Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
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
2.
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
3.
Differentiation ; 115: 53-61, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32891959

RESUMO

Hematopoietic stem cell-containing intra-aortic hematopoietic cell clusters (IAHCs) emerge in the dorsal aorta of the aorta-gonad-mesonephros (AGM) region during midgestation mouse embryos. We previously showed that transduction of Sox17 in CD45lowc-Kithigh cells, which are one component of IAHCs, maintained the cluster formation and the undifferentiated state, but the mechanism of the cluster formation by Sox17 has not been clarified. By microarray gene expression analysis, we found that genes for vascular endothelial-cadherin (VE-cad) and endothelial cell-selective adhesion molecule (ESAM) were expressed at high levels in Sox17-transduced c-Kit+ cells. Here we show the functional role of these adhesion molecules in the formation of IAHCs and the maintenance of the undifferentiated state by in vitro experiments. We detected VE-cad and ESAM expression in endothelial cells of dorsal aorta and IAHCs in E10.5 embryos by whole mount immunohistochemistry. Cells with the middle expression level of VE-cad and the low expression level of ESAM had the highest colony-forming ability. Tamoxifen-dependent nuclear translocation of Sox17-ERT fusion protein induced the formation of cell clusters and the expression of Cdh5 (VE-cad) and ESAM genes. We showed the induction of the Cdh5 (VE-cad) and ESAM expression and the direct interaction of Sox17 with their promoter by luciferase assay and chromatin immunoprecipitation assay, respectively. Moreover, shRNA-mediated knockdown of either Cdh5 (VE-cad) or ESAM gene in Sox17-transduced cells decreased the multilineage-colony forming potential. These findings suggest that VE-cad and ESAM play an important role in the high hematopoietic activity of IAHCs and cluster formation.


Assuntos
Antígenos CD/genética , Caderinas/genética , Moléculas de Adesão Celular/genética , Diferenciação Celular/genética , Proteínas HMGB/genética , Hematopoese/genética , Fatores de Transcrição SOXF/genética , Animais , Aorta/crescimento & desenvolvimento , Aorta/metabolismo , Caderinas/antagonistas & inibidores , Moléculas de Adesão Celular/antagonistas & inibidores , Embrião de Mamíferos , Células Endoteliais/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas HMGB/antagonistas & inibidores , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos , Gravidez , RNA Interferente Pequeno/farmacologia , Fatores de Transcrição SOXF/antagonistas & inibidores
4.
Exp Cell Res ; 365(1): 145-155, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29458175

RESUMO

The aorta-gonad-mesonephros region, from which definitive hematopoiesis first arises in midgestation mouse embryos, has intra-aortic hematopoietic clusters (IAHCs) containing hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). We previously reported expression of the transcription factor Sox17 in IAHCs, and overexpression of Sox17 in CD45lowc-KIThigh cells comprising IAHCs maintains the formation of cell clusters and their multipotency in vitro over multiple passages. Here, we demonstrate the importance of NOTCH1 in IAHC formation and maintenance of the HSC/HPC phenotype. We further show that Notch1 expression is positively regulated by SOX17 via direct binding to its gene promoter. SOX17 and NOTCH1 were both found to be expressed in vivo in cells of IAHCs by whole mount immunostaining. We found that cells transduced with the active form of NOTCH1 or its downstream target, Hes1, maintained their multipotent colony-forming capacity in semisolid medium. Moreover, cells stimulated by NOTCH1 ligand, Jagged1, or Delta-like protein 1, had the capacity to form multilineage colonies. Conversely, knockdown of Notch1 and Hes1 led to a reduction of their multipotent colony-forming capacity. These results suggest that the Sox17-Notch1-Hes1 pathway is critical for maintaining the undifferentiated state of IAHCs.


Assuntos
Aorta/metabolismo , Proteínas HMGB/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Receptor Notch1/metabolismo , Fatores de Transcrição SOXF/metabolismo , Fatores de Transcrição HES-1/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Feto/metabolismo , Gônadas/metabolismo , Mesonefro/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Regiões Promotoras Genéticas/fisiologia
5.
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
6.
Cytokine ; 95: 35-42, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28235674

RESUMO

In the midgestation mouse embryo, hematopoietic cell clusters containing hematopoietic stem/progenitor cells arise in the aorta-gonad-mesonephros (AGM) region. We have previously reported that forced expression of the Sox17 transcription factor in CD45lowc-Kithigh AGM cells, which are the hematopoietic cellular component of the cell clusters, and subsequent coculture with OP9 stromal cells in the presence of three cytokines, stem cell factor (SCF), interleukin-3 (IL-3), and thrombopoietin (TPO), led to the formation and the maintenance of cell clusters with cells at an undifferentiated state in vitro. In this study, we investigated the role of each cytokine in the formation of hematopoietic cell clusters. We cultured Sox17-transduced AGM cells with each of the 7 possible combinations of the three cytokines. The size and the number of Sox17-transduced cell clusters in the presence of TPO, either alone or in combination, were comparable to that observed with the complete set of the three cytokines. Expression of TPO receptor, c-Mpl was almost ubiquitously expressed and maintained in Sox17-transduced hematopoietic cell clusters. In addition, the expression level of c-Mpl was highest in the CD45lowc-Kithigh cells among the Sox17-transduced cell clusters. Moreover, c-Mpl protein was highly expressed in the intra-aortic hematopoietic cell clusters in comparison with endothelial cells of dorsal aorta. Finally, stimulation of the endothelial cells prepared from the AGM region by TPO induced the production of hematopoietic cells. These results suggest that TPO contributes to the formation and the maintenance of hematopoietic cell clusters in the AGM region.


Assuntos
Aorta/citologia , Gônadas/citologia , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Mesonefro/citologia , Trombopoetina/fisiologia , Animais , Aorta/embriologia , Aorta/metabolismo , Células Cultivadas , Gônadas/embriologia , Gônadas/metabolismo , Interleucina-3/fisiologia , Mesonefro/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Receptores de Trombopoetina/metabolismo , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Transdução de Sinais , Fator de Células-Tronco/fisiologia , Transdução Genética
7.
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
8.
Development ; 140(3): 639-48, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23293295

RESUMO

Congenital biliary atresia is an incurable disease of newborn infants, of unknown genetic causes, that results in congenital deformation of the gallbladder and biliary duct system. Here, we show that during mouse organogenesis, insufficient SOX17 expression in the gallbladder and bile duct epithelia results in congenital biliary atresia and subsequent acute 'embryonic hepatitis', leading to perinatal death in ~95% of the Sox17 heterozygote neonates in C57BL/6 (B6) background mice. During gallbladder and bile duct development, Sox17 was expressed at the distal edge of the gallbladder primordium. In the Sox17(+/-) B6 embryos, gallbladder epithelia were hypoplastic, and some were detached from the luminal wall, leading to bile duct stenosis or atresia. The shredding of the gallbladder epithelia is probably caused by cell-autonomous defects in proliferation and maintenance of the Sox17(+/-) gallbladder/bile duct epithelia. Our results suggest that Sox17 plays a dosage-dependent function in the morphogenesis and maturation of gallbladder and bile duct epithelia during the late-organogenic stages, highlighting a novel entry point to the understanding of the etiology and pathogenesis of human congenital biliary atresia.


Assuntos
Atresia Biliar/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas HMGB/metabolismo , Haploinsuficiência , Fatores de Transcrição SOXF/metabolismo , Animais , Animais Recém-Nascidos , Ductos Biliares/metabolismo , Ductos Biliares/patologia , Atresia Biliar/patologia , Proliferação de Células , Colestase/genética , Colestase/patologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Estresse do Retículo Endoplasmático , Epitélio/metabolismo , Epitélio/patologia , Feminino , Vesícula Biliar/metabolismo , Vesícula Biliar/ultraestrutura , Proteínas HMGB/genética , Hepatite Animal/genética , Hepatite Animal/metabolismo , Hepatite Animal/patologia , Hepatócitos/metabolismo , Heterozigoto , Imuno-Histoquímica , Fígado/metabolismo , Fígado/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Gravidez , Fatores de Transcrição SOXF/genética , Fatores de Tempo
9.
Dev Growth Differ ; 56(6): 469-79, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25093513

RESUMO

Sry related high mobility group box 17 (Sox17), which is a marker of endodermal cells and a transcriptional regulator, has a critical role in the maintenance of fetal and neonatal hematopoietic stem cells (HSC). Sox17 has been identified as a key regulator of the development and differentiation of fetal hematopoietic progenitors from the aorta-gonad-mesonephros (AGM) region. The co-culture of Sox17-transduced hematopoietic progenitor cells (CD45(low) c-Kit(high) cells) from AGM regions on OP9 stromal cells gives rise to multipotential hematopoietic stem/progenitor cells. Here, we show that in a primary transplantation experiment, Sox17-transduction in CD45(low) c-Kit(high) cells of embryonic day (E) 10.5 AGM increased the absolute number of common myeloid progenitors (CMPs) in the bone marrow (BM) of recipient mice in comparison to that of granulocyte/macrophage progenitors (GMPs) and the megakaryocyte/erythroid progenitors (MEPs). When Sox17-transduced cells were cultured with OP9 stromal cells, Sox17-transduced GMPs (Sox17-GMPs), Sox17-transduced CMPs (Sox17-CMPs), and Sox17-transduced MEPs (Sox17-MEPs) were generated. Sox17-GMPs and Sox17-CMPs maintained their self-renewal capacity and the hematopoietic ability upon co-culture with the OP9 stromal cells for some passages. Moreover, Sox17-GMPs exhibited the increase in expression of c-Mpl and GATA-2 in comparison to GMPs of BM and Sox17-CMPs showed the increase in expression of c-Mpl, NF-E2, and ß-globin genes in comparison to CMPs of BM. Furthermore, when Sox17-transduced cells were cultured in methylcellulose to examine the colony-forming ability, Sox17-GMPs and Sox17-CMPs maintained the formation of mixed colonies for some passages. Taken together, Sox17 is suggested to regulate the maintenance and differentiation of hematopoietic progenitors derived from AGM regions at midgestation, in particular myeloid progenitors.


Assuntos
Proteínas HMGB/metabolismo , Células Mieloides/citologia , Fatores de Transcrição SOXF/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Técnicas de Cocultura , Proteínas HMGB/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Células Progenitoras de Megacariócitos e Eritrócitos/citologia , Células Progenitoras de Megacariócitos e Eritrócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXF/genética
10.
Elife ; 122024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38529532

RESUMO

Increased levels of lactate, an end-product of glycolysis, have been proposed as a potential surrogate marker for metabolic changes during neuronal excitation. These changes in lactate levels can result in decreased brain pH, which has been implicated in patients with various neuropsychiatric disorders. We previously demonstrated that such alterations are commonly observed in five mouse models of schizophrenia, bipolar disorder, and autism, suggesting a shared endophenotype among these disorders rather than mere artifacts due to medications or agonal state. However, there is still limited research on this phenomenon in animal models, leaving its generality across other disease animal models uncertain. Moreover, the association between changes in brain lactate levels and specific behavioral abnormalities remains unclear. To address these gaps, the International Brain pH Project Consortium investigated brain pH and lactate levels in 109 strains/conditions of 2294 animals with genetic and other experimental manipulations relevant to neuropsychiatric disorders. Systematic analysis revealed that decreased brain pH and increased lactate levels were common features observed in multiple models of depression, epilepsy, Alzheimer's disease, and some additional schizophrenia models. While certain autism models also exhibited decreased pH and increased lactate levels, others showed the opposite pattern, potentially reflecting subpopulations within the autism spectrum. Furthermore, utilizing large-scale behavioral test battery, a multivariate cross-validated prediction analysis demonstrated that poor working memory performance was predominantly associated with increased brain lactate levels. Importantly, this association was confirmed in an independent cohort of animal models. Collectively, these findings suggest that altered brain pH and lactate levels, which could be attributed to dysregulated excitation/inhibition balance, may serve as transdiagnostic endophenotypes of debilitating neuropsychiatric disorders characterized by cognitive impairment, irrespective of their beneficial or detrimental nature.


Assuntos
Disfunção Cognitiva , Endofenótipos , Animais , Camundongos , Humanos , Encéfalo/metabolismo , Disfunção Cognitiva/metabolismo , Modelos Animais de Doenças , Lactatos/metabolismo , Concentração de Íons de Hidrogênio
11.
Exp Cell Res ; 318(6): 705-15, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22289156

RESUMO

Long-term reconstituting hematopoietic stem cells first arise from the aorta of the aorta-gonad-mesonephros (AGM) region in a mouse embryo. We have previously reported that in cultures of the dispersed AGM region, CD45(low)c-Kit(+) cells possess the ability to reconstitute multilineage hematopoietic cells, but investigations are needed to show that this is not a cultured artifact and to clarify when and how this population is present. Based on the expression profile of CD45 and c-Kit in freshly dissociated AGM cells from embryonic day 9.5 (E9.5) to E12.5 and aorta cells in the AGM from E13.5 to E15.5, we defined six cell populations (CD45(-)c-Kit(-), CD45(-)c-Kit(low), CD45(-)c-Kit(high), CD45(low)c-Kit(high), CD45(high)c-Kit(high), and CD45(high)c-Kit(very low)). Among these six populations, CD45(low)c-Kit(high) cells were most able to form hematopoietic cell colonies, but their ability decreased after E11.5 and was undetectable at E13.5 and later. The CD45(low)c-Kit(high) cells showed multipotency in vitro. We demonstrated further enrichment of hematopoietic activity in the Hoechst dye-effluxing side population among the CD45(low)c-Kit(high) cells. Here, we determined that CD45(low)c-Kit(high) cells arise from the lateral plate mesoderm using embryonic stem cell-derived differentiation system. In conclusion, CD45(low)c-Kit(high) cells are the major hematopoietic cells of mouse AGM.


Assuntos
Aorta/citologia , Gônadas/citologia , Células-Tronco Hematopoéticas/citologia , Antígenos Comuns de Leucócito/metabolismo , Mesonefro/citologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Animais , Aorta/embriologia , Aorta/metabolismo , Diferenciação Celular , Gônadas/embriologia , Gônadas/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Mesonefro/embriologia , Camundongos
12.
Inflamm Regen ; 43(1): 41, 2023 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-37553580

RESUMO

BACKGROUND: During mouse embryonic development, definitive hematopoiesis is first detected around embryonic day (E) 10.5 in the aorta-gonad-mesonephros (AGM) region. Hematopoietic stem cells (HSCs) arise in the dorsal aorta's intra-aortic hematopoietic cell clusters (IAHCs). We have previously reported that a transcription factor Sox17 is expressed in IAHCs, and that, among them, CD45lowc-Kithigh cells have high hematopoietic activity. Furthermore, forced expression of Sox17 in this population of cells can maintain the formation of hematopoietic cell clusters. However, how Sox17 does so, particularly downstream signaling involved, remains poorly understood. The purpose of this study is to search for new Sox17 targets which contribute to cluster formation with hematopoietic activity. METHODS: RNA-sequencing (RNA-seq) analysis was done to identify genes that are upregulated in Sox17-expressing IAHCs as compared with Sox17-negative ones. Among the top 7 highly expressed genes, Rasip1 which had been reported to be a vascular-specific regulator was focused on in this study, and firstly, the whole-mount immunostaining was done. We conducted luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay to examine whether Sox17 regulates Rasip1 gene expression via binding to its enhancer element. We also analyzed the cluster formation and the multilineage colony-forming ability of Rasip1-transduced cells and Rasip1-knockdown Sox17-transduced cells. RESULTS: The increase of the Rasip1 expression level was observed in Sox17-positive CD45lowc-Kithigh cells as compared with the Sox17-nonexpressing control. Also, the expression level of the Rasip1 gene was increased by the Sox17-nuclear translocation. Rasip1 was expressed on the membrane of IAHCs, overlapping with the endothelial cell marker, CD31, and hematopoietic stem/progenitor marker (HSPC), c-Kit. Rasip1 expression was observed in most part of c-Kit+Sox17+ cells in IAHCs. Luciferase reporter assay and ChIP assay indicated that one of the five putative Sox17-binding sites in the Rasip1 enhancer region was important for Rasip1 expression via Sox17 binding. Rasip1 knockdown in Sox17-transduced cells decreased the cluster formation and diminished the colony-forming ability, while overexpression of Rasip1 in CD45lowc-Kithigh cells led to a significant but transient increase in hematopoietic activity. CONCLUSIONS: Rasip1 knockdown in Sox17-transduced CD45lowc-Kithigh cells displayed a significant decrease in the multilineage colony-forming ability and the cluster size. Rasip1 overexpression in Sox17-untransduced CD45lowc-Kithigh cells led to a significant but transient increase in the multilineage colony-forming ability, suggesting the presence of a cooperating factor for sustained hematopoietic activity.

13.
Genes Cells ; 15(9): 983-94, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20718937

RESUMO

The discovery of a major hematopoietic stem cell pool in midgestation mouse embryo has defined the placenta as an important hematopoietic anatomical site. In this study, we examined the flow cytometric pattern of mouse placenta cells on embryonic days (E) 10.5 to E15.5, in view of CD45 and c-Kit expression. We also determined which population of these cells shows differentiation potential toward multiple hematopoietic lineages by performing coculture with OP9 stromal cells and colony-forming assay in methylcellulose. Only CD45(+)c-Kit(+) population showed the ability to form hematopoietic colonies including multiple lineages. To distinguish which fraction of placenta cells have the hematopoietic activity, we used GFP transgenic mice in which the fetal part of the placenta is GFP positive and the maternal part is GFP negative. E11.5 and E13.5 CD45(+)c-Kit(+) placental cells that have ability to form hematopoietic colonies are the fetal GFP positive placental cells. E11.5 and E13.5 CD45(+)c-Kit(+) placental cells that have an ability to form hematopoietic colonies mainly reside in Hoechst dye-effluxing side population area (SP). Taken together, in the placenta of mouse embryo, we conclude that SP cells in the CD45(+)c-Kit(+) fetal placental cells have the ability to form hematopoietic colonies.


Assuntos
Células-Tronco Hematopoéticas/citologia , Antígenos Comuns de Leucócito/metabolismo , Placenta/citologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Animais , Antígenos CD34/metabolismo , Linhagem Celular , Separação Celular , Células Cultivadas , Técnicas de Cocultura , Ensaio de Unidades Formadoras de Colônias , Feminino , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Placenta/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Gravidez , Células Estromais/citologia , Células Estromais/metabolismo , Fatores de Tempo
14.
Dev Growth Differ ; 53(7): 870-7, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21883169

RESUMO

During murine embryonic development, primitive hematopoiesis occurs in the yolk sac (YS). Recent studies have shown that the YS also harbors definitive hematopoietic activity. However, the population of YS cells contributing to definitive hematopoiesis has not been identified. In this study, we characterized the hematopoietic cell populations in the YS of mouse embryos from E9.5 to E14.5 in view of the expression profiles of CD45 and c-Kit. The YS cells from E9.5 to E11.5 could be divided into six populations: CD45(-) c-Kit(-) , CD45(-) c-Kit(low) , CD45(-) c-Kit(high) , CD45(low) c-Kit(high) , CD45(high) c-Kit(high) and CD45(high) c-Kit(very low) . Among these populations, CD45(low) c-Kit(high) cells showed the highest multilineage hematopoietic colony-forming activity. Later in development, the YS cells from E12.5 to E14.5 lost the second and fourth populations (i.e., they retained CD45(-) c-Kit(-) , CD45(-) c-Kit(high) , CD45(high) c-Kit(high) and CD45(high) c-Kit(very low) cells), and concurrently with the disappearance of the CD45(low) c-Kit(high) population, no significant hematopoietic activity was found in any of the populations on and after E12.5. CD45(low) c-Kit(high) YS cells, which had a round morphology with a large nucleus, possessed the ability to differentiate into myeloid and B lymphoid cells when cultured with stromal cells. These findings suggest that CD45(low) c-Kit(high) YS cells include more undifferentiated cells than the other YS cell populations and possess in vitro potency to differentiate into multilineage hematopoietic cells. Furthermore, this cell population disappears from the YS at around E12.5, when the site of hematopoiesis has already shifted to the fetal liver and the placenta.


Assuntos
Embrião de Mamíferos/citologia , Hematopoese , Antígenos Comuns de Leucócito/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Saco Vitelino/citologia , Animais , Linfócitos B/citologia , Biomarcadores , Técnicas de Cultura de Células , Diferenciação Celular , Separação Celular , Técnicas de Cocultura , Ensaio de Unidades Formadoras de Colônias , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Embrião de Mamíferos/metabolismo , Feminino , Citometria de Fluxo , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Monócitos/citologia , Células Estromais/citologia , Células Estromais/metabolismo , Saco Vitelino/metabolismo
15.
J Exp Med ; 199(5): 737-42, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14981116

RESUMO

In midgestation mouse embryos, the aorta-gonad-mesonephros (AGM) region generates hematopoietic stem cells and definitive hematopoiesis is regulated by cell-cell interaction and signaling molecules. We showed that a Ras/mitogen-activated protein (MAP) kinase signaling-specific inhibitor and a dominant negative mutant Ras blocked the production of CD45+ hematopoietic cells in embryonic day 11.5 AGM culture, indicating an essential role for the MAP kinase pathway in AGM hematopoiesis. Overexpression of the Ras/MAP kinase pathway regulator, Spred-2, in the AGM culture significantly reduced the number of CD45+ cells. In contrast, production of CD45+ cells from the AGM region of Spred-2-null mice was up-regulated as compared with wild-type littermates. Furthermore, Spred-2-deficient mice exhibited elevated hematopoietic colony formation from vascular endothelial-cadherin+ cells. These data indicate that Spred-2 functions as a negative regulator of AGM hematopoiesis by inhibiting hematopoietic cytokine signaling.


Assuntos
Hematopoese/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Repressoras/fisiologia , Animais , Aorta/embriologia , Aorta/enzimologia , Sequência de Bases , Citocinas/metabolismo , Primers do DNA/genética , Ativação Enzimática , Feminino , Idade Gestacional , Gônadas/embriologia , Gônadas/enzimologia , Sistema de Sinalização das MAP Quinases , Masculino , Mesonefro/embriologia , Mesonefro/enzimologia , Camundongos , Camundongos Knockout , Gravidez , Proteínas Repressoras/genética
16.
J Cancer Prev ; 25(4): 204-212, 2020 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-33409253

RESUMO

A transcription factor Sry-related high mobility group box (Sox) 17 is involved in developmental processes including spermatogenesis, cardiovascular system, endoderm formation, and so on. In this article, we firstly review the studies on the relation between the Sox17 expression and tumor malignancy. Although Sox17 positively promotes various tissue development, most of the cancers associated with Sox17 show decreased expression levels of Sox17, and an inverse correlation between Sox17 expression and malignancy is revealed. We briefly discuss the mechanism of such Sox17 down-regulation by focusing on DNA methylation of CpG sites located in the Sox17 gene promoter. Next, we overview the function of Sox17 in the fetal hematopoiesis, particularly in the dorsal aorta in midgestation mouse embryos. The Sox17 expression in hematopoietic stem cell (HSC)-containing intra-aortic hematopoietic cell cluster (IAHCs) is important for the cluster formation with the hematopoietic ability. The sustained expression of Sox17 in adult bone marrow HSCs and the cells in IAHCs of the dorsal aorta indicate abnormalities that are low lymphocyte chimerism and the aberrant proliferation of common myeloid progenitors in transplantation experiments. We then summarize the perspectives of Sox17 research in cancer control.

17.
Mol Cell Biol ; 23(23): 8486-94, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14612394

RESUMO

Development of hematopoietic cells in the aorta-gonad-mesonephros (AGM) region in the midgestation mouse embryo involves a multistep process, sequentially changing from endothelial cell-like cells, including hemangioblasts, into hematopoietic stem cells, progenitors, and/or lineage-committed cells. An adaptor molecule, Lnk, is known to negatively control the production of pro- and pre-B cells and hematopoietic progenitor cells in adult bone marrow. Here we show a role of Lnk in hematopoietic development in the AGM region. Lnk was predominantly expressed in the endothelial cells lining the dorsal aorta at embryonic day 11.5 (E11.5). Overexpression of Lnk in the primary culture of the AGM region at E11.5 suppressed the emergence of CD45+ hematopoietic cells. Point mutation in the SH2 domain of Lnk, which abolishes the binding capability of Lnk to c-Kit upon stimulation with stem cell factor (SCF), led to loss of Lnk-dependent inhibition of hematopoietic cell development in AGM cultures, suggesting Lnk-mediated inhibition of the SCF/c-Kit signaling pathway. In cultured AGM cells from Lnk homozygous mutant mouse embryos, the number of emerged CD45+ cells was 2.5-fold larger than that from heterozygous littermates. Furthermore, aorta cells of E11.5 Lnk homozygous mutant mice also showed enhanced hematopoietic colony-forming activity. Thus, Lnk is a negative regulator of hematopoiesis in the AGM region.


Assuntos
Hematopoese/fisiologia , Proteínas/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Aorta/embriologia , Sequência de Bases , Técnicas de Cultura , DNA/genética , Fator 2 de Crescimento de Fibroblastos/fisiologia , Expressão Gênica , Gônadas/embriologia , Heterozigoto , Homozigoto , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Mesonefro/embriologia , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Mutação Puntual , Proteínas/química , Proteínas/genética , Transdução de Sinais , Fator de Células-Tronco/fisiologia , Domínios de Homologia de src
18.
Biochem J ; 396(1): 183-92, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16460309

RESUMO

Activation of the superoxide-producing phagocyte NADPH oxidase, crucial for host defence, requires an SH3 (Src homology 3)-domain-mediated interaction of the regulatory protein p47phox with p22phox, a subunit of the oxidase catalytic core flavocytochrome b558. Although previous analysis of a crystal structure has demonstrated that the tandem SH3 domains of p47phox sandwich a short PRR (proline-rich region) of p22phox (amino acids 151-160), containing a polyproline II helix, it has remained unknown whether this model is indeed functional in activation of the oxidase. In the present paper we show that the co-operativity between the two SH3 domains of p47phox, as expected from the model, is required for oxidase activation. Deletion of the linker between the p47phox SH3 domains results not only in a defective binding to p22phox but also in a loss of the activity to support superoxide production. The present analysis using alanine-scanning mutagenesis identifies Pro152, Pro156 and Arg158 in the p22phox PRR as residues indispensable for the interaction with p47phox. Pro152 and Pro156 are recognized by the N-terminal SH3 domain, whereas Arg158 contacts with the C-terminal SH3 domain. Amino acid substitution for any of the three residues in the p22phox PRR abrogates the superoxide-producing activity of the oxidase reconstituted in intact cells. The bis-SH3-mediated interaction of p47phox with p22phox thus functions to activate the phagocyte oxidase. Furthermore, we provide evidence that a region C-terminal to the PRR of p22phox (amino acids 161-164), adopting an a-helical conformation, participates in full activation of the phagocyte oxidase by fortifying the association with the p47phox SH3 domains.


Assuntos
NADPH Oxidases/fisiologia , Domínios de Homologia de src , Substituição de Aminoácidos , Animais , Células CHO , Cricetinae , Cricetulus , Ativação Enzimática , Humanos , Mutagênese Sítio-Dirigida , NADPH Oxidases/química , Fagócitos/enzimologia , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/fisiologia , Relação Estrutura-Atividade , Superóxidos/metabolismo , Transfecção
19.
Neurosci Res ; 53(2): 176-82, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16112216

RESUMO

Mammalian cells that have been committed to a certain cell lineage cannot be directed to other lineages. However, some astrocytes in the mammalian brains have been reported to represent plasticity to redirect to other cell lineages. We found that mouse hippocampal astrocytes cultured in aggregate forms of "astrosphere", redirected to MAP2-positive immature neurons. In astrospheres, basic HLH factors positively regulating neuronal differentiation were up-regulated and Id3 inhibiting basic HLH factors was down-regulated. Ectopic Id3 induction repressed redirection of astrocytes to a neuronal lineage, suggesting that astrosphere formation induced plasticity of astrocytes by changing the gene expression patterns.


Assuntos
Astrócitos/citologia , Técnicas de Cultura de Células , Diferenciação Celular , Hipocampo/citologia , Neurônios/citologia , Animais , Agregação Celular , Técnicas de Cultura de Células/métodos , Linhagem da Célula , Células Cultivadas , Imuno-Histoquímica , Proteínas Inibidoras de Diferenciação , Camundongos , Fator de Crescimento Neural/metabolismo , Proteínas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Exp Hematol ; 31(4): 283-9, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12691915

RESUMO

OBJECTIVE: Definitive hematopoiesis starts in the aorta-gonad-mesonephros (AGM) region during mouse development and remarkably expands in the liver at a later stage of ontogeny. gp130 is a signal transducing receptor component shared by all the IL-6 family cytokines, whose gene ablation in mouse results in the significant reduction in the fetal liver hematopoiesis. The present study aims to evaluate the role of gp130 signaling in the fetal mouse AGM hematopoiesis. METHODS AND MATERIALS: Mouse AGM regions from the wild-type and gp130-deficient mice on embryonic day 11.5 were dissociated and cultured with a mixture of cytokines, including one which activates gp130. Wild-type human gp130 and its mutant constructs were introduced into cultured gp130-deficient AGM cells using retrovirus system. To further analyze gp130 downstream signaling, a dominant-negative mutant of STAT3 was also introduced. RESULTS: The gp130 deficiency in the culture of fetal mouse AGM cells resulted in the failure of the expansion of the c-kit(+), Sca-1(+), and lineage markers(-) population. Such failure was rescued by introduction of a wild-type gp130 expression construct but not its mutant constructs having no ability to activate STAT3. In the normal AGM cell culture, introduction of a dominant-negative form of STAT3 in which Y(705) was changed to phenylalanine suppressed the expansion of hematopoietic cell colonies. CONCLUSION: gp130 plays an indispensable role in the expansion of hematopoietic precursor cells in the fetal mouse AGM. In particular, the activation of STAT3 by gp130 is found to be important in this process.


Assuntos
Antígenos CD/fisiologia , Aorta/embriologia , Gônadas/embriologia , Hematopoese , Glicoproteínas de Membrana/fisiologia , Mesonefro/embriologia , Transdução de Sinais , Animais , Antígenos CD/genética , Aorta/citologia , Contagem de Células , Divisão Celular , Células Cultivadas , Receptor gp130 de Citocina , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Expressão Gênica , Regulação da Expressão Gênica , Genes bcl-1/genética , Genes myb/genética , Gônadas/citologia , Proteínas de Fluorescência Verde , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/imunologia , Antígenos Comuns de Leucócito/análise , Proteínas Luminescentes/genética , Glicoproteínas de Membrana/genética , Mesonefro/citologia , Camundongos , Camundongos Endogâmicos ICR , Mutação , Oncostatina M , Peptídeos/farmacologia , Fosforilação , Fator de Transcrição STAT3 , Fator de Células-Tronco/farmacologia , Transativadores/genética , Transativadores/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA