Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Genes Cells ; 29(3): 231-253, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38253356

RESUMO

The cell-assisted lipotransfer technique, integrating adipose-derived mesenchymal stem cells (ADMSCs), has transformed lipofilling, enhancing fat graft viability. However, the multipotent nature of ADMSCs poses challenges. To improve safety and graft vitality and to reduce unwanted lineage differentiation, this study refines the methodology by priming ADMSCs into preadipocytes-unipotent, self-renewing cells. We explored the impact of fibroblast growth factor-1 (FGF-1), fibroblast growth factor-2 (FGF-2), and epidermal growth factor (EGF), either alone or in combination, on primary human ADMSCs during the proliferative phase. FGF-2 emerged as a robust stimulator of cell proliferation, preserving stemness markers, especially when combined with EGF. Conversely, FGF-1, while not significantly affecting cell growth, influenced cell morphology, transitioning cells to a rounded shape with reduced CD34 expression. Furthermore, co-priming with FGF-1 and FGF-2 enhanced adipogenic potential, limiting osteogenic and chondrogenic tendencies, and possibly promoting preadipocyte commitment. These preadipocytes exhibited unique features: rounded morphology, reduced CD34, decreased preadipocyte factor 1 (Pref-1), and elevated C/EBPα and PPARγ, alongside sustained stemness markers (CD73, CD90, CD105). Mechanistically, FGF-1 and FGF-2 activated key adipogenic transcription factors-C/EBPα and PPARγ-while inhibiting GATA3 and Notch3, which are adipogenesis inhibitors. These findings hold the potential to advance innovative strategies for ADMSC-mediated lipofilling procedures.


Assuntos
Fator 1 de Crescimento de Fibroblastos , Células-Tronco Mesenquimais , Humanos , Adipogenia , Diferenciação Celular , Células Cultivadas , Fator de Crescimento Epidérmico/farmacologia , Fator 1 de Crescimento de Fibroblastos/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , PPAR gama/metabolismo
2.
Genes Cells ; 27(1): 25-42, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34837452

RESUMO

Cancer stem cells are a promising target for cancer eradication due to their responsibility for therapy-resistance and cancer recurrence. Previously, we have demonstrated that glioma stem cells (GSCs) recruit and induce the differentiation of bone marrow (BM) monocytes into tumor-infiltrating macrophages, which phagocytose hemorrhaged erythrocytes and store GSC-beneficial iron in mouse xenografts, suggesting a self-expanding strategy of GSCs that exploits host hematopoiesis of myeloid cells. However, it remains unclear whether a self-advantageous effect of GSCs also occurs on erythroid cells during glioma development. Here, we found that, in the primary cultures of mouse fetal liver proerythroblasts (proEs), conditioned media prepared from glioma cells including patient-derived glioblastoma (GBM) cells significantly facilitated the differentiation of proEs into erythroblasts. Importantly, in-vivo erythroid analysis in intracranially GSC-transplanted mice showed an enhanced erythropoiesis in the BM. In addition, the sphere forming ability of patient-derived GBM cells was significantly suppressed by hypoxia treatment and iron chelation, suggesting higher demands of GSCs for oxygen and iron, which may be supplied by GSCs- and their progeny-induced erythrocyte production. Our findings provide a new insight into survival and expanding strategies of GSCs that systemically exploit host erythropoiesis.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Animais , Eritropoese , Humanos , Camundongos , Células-Tronco Neoplásicas
3.
Stem Cells ; 38(8): 921-935, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32346916

RESUMO

Spontaneous necrosis is a defining feature of glioblastomas (GBMs), the most malignant glioma. Despite its strong correlations with poor prognosis, it remains unclear whether necrosis could be a possible cause or mere consequence of glioma progression. Here we isolated a particular fraction of necrotic products spontaneously arising from glioma cells, morphologically and biochemically defined as autoschizis-like products (ALPs). When administered to granulocyte macrophage colony-stimulating factor (GM-CSF)-primed bone marrow-derived macrophage/dendritic cells (Mφ/DCs), ALPs were found to be specifically engulfed by Mφs expressing a tumor-associated macrophage (TAM) marker CD204. ALPs from glioma stem cells (GSCs) had higher activity for the TAM development than those from non-GSCs. Of note, expression of the Il12b gene encoding a common subunit of IL-12/23 was upregulated in ALPs-educated Mφs. Furthermore, IL-12 protein evidently enhanced the sphere-forming activity of GBM patient-derived cells, although interestingly IL-12 is generally recognized as an antitumoral M1-Mφ marker. Finally, in silico analysis of The Cancer Genome Atlas (TCGA) transcriptome data of primary and recurrent GBMs revealed that higher expression of these IL-12 family genes was well correlated with more infiltration of M1-type TAMs and closely associated with poorer prognosis in recurrent GBMs. Our results highlight a role of necrosis in GSC-driven self-beneficial niche construction and glioma progression, providing important clues for developing new therapeutic strategies against gliomas.


Assuntos
Glioma/genética , Proteína Goosecoid/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Macrófagos Associados a Tumor/metabolismo , Animais , Feminino , Humanos , Camundongos , Transdução de Sinais
4.
Genes Cells ; 21(3): 241-51, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26805963

RESUMO

Cancer stem cells (CSCs) are maintained under special microenvironment called niche, and elucidation and targeting of the CSC niche will be a feasible strategy for cancer eradication. Tumor-associated macrophages (TAMs) are known to be involved in cancer progression and thus can be a component of CSC niche. Although TAMs are known to play multiple roles in tumor progression, involvement of CSCs in TAM development fully remains to be elucidated. Using rat C6 glioma side population (SP) cells as a model of glioma CSCs, we here show that CSCs induce the TAM development by promoting survival and differentiation of bone marrow-derived monocytes. CSC-induced macrophages can be separated into two distinct subsets of cells, CD11c(low) and CD11c(high) cells. Interestingly, only the CD11c(high) subset of cells have protumoral activity, as shown by intracranial transplantation into immune-deficient mice together with CSCs. These CD11c(high) macrophages were observed in the tumor formed by co-transplantation with CSCs. Furthermore, CSCs produced GM-CSF and anti-GM-CSF antibody inhibited CSC-induced TAM development. In conclusion, CSCs have the ability to self-create their own niche involving TAMs through CSC-derived GM-CSF, which can thus be a therapeutic target in view of CSC niche disruption.


Assuntos
Neoplasias Encefálicas/metabolismo , Antígenos CD11/metabolismo , Glioma/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Macrófagos/citologia , Células-Tronco Neoplásicas/transplante , Animais , Neoplasias Encefálicas/patologia , Antígenos CD11/genética , Diferenciação Celular , Linhagem Celular Tumoral , Feminino , Glioma/patologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Monócitos/citologia , Monócitos/metabolismo , Células-Tronco Neoplásicas/metabolismo , Ratos , Nicho de Células-Tronco
5.
Stem Cells ; 34(5): 1151-62, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26822103

RESUMO

Cancer stem cells (CSCs) are believed to be maintained within a microenvironmental niche. Here we used polymer microarrays for the rapid and efficient identification of glioma CSC (GSC) niche mimicries and identified a urethane-based synthetic polymer, upon which two groups of niche components, namely extracellular matrices (ECMs) and iron are revealed. In cultures, side population (SP) cells, defined as GSCs in the rat C6 glioma cell line, are more efficiently sustained in the presence of their differentiated progenies expressing higher levels of ECMs and transferrin, while in xenografts, ECMs are supplied by the vascular endothelial cells (VECs), including SP cell-derived ones with distinctively greater ability to retain xenobiotics than host VECs. Iron is stored in tumor infiltrating host macrophages (Mφs), whose protumoral activity is potently enhanced by SP cell-secreted soluble factor(s). Finally, coexpression of ECM-, iron-, and Mφ-related genes is found to be predictive of glioma patients' outcome. Our polymer-based approach reveals the intrinsic capacities of GSCs, to adapt the environment to organize a self-advantageous microenvironment niche, for their maintenance and expansion, which redefines the current concept of anti-CSC niche therapy and has the potential to accelerate cancer therapy development. Stem Cells 2016;34:1151-1162.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Polímeros/farmacologia , Nicho de Células-Tronco , Alicerces Teciduais/química , Animais , Neoplasias Encefálicas/genética , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/genética , Humanos , Ferro/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Poliuretanos/farmacologia , Ratos , Receptores Depuradores Classe A/genética , Receptores Depuradores Classe A/metabolismo , Células da Side Population/citologia , Células da Side Population/efeitos dos fármacos , Nicho de Células-Tronco/efeitos dos fármacos , Nicho de Células-Tronco/genética , Transferrina/metabolismo , Resultado do Tratamento
6.
BMC Cancer ; 16(1): 847, 2016 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-27814696

RESUMO

BACKGROUND: Elucidating the precise properties of cancer stem cells (CSCs) is indispensable for the development of effective therapies against tumors, because CSCs are key drivers of tumor development, metastasis and relapse. We previously reported that the Hoechst 33342 dye-low staining side population (SP) method can enrich for CSCs in the C6 glioma cell line, and that the positively stained main population (MP) cells are non-CSCs. Presence of cancer stem-like SP cells is reported in various types of cancer. Although altered cellular energy metabolism is a hallmark of cancer, very little has been studied on the applicability of fluorescent probes for the understanding of CSC energy metabolism. METHODS: The metabolic status of C6 SP and MP cells are evaluated by CellROX, MitoTracker Green (MTG) and JC-1 for cellular oxidative stress, mitochondrial amount, and mitochondrial membrane potential, respectively. RESULTS: SP cells were found to exhibit significantly lower fluorescent intensities of CellROX and MTG than MP cells. However, inhibition of ATP binding cassette (ABC) transporters by verapamil enhanced the intensities of these probes in SP cells to the levels similar to those in MP cells, indicating that SP cells expel the probes outside of the cells through ABC transporters. Next, SP cells were stained with JC-1 dye which exhibits membrane potential dependent accumulation in mitochondrial matrix, followed by formation of aggregates. The mitochondrial membrane potential indicated by the aggregates of JC-1 was 5.0-fold lower in SP cells than MP cells. Inhibition of ABC transporters enhanced the fluorescent intensities of the JC-1 aggregates in both SP and MP cells, the former of which was still 2.2-fold lower than the latter. This higher JC-1 signal in MP cells was further found to be due to the Hoechst 33342 dye existing in MP cells. When SP and MP cells were recultured to deprive the intracellular Hoechst 33342 dye and then stained with JC-1 in the presence of verapamil, the intensities of JC-1 aggregates in such SP and MP cells became comparable. CONCLUSION: Inhibiting ABC transporters and depriving Hoechst 33342 dye are required for the accurate assessment of side population-defined C6 glioma stem cell metabolism using fluorescent probes.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Glioma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células da Side Population/metabolismo , Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Animais , Biomarcadores , Linhagem Celular Tumoral , Corantes Fluorescentes , Glioma/patologia , Humanos , Imunofenotipagem , Células-Tronco Neoplásicas/patologia
7.
Stem Cells ; 32(6): 1602-15, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24302516

RESUMO

Self-renewing proliferation of neural stem cells (NSCs) is intimately linked to the inhibition of neuronal and glial differentiation, however, their molecular linkage has been poorly understood. We have proposed a model previously explaining partly this linkage, in which fibroblast growth factor 2 (FGF2) and Wnt signals cooperate to promote NSC self-renewal via ß-catenin accumulation, which leads to the promotion of proliferation by lymphoid enhancer factor (LEF)/T-cell factor (TCF)-mediated cyclin D1 expression and at the same time to the inhibition of neuronal differentiation by ß-catenin-mediated potentiation of Notch signaling. To fully understand the mechanisms underlying NSC self-renewal, it needs to be clarified how these growth factor signals inhibit glial differentiation as well. Here, we demonstrate that cyclin D1, a NSC growth promoting signaling component and also a common component of FGF2 and Wnt signaling pathways, inhibits astroglial differentiation of NSCs. Interestingly, this effect of cyclin D1 is mediated even though its cell cycle progression activity is blocked. Forced downregulation of cyclin D1 enhances astrogliogenesis of NSCs in culture and in vivo. We further demonstrate that cyclin D1 binds to STAT3, a transcription factor downstream of astrogliogenic cytokines, and suppresses its transcriptional activity on the glial fibrillary acidic protein (Gfap) gene. Taken together with our previous finding, we provide a novel molecular mechanism for NSC self-renewal in which growth promoting signaling components activated by FGF2 and Wnts inhibit neuronal and glial differentiation.


Assuntos
Astrócitos/citologia , Ciclina D1/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese , Transdução de Sinais , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/enzimologia , Proteína Morfogenética Óssea 2/farmacologia , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Proteína p300 Associada a E1A/metabolismo , Ativação Enzimática/efeitos dos fármacos , Feminino , Proteína Glial Fibrilar Ácida , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Fator Inibidor de Leucemia/farmacologia , Camundongos Endogâmicos ICR , Proteínas do Tecido Nervoso/genética , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Estabilidade Proteica/efeitos dos fármacos , Interferência de RNA/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética , beta Catenina/metabolismo
8.
Gynecol Oncol ; 129(3): 598-605, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23500085

RESUMO

GOALS: We previously demonstrated that side-population (SP) cells in human endometrial cancer cells (Hec1 cells) and in rat endometrial cells expressing oncogenic human K-Ras protein (RK12V cells) have features of cancer stem cells (CSCs). Hec1-SP cells showed enhanced migration and the potential to differentiate into the mesenchymal cell lineage. In this study, we analyzed the association of the epithelial-mesenchymal transition (EMT) with the properties of these endometrial CSCs. We also assessed the effects of salinomycin (a compound with EMT-specific toxicity) on the proliferative capacity, migration and invasiveness of these endometrial CSCs using Hec1-SP cells. METHOD: We performed microarray expression analysis to screen for up-regulated genes in CSCs using a set of RK12V-SP cells and -non-SP(NSP) cells and used the Metacore package to identify the Gene GO pathway MAPs involved in the up-regulated genes. To analyze their association with EMT, the expression of several EMT associated genes in Hec1-SP cells was investigated by real time PCR and compared with that in Hec1-NSP cells. We assessed the expression of BAX, BCL2, LEF1, cyclinD and fibronectin by real time PCR. We also evaluated the viabilities, migration and invasive activities, and tumorigenicities of these SP cells and NSP cells in the presence or absence of salinomycin. RESULTS: We demonstrated that i) EMT processes were observed in both RK12V-SP cells and Hec1-SP cells, ii) the level of fibronectin was enhanced in Hec1-SP cells and salinomycin reduced the level of fibronectin expression, iii) salinomycin induced apoptosis and inhibited Wnt signaling, and iv) salinomycin inhibited the proliferation, migration, invasiveness and tumorigenicity of these SP cells. CONCLUSION: This is the first report of an inhibitory effect of salinomycin on the properties of endometrial CSCs.


Assuntos
Movimento Celular/efeitos dos fármacos , Neoplasias do Endométrio/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Piranos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Endométrio/efeitos dos fármacos , Endométrio/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Feminino , Fibronectinas/biossíntese , Humanos , Invasividade Neoplásica , Células-Tronco Neoplásicas/patologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteínas Wnt/metabolismo , Proteína X Associada a bcl-2/biossíntese
9.
Adv Exp Med Biol ; 777: 73-85, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23161076

RESUMO

A pentaspan membrane glycoprotein prominin-1 (frequently called CD133 in human) is widely used as a surface marker to identify and isolate normal stem/progenitor cells from various organs, although it is also expressed in some types of differentiated cells. Since CD133 was identified as a universal marker to isolate cancer stem cells (CSCs) in tumors derived from multiple tissues, much attention has been directed toward the relationship between its gene regulation and identity of CSCs (i.e., cancer stemness). Prominin-1 (PROM1) gene possesses five alternative promoters yielding multiple first exons within the 5'-untranslated region (UTR) and also splicing variants affecting the open reading frame (ORF) sequence, implicating the complicated gene regulation in a context-dependent manner. This chapter aims to organize the accumulated findings on prominin-1 with a focus on its altered expression and regulation in normal and cancerous cells and to discuss potential regulatory networks underlying cancer stemness.


Assuntos
Regulação da Expressão Gênica , Células-Tronco Neoplásicas , Éxons , Humanos , Glicoproteínas de Membrana/metabolismo , Células-Tronco Neoplásicas/metabolismo , Regiões Promotoras Genéticas
10.
Inflamm Regen ; 43(1): 46, 2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37759310

RESUMO

BACKGROUND: Pancreatic adenocarcinoma (PAAD) is one of the most fatal human cancers, but effective therapies remain to be established. Cancer stem cells (CSCs) are highly resistant to anti-cancer drugs and a deeper understanding of their microenvironmental niche has been considered important to provide understanding and solutions to cancer eradication. However, as the CSC niche is composed of a wide variety of biological and physicochemical factors, the development of multidisciplinary tools that recapitulate their complex features is indispensable. Synthetic polymers have been studied as attractive biomaterials due to their tunable biofunctionalities, while hydrogelation technique further renders upon them a diversity of physical properties, making them an attractive tool for analysis of the CSC niche. METHODS: To develop innovative materials that recapitulate the CSC niche in pancreatic cancers, we performed polymer microarray analysis to identify niche-mimicking scaffolds that preferentially supported the growth of CSCs. The niche-mimicking activity of the identified polymers was further optimized by polyethylene glycol (PEG)-based hydrogelation. To reveal the biological mechanisms behind the activity of the optimized hydrogels towards CSCs, proteins binding onto the hydrogel were analyzed by liquid chromatography with tandem mass spectrometry (LC-MS/MS), and the potential therapeutic targets were validated by looking at gene expression and patients' outcome in the TCGA database. RESULTS: PA531, a heteropolymer composed of 2-methoxyethyl methacrylate (MEMA) and 2-(diethylamino)ethyl methacrylate (DEAEMA) (5.5:4.5) that specifically supports the growth and maintenance of CSCs was identified by polymer microarray screening using the human PAAD cell line KLM1. The polymer PA531 was converted into five hydrogels (PA531-HG1 to HG5) and developed to give an optimized scaffold with the highest CSC niche-mimicking activities. From this polymer that recapitulated CSC binding and control, the proteins fetuin-B and angiotensinogen were identified as candidate target molecules with clinical significance due to the correlation between gene expression levels and prognosis in PAAD patients and the proteins associated with the niche-mimicking polymer. CONCLUSION: This study screened for biofunctional polymers suitable for recapitulation of the pancreatic CSC niche and one hydrogel with high niche-mimicking abilities was successfully fabricated. Two soluble factors with clinical significance were identified as potential candidates for biomarkers and therapeutic targets in pancreatic cancers. Such a biomaterial-based approach could be a new platform in drug discovery and therapy development against CSCs, via targeting of their niche.

11.
Cancer Invest ; 30(5): 390-7, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22571340

RESUMO

Synovial sarcoma (SS) is an aggressive tumor that accounts for almost 10% of all soft tissue sarcomas. In this study, we found the expression of CD133 in human SS specimens, thus, we focused on the function of CD133 in SS. Separation of the CD133-positive and -negative subpopulations in SS cell lines clarified that the CD133-negative subpopulation exhibited enhanced growth and hyperphosphorylation of AKT. Treatment of Akt inhibitor suppressed the cell growth of CD133-negative subpopulation to the levels of CD133-positive cells. These results suggest that CD133 has negative effect on the growth of cells through AKT-dependent signalling pathway.


Assuntos
Antígenos CD/fisiologia , Glicoproteínas/fisiologia , Peptídeos/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Sarcoma Sinovial/etiologia , Transdução de Sinais/fisiologia , Antígeno AC133 , Animais , Antígenos CD/análise , Linhagem Celular Tumoral , Feminino , Glicoproteínas/análise , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/análise
12.
Ann Diagn Pathol ; 16(2): 134-40, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21396870

RESUMO

A clear cell variant of solid-pseudopapillary tumor (SPT) of the pancreas was initially reported in 2006 as a tumor that arose in the pancreatic body and tail in young adults; to date, only 4 cases of this entity have been reported. Here, we present the case of a 58-year-old man with clear cell variant of SPT with distinctive clinicopathologic features. The tumor was well demarcated, was 2.6 cm in size and mostly composed of multivacuolated clear cells with solid growth, and exhibited the characteristic immunohistochemical positivity of ß-catenin in the cytoplasm and nuclei of the neoplastic cells. In contrast to classical SPT with nuclear positivity, this case was negative for E-cadherin. Direct DNA sequencing of exon 3 of ß-catenin gene demonstrated a single amino acid substitution (serine to phenylalanine) in codon 37, which is the phosphorylation site by GSKß and frequently found in classical SPT. Electron microscopy demonstrated enlarged mitochondria and endoplasmic reticulum. Despite the fact that previous cases of clear cell variant of SPT arose mainly in the pancreatic body and tail in female young adults (age, 26-32 years), this case suggested that it is possible for a clear cell variant of SPT to arise in the pancreatic head in a middle-aged man. Because the recognition of the clear cell variant of SPT is important for the appropriate diagnosis of primary pancreatic tumor, the present case with its distinctive characteristics may provide new information for a more profound understanding of the pancreatic SPT.


Assuntos
Adenocarcinoma de Células Claras/patologia , Carcinoma Papilar/patologia , Neoplasias Pancreáticas/patologia , Adenocarcinoma de Células Claras/genética , Adenocarcinoma de Células Claras/metabolismo , Carcinoma Papilar/genética , Carcinoma Papilar/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Mutação , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , beta Catenina/genética
13.
Inflamm Regen ; 42(1): 54, 2022 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-36451253

RESUMO

For all living organisms, the adaptation to outside environments is an essential determinant to survive natural and artificial selections and to sustain the whole ecosystem intact with functional biodiversity. Likewise, cancer cells have similar characteristics that evade not only stresses from the host-internal innate and adaptive immune systems but also those from host-externally administered therapeutic interventions. Such selfish characteristics of cancer cells lead to the formation of cancerous ecosystem with a wide variety of phenotypic heterogeneity, which should be called cancer "egosystem" from the host point of view. Recently increasing evidence demonstrates that cancer stem cells (CSCs) are responsible for this cancer egosystem by effectively exploiting host inflammatory and hematopoietic cells and thereby reconstructing their own advantageous niches, which may well be a driving force in cancer recurrence. CSCs are further likely to render multiple niches mutually interconnected and cooperating as a network to support back CSCs themselves. Here, we summarize a recently identified iron-replenishing niche network self-organized by glioma CSCs (GSCs) through remote regulation of host myeloid and erythroid lineage cells. GSCs recruit bone marrow (BM)-derived inflammatory monocytes into tumor parenchyma, facilitate their differentiation into macrophages (Mφs) and skew their polarization into pro-tumoral phenotype, i.e., tumor-associated Mφs (TAMs). Meanwhile, GSCs distantly enhance erythropoiesis in host hematopoietic organs like BM and spleen potentially by secreting some soluble mediators that maintain continuous supply of erythrocytes within tumors. In addition, as normal red pulp Mφs (RPMs) under steady state conditions in spleen recycle iron by phagocytosing the aged or damaged erythrocytes (a/dECs) and release it in time of need, TAMs at least in gliomas phagocytose the hemorrhaged erythrocytes within tumors and potentially serve as a source of iron, an important nutrient indispensable to GSC survival and glioma progression. Taken together, these studies provide the substantial evidence that CSCs have a unique strategy to orchestrate multiple niches as an ecosystem that threatens the host living, which in this sense must be an egosystem. Targeting such an adaptive subpopulation of CSCs could achieve drastic disturbance of the CSC niches and subsequent extinction of malignant neoplasms.

14.
Inflamm Regen ; 42(1): 34, 2022 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-36324180

RESUMO

Due to increased resistance to standard chemo/radiotherapies and relapse, highly tumorigenic cancer stem cells (CSCs) have been proposed as a promising target for the development of effective cancer treatments. In order to develop innovative cancer therapies that target CSCs, much attention has focused on the iron metabolism of CSCs, which has been considered to contribute to self-renewal of CSCs. Here, we review recent advances in iron metabolism and conventional iron metabolism-targeted cancer therapies, as well as therapy resistance of CSCs and potential treatment options to overcome them, which provide important insights into therapeutic strategies against intractable cancers. Potential treatment options targeting iron homeostasis, including small-molecule inhibitors, nanotechnology platforms, ferroptosis, and 5-ALA-PDT, might be a focus of future research for the development of innovative cancer therapies that tackle CSCs.

15.
Cells ; 11(13)2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35805138

RESUMO

Glioblastoma multiforme (GBM) is the most incurable tumor (due to the difficulty in complete surgical resection and the resistance to conventional chemo/radiotherapies) that displays a high relapse frequency. Cancer stem cells (CSCs) have been considered as a promising target responsible for therapy resistance and cancer recurrence. CSCs are known to organize a self-advantageous microenvironment (niche) for their maintenance and expansion. Therefore, understanding how the microenvironment is reconstructed by the remaining CSCs after conventional treatments and how it eventually causes recurrence should be essential to inhibit cancer recurrence. However, the number of studies focusing on recurrence is limited, particularly those related to tumor immune microenvironment, while numerous data have been obtained from primary resected samples. Here, we summarize recent investigations on the immune microenvironment from the viewpoint of recurrent GBM (rGBM). Based on the recurrence-associated immune cell composition reported so far, we will discuss how CSCs manipulate host immunity and create the special microenvironment for themselves to regrow. An integrated understanding of the interactions between CSCs and host immune cells at the recurrent phase will lead us to develop innovative therapies and diagnoses to achieve GBM eradication.


Assuntos
Glioblastoma , Glioblastoma/patologia , Humanos , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/patologia , Nicho de Células-Tronco , Microambiente Tumoral
16.
Pathol Res Pract ; 223: 153465, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33989885

RESUMO

We demonstrate that the introduction of GLI1 is sufficient for immortalized human astrocytes to be transformed whereas FOXM1 fails to induce malignant transformation, suggesting differences between GLI1 and FOXM1 in terms of transforming ability despite both transcription factors being overexpressed in malignant gliomas. Moreover, in investigations of mechanisms underlying relatively less-malignant features of GLI1-transformed astrocytes, we found that p27KIP1-positive cells were frequently observed in xenografts derived from GLI1-transformed astrocytes compared to those from RAS-transformed cells. As shRNA-mediated knockdown of p27KIP1 accelerates tumor progression of GLI1-transformed astrocytes, downregulation of p27KIP1 contributes to malignant features of transformed astrocytes. We propose that the models using immortalized/transformed astrocytes are useful to identify the minimal and most crucial set of changes required for glioma formation.


Assuntos
Astrócitos/metabolismo , Neoplasias Encefálicas/genética , Transformação Celular Neoplásica/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Glioma/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteína GLI1 em Dedos de Zinco/genética , Animais , Astrócitos/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Glioma/patologia , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Carga Tumoral , Proteína GLI1 em Dedos de Zinco/metabolismo
17.
Sci Rep ; 11(1): 4220, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33602962

RESUMO

Y-box binding protein 2 (YBX2) has been associated with the properties of both germ cells and cancer cells. We hypothesized that YBX2 might contribute to the characteristics of cancer stem cells (CSCs). In this study, we clarified the function of YBX2 in endometrial cancer stem cells. We established a human YBX2-expressing Ishikawa (IK) cell line (IK-YBX2 cells). We analyzed gene expression associated with stemness and isolated SP cells from IK-YBX2 cells. The SP population of IK-YBX2 cells, the expression of ALDH1 and serial sphere-forming capacity were associated with levels of YBX2 expression. IK-YBX2 cells were resistant to anti-cancer drugs. In gene expression analysis, a gene for cancer testis antigen, CT45, was generally overexpressed in IK-YBX2 cells. YBX2-mediated CT45 expression was associated with increased levels of self-renewal capacity and paclitaxel resistance. The level of CT45 expression was enhanced in high-grade and/or advanced stages of human endometrial cancer tissues. We conclude that expression of YBX2 is essential for the stem cell-like phenotype. CT45 contributes to stemness associated with YBX2 and might be related to the progression of endometrial cancer.


Assuntos
Antígenos de Neoplasias/genética , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias do Endométrio/genética , Células-Tronco Neoplásicas/patologia , Proteínas de Ligação a RNA/genética , Linhagem Celular Tumoral , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Células-Tronco Neoplásicas/efeitos dos fármacos , Paclitaxel/farmacologia
18.
Mol Cancer ; 9: 39, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20167130

RESUMO

BACKGROUND: An increasing number of studies support the presence of stem-like cells in human malignancies. These cells are primarily responsible for tumor initiation and thus considered as a potential target to eradicate tumors. CD133 has been identified as an important cell surface marker to enrich the stem-like population in various human tumors. To reveal the molecular machinery underlying the stem-like features in tumor cells, we analyzed a promoter of CD133 gene using human colon carcinoma Caco-2 and synovial sarcoma Fuji cells, which endogenously express CD133 gene. RESULTS: A reporter analysis revealed that P5 promoter, located far upstream in a human CD133 gene locus, exhibits the highest activity among the five putative promoters (P1 to P5). Deletion and mutation analysis identified two ETS binding sites in the P5 region as being essential for its promoter activity. Electrophoretic mobility shift assays demonstrated the specific binding between nuclear factors and the ETS binding sequence. Overexpression of dominant-negative forms of Ets2 and Elk1 resulted in the significant decrease of P5 activity. Furthermore, treatment of Fuji cells with a specific MEK/ERK inhibitor, U0126, also markedly decreased CD133 expression, but there was no significant effect in Caco-2 cells, suggesting cell type-specific regulation of CD133 expression. Instead, the side population, another hallmark of TSLCs, was dramatically diminished in Caco-2 cells by U0126. Finally, Ras-mediated oncogenic transformation in normal human astrocytes conferred the stem-like capability to form neurosphere-like colonies with the increase of CD133 mRNA expression. CONCLUSIONS: In conclusion, the Ras/ERK pathway at least in part contributes to the maintenance and the acquisition of stem-like hallmarks, although the extent of its contribution is varied in a cell type-specific manner. These findings could help our comprehensive understanding of tumor stemness, and also improve the development of eradicative therapies against human malignancies.


Assuntos
Antígenos CD/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glicoproteínas/genética , Sistema de Sinalização das MAP Quinases , Células-Tronco Neoplásicas/enzimologia , Células-Tronco Neoplásicas/patologia , Peptídeos/genética , Regiões Promotoras Genéticas , Proteínas ras/metabolismo , Antígeno AC133 , Animais , Antígenos CD/metabolismo , Astrócitos/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Transformação Celular Neoplásica/patologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica , Genes Dominantes/genética , Glicoproteínas/metabolismo , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos SCID , Peptídeos/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-ets/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica
19.
Inflamm Regen ; 40: 33, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32952746

RESUMO

Due to the nature of enhanced resistance to conventional chemo/radiotherapies and metastasis, highly tumorigenic cancer stem cells (CSCs) have been proposed as a promising target for cancer eradication. To tackle the therapeutic difficulties of cancers involving CSCs, extensive research efforts have been directed toward understanding the extracellular microenvironments of CSCs, i.e., CSC niche, which plays important roles in CSC maintenance and expansion. Here we review recently identified mechanisms of maintenance and expansion of glioma CSCs (GSCs) leading to glioma progression and recurrence, with particular emphasis on the reports made by studies with a unique approach using polymer microarrays screening and with a unique viewpoint of necrotic particles. The polymer-based approach identified two groups of niche components, extracellular matrices (ECMs) and iron, and uncovered that co-expression of ECM-, iron-, and macrophage-related genes is predictive of glioma patients' outcome. The study in view of a unique fraction of GSC-derived necrotic particles proposed that such particles develop GSC-supportive tumor-associated macrophages (TAMs). Taken together, these studies provide new insights into the mechanisms underlying GSC-driven niche development, i.e., organization of the self-advantageous niche microenvironments for GSC maintenance and expansion leading to glioma progression and recurrence. A series of such studies can redefine the current concept of anti-GSC niche therapy that targets ligands/receptors supporting GSCs, and have potential to accelerate cancer therapy development.

20.
Lab Invest ; 89(6): 645-56, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19333234

RESUMO

SYT-SSX protein, resulted from chromosomal translocation, causes synovial sarcoma, which is a malignant tumor accounting for 10% of soft tissue sarcoma. However, biological functions of SYT (synovial sarcoma translocation), also known as SS18, are largely unclear, whereas it has been proven that Syt-null mice die at early stages of embryonic development. Here, we generated Syt-deficient mice and confirmed the reported phenotypes, including growth retardation, open neural tube and haplo-insufficient lethality, and therefore, there is no doubt that Syt is essential for embryonic development. However, placental defects, described in the earlier report, were rarely seen in our mice and we frequently observed cardiac defect in Syt-deficient mice. As the mechanisms responsible for embryonic lethality seem to be complicate, we performed additional experiments. By using primary cultured embryonic fibroblasts, we showed that Syt(-/-) MEFs deregulate actin organization and suppressed cell migration. These observations suggest that Syt may contribute to the signaling pathway important for various cellular functions in vivo and in vitro, and we propose that Syt-deficient MEFs would be a powerful means to understand the biological roles of SYT in vitro.


Assuntos
Movimento Celular , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Repressoras/fisiologia , Actinas/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Perda do Embrião , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Feminino , Fibroblastos/fisiologia , Cardiopatias Congênitas/genética , Humanos , Camundongos , Camundongos Knockout , Tubo Neural/anormalidades , Placenta/anormalidades , Placenta/metabolismo , Gravidez , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA