismo , Redes y Vías Metabólicas/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Microambiente Tumoral
20.
Expert Rev Anticancer Ther ; 21(6): 605-619, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33857392

RESUMEN

Introduction: The phosphatidylinositol 3-kinase/protein kinase-B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway is a fundamental regulator of cell proliferation and survival. Dysregulation in this pathway leads to the development of cancer. Accumulating evidence indicates that dysregulation in this pathway is involved in cancer initiation, progression, and recurrence. However, the pathway consists of various signal transducing factors related with cellular events, such as transformation, tumorigenesis, cancer progression, and drug resistance. Therefore, it is very important to determine the targets in this pathway for cancer therapy. Although many drugs inhibiting this signaling pathway are in clinical trials or have been approved for treating solid tumors and hematologic malignancies, further understanding of the signaling mechanism is required to achieve better therapeutic efficacy.Areas covered: In this review, we have describe the PI3K/AKT/mTOR pathway in detail, along with its critical role in cancer stem cells, for identifying potential therapeutic targets. We also summarize the recent developments in different types of signaling inhibitors.Expert opinion: Downregulation of the PI3K/AKT/mTOR pathway is very important for treating all types of cancers. Thus, further studies are required to establish novel prognostic factors to support the current progress in cancer treatment with emphasis on this pathway.


Asunto(s)
Neoplasias , Proteínas Proto-Oncogénicas c-akt , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/uso terapéutico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA