Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
PLoS One ; 19(4): e0302130, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38625917

RESUMO

PARP inhibitors have been developed as anti-cancer agents based on synthetic lethality in homologous recombination deficient cancer cells. However, resistance to PARP inhibitors such as olaparib remains a problem in clinical use, and the mechanisms of resistance are not fully understood. To investigate mechanisms of PARP inhibitor resistance, we established a BRCA1 knockout clone derived from the pancreatic cancer MIA PaCa-2 cells, which we termed C1 cells, and subsequently isolated an olaparib-resistant C1/OLA cells. We then performed RNA-sequencing and pathway analysis on olaparib-treated C1 and C1/OLA cells. Our results revealed activation of cell signaling pathway related to NAD+ metabolism in the olaparib-resistant C1/OLA cells, with increased expression of genes encoding the NAD+ biosynthetic enzymes NAMPT and NMNAT2. Moreover, intracellular NAD+ levels were significantly higher in C1/OLA cells than in the non-olaparib-resistant C1 cells. Upregulation of intracellular NAD+ levels by the addition of nicotinamide also induced resistance to olaparib and talazoparib in C1 cells. Taken together, our findings suggest that upregulation of intracellular NAD+ is one of the factors underlying the acquisition of PARP inhibitor resistance.


Assuntos
Antineoplásicos , Neoplasias Pancreáticas , Piperazinas , Humanos , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , NAD , Linhagem Celular Tumoral , Antineoplásicos/farmacologia , Ftalazinas/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Proteína BRCA1
2.
Biochem Biophys Res Commun ; 692: 149309, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38048727

RESUMO

Poly (ADP-ribose) glycohydrolase (PARG) is an enzyme that mainly degrades poly (ADP-ribose) (PAR) synthesized by poly (ADP-ribose) polymerase (PARP) family proteins. Although PARG is involved in many biological phenomena, including DNA repair, cell differentiation, and cell death, little is known about the relationship between osteoclast differentiation and PARG. It has also not been clarified whether PARG is a valuable target for therapeutic agents in the excessive activity of osteoclast-related bone diseases such as osteoporosis. In the present study, we examined the effects of PARG inhibitor PDD00017273 on osteoclast differentiation in RANKL-induced RAW264 cells. PDD00017273 induced the accumulation of intracellular PAR and suppressed the number of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells. PDD00017273 also downregulated osteoclast differentiation marker genes such as Trap, cathepsin K (Ctsk), and dendrocyte expressed seven transmembrane protein (Dcstamp) and protein expression of nuclear factor of activated T cells 1 (NFATc1), a master regulator of osteoclast differentiation. Taken together, our findings suggest that dysfunction of PARG suppresses osteoclast differentiation via the PAR accumulation and partial inactivation of the NFATc1.


Assuntos
Osteoclastos , Ribose , Glicosídeo Hidrolases/metabolismo , Osteoclastos/metabolismo , Poli(ADP-Ribose) Polimerase-1 , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Camundongos
3.
Stem Cells Dev ; 32(21-22): 670-680, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37639359

RESUMO

The parathyroid gland plays an essential role in mineral and bone metabolism. Cultivation of physiological human parathyroid cells has yet to be established and the method by which parathyroid cells differentiate from pluripotent stem cells remains uncertain. Therefore, it has been hard to clarify the mechanisms underlying the onset of parathyroid disorders, such as hyperparathyroidism. In this study, we developed a new method of parathyroid cell differentiation from human induced pluripotent stem (iPS) cells. Parathyroid cell differentiation occurred in accordance with embryologic development. Differentiated cells, which expressed the parathyroid hormone, adopted unique cell aggregation similar to the parathyroid gland. In addition, these differentiated cells were identified as calcium-sensing receptor (CaSR)/epithelial cell adhesion molecule (EpCAM) double-positive cells. Interestingly, stimulation with transforming growth factor-α (TGF-α), which is considered a causative molecule of parathyroid hyperplasia, increased the CaSR/EpCAM double-positive cells, but this effect was suppressed by erlotinib, which is an epidermal growth factor receptor (EGFR) inhibitor. These results suggest that TGF-α/EGFR signaling promotes parathyroid cell differentiation from iPS cells in a similar manner to parathyroid hyperplasia.


Assuntos
Células-Tronco Pluripotentes Induzidas , Glândulas Paratireoides , Humanos , Glândulas Paratireoides/metabolismo , Glândulas Paratireoides/patologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Hiperplasia/metabolismo , Hiperplasia/patologia , Fator de Crescimento Transformador alfa/farmacologia , Fator de Crescimento Transformador alfa/metabolismo , Molécula de Adesão da Célula Epitelial/metabolismo , Molécula de Adesão da Célula Epitelial/farmacologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Diferenciação Celular , Receptores de Detecção de Cálcio/genética , Receptores de Detecção de Cálcio/metabolismo
4.
Int J Mol Sci ; 23(9)2022 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-35563432

RESUMO

Poly ADP-ribosylation (PARylation) is a post-translational modification catalyzed by poly (ADP-ribose) polymerase (PARP) family proteins such as PARP1. Although PARylation regulates important biological phenomena such as DNA repair, chromatin regulation, and cell death, little is known about the relationship between osteoblast differentiation and the PARylation cycle involving PARP1 and the poly (ADP-ribose)-degrading enzyme poly (ADP-ribose) glycohydrolase (PARG). Here, we examined the effects of PARP inhibitor olaparib, an approved anti-cancer agent, and PARG inhibitor PDD00017273 on osteoblast differentiation. Olaparib decreased alkaline phosphatase (ALP) activity and suppressed mineralized nodule formation evaluated by Alizarin Red S staining in preosteoblastic MC3T3-E1 cells, while PDD00017273 promoted ALP activity and mineralization. Furthermore, PDD00017273 up-regulated the mRNA expression levels of osteocalcin and bone sialoprotein, as osteoblast differentiation markers, and osterix as transcription inducers for osteoblast differentiation, whereas olaparib down-regulated the expression of these genes. These findings suggest that PARG inhibition by PDD00017273 accelerates osteoblast differentiation in MC3T3-E1 cells. Thus, PARG inhibitor administration could provide therapeutic benefits for metabolic bone diseases such as osteoporosis.


Assuntos
Poli(ADP-Ribose) Polimerases , Ribose , Difosfato de Adenosina , Glicosídeo Hidrolases/metabolismo , Osteoblastos/metabolismo , Poli(ADP-Ribose) Polimerase-1/genética , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo
6.
Oncoimmunology ; 9(1): 1814620, 2020 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-33457097

RESUMO

Immunotherapy using dendritic cells (DCs) is a promising treatment modality for cancer. However, the limited number of functional DCs from peripheral blood has been linked to the unsatisfactory clinical efficacies of current DC-based cancer immunotherapies. We previously generated proliferating antigen-presenting cells (APCs) by genetically engineering myeloid cells derived from induced pluripotent stem cells (iPSC-pMCs), which offer infinite functional APCs for broad applications in cancer therapy. Herein, we aimed to further enhance the antitumor effect of these cells by genetic modification. GM-CSF gene transfer did not affect the morphology, or surface phenotype of the original iPSC-pMCs, however, it did impart good viability to iPSC-pMCs. The resultant cells induced GM-CSF-dependent CD8+ T cell homeostatic proliferation, thereby enhancing antigen-specific T cell priming in vitro. Administration of the tumor antigen-loaded GM-CSF-producing iPSC-pMCs (GM-pMCs) efficiently stimulated antigen-specific T cells and promoted effector cell infiltration of the tumor tissues, leading to an augmented antitumor effect. To address the potential tumorigenicity of iPSC-derived products, irradiation was applied and found to restrict the proliferation of GM-pMCs, while retaining their T cell-stimulatory capacity. Furthermore, the irradiated cells exerted an antitumor effect equivalent to that of bone marrow-derived DCs obtained from immunocompetent mice. Additionally, combination with immune checkpoint inhibitors increased the infiltration of CD8+ or NK1.1+ effector cells and decreased CD11b+/Gr-1+ cells without causing adverse effects. Hence, although GM-pMCs have certain characteristics that differ from endogenous DCs, our findings suggest the applicability of these cells for broad clinical use and will provide an unlimited source of APCs with uniform quality.


Assuntos
Células Dendríticas , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Animais , Antígenos de Neoplasias/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Ativação Linfocitária , Camundongos , Linfócitos T Citotóxicos
7.
Nat Commun ; 9(1): 2202, 2018 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-29875383

RESUMO

We previously identified CD34-negative (CD34-) severe combined immunodeficiency (SCID)-repopulating cells as primitive hematopoietic stem cells (HSCs) in human cord blood. In this study, we develop a prospective ultra-high-resolution purification method by applying two positive markers, CD133 and GPI-80. Using this method, we succeed in purifying single long-term repopulating CD34- HSCs with self-renewing capability residing at the apex of the human HSC hierarchy from cord blood, as evidenced by a single-cell-initiated serial transplantation analysis. The gene expression profiles of individual CD34+ and CD34- HSCs and a global gene expression analysis demonstrate the unique molecular signature of CD34- HSCs. We find that the purified CD34- HSCs show a potent megakaryocyte/erythrocyte differentiation potential in vitro and in vivo. Megakaryocyte/erythrocyte progenitors may thus be generated directly via a bypass route from the CD34- HSCs. Based on these data, we propose a revised road map for the commitment of human CD34- HSCs in cord blood.


Assuntos
Autorrenovação Celular , Sangue Fetal/citologia , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Animais , Antígenos CD34/metabolismo , Diferenciação Celular/genética , Linhagem da Célula/genética , Separação Celular/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Estudos Prospectivos , Análise de Célula Única/métodos
8.
Stem Cell Reports ; 10(3): 920-932, 2018 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-29478902

RESUMO

It is generally thought that the proliferative capacity and differentiation potential of somatic stem cells, including mesenchymal stromal/stem cells (MSCs) and hematopoietic stem cells, decline with age. We investigated the effects of aging on human bone-derived MSCs expressing CD271 and SSEA-4 (double-positive MSCs [DPMSCs]). The percentages of DPMSCs in bone tissue decreased significantly with age. The DPMSCs from elderly patients (old DPMSCs) showed cellular senescence, which was evidenced by low growth potential, high senescence-associated ß-galactosidase activity, and elevated p16 and p21 CDK inhibitor levels. Moreover, old DPMSCs showed weak osteogenic differentiation potential and less hematopoiesis-supporting activity in comparison with young DPMSCs. Interestingly, the addition of transforming growth factor ß2 (TGF-ß2) induced cellular senescence in young DPMSCs. With the exception of the adipogenic differentiation potential, all of the aging phenomena observed in old DPMSCs were reversed by the addition of anti-TGF-ß antibodies. These results suggest that, in part, old DPMSCs accelerate cellular senescence through TGF-ß signaling.


Assuntos
Senescência Celular/fisiologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Antígenos Embrionários Estágio-Específicos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Adipogenia/fisiologia , Adulto , Idoso de 80 Anos ou mais , Envelhecimento/metabolismo , Envelhecimento/fisiologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Feminino , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Humanos , Masculino , Osteogênese/fisiologia , Transdução de Sinais/fisiologia , beta-Galactosidase/metabolismo
9.
Cell Transplant ; 26(6): 1043-1058, 2017 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-27938494

RESUMO

In the murine hematopoietic stem cell (HSC) compartment, thrombopoietin (THPO)/MPL (THPO receptor) signaling plays an important role in the maintenance of adult quiescent HSCs. However, the role of THPO/MPL signaling in the human primitive HSC compartment has not yet been elucidated. We have identified very primitive human cord blood (CB)-derived CD34- severe combined immunodeficiency (SCID)-repopulating cells (SRCs) using the intra-bone marrow injection method. In this study, we investigated the roles of the MPL expression in the human primitive HSC compartment. The SRC activities of the highly purified CB-derived 18Lin-CD34+/-MPL+/- cells were analyzed using NOG mice. In the primary recipient mice, nearly all mice that received CD34+/-MPL+/- cells were repopulated with human CD45+ cells. Nearly all of these mice that received CD34+MPL+/- and CD34-MPL- cells showed a secondary repopulation. Interestingly, the secondary recipient mice that received CD34+/-MPL- cells showed a distinct tertiary repopulation. These results clearly indicate that the CD34+/- SRCs not expressing MPL sustain a long-term (LT) (>1 year) human cell repopulation in NOG mice. Moreover, CD34- SRCs generate CD34+CD38-CD90+ SRCs in vitro and in vivo. These findings provide a new concept that CD34-MPL- SRCs reside at the apex of the human HSC hierarchy.


Assuntos
Antígenos CD34/metabolismo , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Receptores de Trombopoetina/metabolismo , Animais , Antígenos CD34/genética , Células Cultivadas , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Feminino , Humanos , Camundongos , Camundongos SCID , Receptores de Trombopoetina/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
11.
Stem Cells Dev ; 25(1): 27-42, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26595762

RESUMO

Murine bone marrow (BM)-derived very small embryonic-like stem cells (BM VSELs), defined by a lineage-negative (Lin(-)), CD45-negative (CD45(-)), Sca-1-positive (Sca-1(+)) immunophenotype, were previously reported as postnatal pluripotent stem cells (SCs). We developed a highly efficient method for isolating Lin(-)CD45(-)Sca-1(+) small cells using enzymatic treatment of murine bone. We designated these cells as bone-derived VSELs (BD VSELs). The incidences of BM VSELs in the BM-derived nucleated cells and that of BD VSELs in bone-derived nucleated cells were 0.002% and 0.15%, respectively. These BD VSELs expressed a variety of hematopoietic stem cell (HSC), mesenchymal stem cell (MSC), and endothelial cell markers. The gene expression profile of the BD VSELs was clearly distinct from those of HSCs, MSCs, and ES cells. In the steady state, the BD VSELs proliferated slowly, however, the number of BD VSELs significantly increased in the bone after acute liver injury. Moreover, green fluorescent protein-mouse derived BD VSELs transplanted via tail vein injection after acute liver injury were detected in the liver parenchyma of recipient mice. Immunohistological analyses suggested that these BD VSELs might transdifferentiate into hepatocytes. This study demonstrated that the majority of the Lin(-)CD45(-)Sca-1(+) VSEL phenotypic cells reside in the bone rather than the BM. However, the immunophenotype and the gene expression profile of BD VSELs were clearly different from those of other types of SCs, including BM VSELs, MSCs, HSCs, and ES cells. Further studies will therefore be required to elucidate their cellular and/or SC characteristics and the potential relationship between BD VSELs and BM VSELs.


Assuntos
Antígenos Ly/metabolismo , Osso e Ossos/citologia , Linhagem da Célula , Separação Celular/métodos , Células-Tronco Embrionárias/citologia , Antígenos Comuns de Leucócito/metabolismo , Proteínas de Membrana/metabolismo , Envelhecimento/patologia , Animais , Osso e Ossos/metabolismo , Tamanho Celular , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo
12.
Cancer Immunol Res ; 3(6): 668-77, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25672396

RESUMO

The use of dendritic cells (DC) to prime tumor-associated antigen-specific T-cell responses provides a promising approach to cancer immunotherapy. Embryonic stem cells (ESC) and induced pluripotent stem cells (iPSC) can differentiate into functional DCs, thus providing an unlimited source of DCs. However, the previously established methods of generating practical volumes of DCs from pluripotent stem cells (PSC) require a large number of PSCs at the start of the differentiation culture. In this study, we generated mouse proliferating myeloid cells (pMC) as a source of antigen-presenting cells (APC) using lentivirus-mediated transduction of the c-Myc gene into mouse PSC-derived myeloid cells. The pMCs could propagate almost indefinitely in a cytokine-dependent manner, while retaining their potential to differentiate into functional APCs. After treatment with IL4 plus GM-CSF, the pMCs showed impaired proliferation and differentiated into immature DC-like cells (pMC-DC) expressing low levels of major histocompatibility complex (MHC)-I, MHC-II, CD40, CD80, and CD86. In addition, exposure to maturation stimuli induced the production of TNFα and IL12p70, and enhanced the expression of MHC-II, CD40, and CD86, which is thus suggestive of typical DC maturation. Similar to bone marrow-derived DCs, they stimulated a primary mixed lymphocyte reaction. Furthermore, the in vivo transfer of pMC-DCs pulsed with H-2K(b)-restricted OVA257-264 peptide primed OVA-specific cytotoxic T cells and elicited protection in mice against challenge with OVA-expressing melanoma. Overall, myeloid cells exhibiting cytokine-dependent proliferation and DC-like differentiation may be used to address issues associated with the preparation of DCs.


Assuntos
Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Diferenciação Celular , Células Mieloides/citologia , Células Mieloides/imunologia , Células-Tronco Pluripotentes/citologia , Transferência Adotiva , Animais , Apresentação de Antígeno , Células Apresentadoras de Antígenos/metabolismo , Antígenos de Neoplasias/imunologia , Antígenos de Superfície/metabolismo , Proliferação de Células , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Imunofenotipagem , Melanoma/imunologia , Melanoma/patologia , Melanoma/terapia , Camundongos , Células Mieloides/metabolismo , Neoplasias/imunologia , Peptídeos/imunologia , Fenótipo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo
13.
Cell Transplant ; 24(1): 97-113, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24172908

RESUMO

It is well documented that specialized mesenchymal stem/stromal cells (MSCs) constitute the hematopoietic stem cell (HSC) niche in the bone marrow (BM), and these MSCs support/maintain the HSCs in an undifferentiated state. A number of studies have demonstrated that BM-derived MSCs (BM-MSCs) can support HSCs in vitro. However, it remains unclear whether nonhematopoietic tissue-derived MSC-like cells, such as dental pulp stem cells (DPSCs), have the ability to support HSCs. In this study, we prospectively isolated DPSCs from mouse mandibular incisors by fluorescence-activated cell sorting (FACS) using BM-MSC markers, such as PDGFRα and Sca-1. The PDGFRα and Sca-1 double-positive DPSCs and BM-MSCs showed similar morphologies and expression patterns of MSC markers. The ability of the DPSCs to support hematopoietic stem/progenitor cells (HSPCs) was then analyzed by an in vitro coculture system. Moreover, their HSC-supporting activity was evaluated by in vivo xenotransplantation assays using NOD/Shi-scid/IL-2Rγc(null) (NOG) mice. Interestingly, the DPSCs supported human cord blood (CB)-derived CD34-positive (CD34(+)), as well as CD34-negative (CD34(-)), HSCs. The supporting activities of DPSCs for human CB-derived CD34(+) and CD34(-) HSCs were comparable to those of BM-MSCs. The results of the present study demonstrated, for the first time, that prospectively isolated murine PDGFRα and Sca-1 double-positive DPSCs could support primitive human CD34(+) and CD34(-) HSCs in vitro.


Assuntos
Polpa Dentária/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Animais , Antígenos de Diferenciação/metabolismo , Células Cultivadas , Técnicas de Cocultura , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Polpa Dentária/citologia , Feminino , Sangue Fetal , Células-Tronco Hematopoéticas/citologia , Xenoenxertos , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Nus , Camundongos SCID
14.
Stem Cells ; 33(5): 1554-65, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25537923

RESUMO

Hematopoietic stem cells (HSCs) are maintained in a specialized bone marrow (BM) niche, which consists of osteoblasts, endothelial cells, and a variety of mesenchymal stem/stromal cells (MSCs). However, precisely what types of MSCs support human HSCs in the BM remain to be elucidated because of their heterogeneity. In this study, we succeeded in prospectively isolating/establishing three types of MSCs from human BM-derived lineage- and CD45-negative cells, according to their cell surface expression of CD271 and stage-specific embryonic antigen (SSEA)-4. Among them, the MSCs established from the Lineage(-) CD45(-) CD271(+) SSEA-4(+) fraction (DP MSC) could differentiate into osteoblasts and chondrocytes, but they lacked adipogenic differentiation potential. The DP MSCs expressed significantly higher levels of well-characterized HSC-supportive genes, including IGF-2, Wnt3a, Jagged1, TGFß3, nestin, CXCL12, and Foxc1, compared with other MSCs. Interestingly, these osteo-chondrogenic DP MSCs possessed the ability to support cord blood-derived primitive human CD34-negative severe combined immunodeficiency-repopulating cells. The HSC-supportive actions of DP MSCs were partially carried out by soluble factors, including IGF-2, Wnt3a, and Jagged1. Moreover, contact between DP MSCs and CD34-positive (CD34(+) ) as well as CD34-negative (CD34(-) ) HSCs was important for the support/maintenance of the CD34(+/-) HSCs in vitro. These data suggest that DP MSCs might play an important role in the maintenance of human primitive HSCs in the BM niche. Therefore, the establishment of DP MSCs provides a new tool for the elucidation of the human HSC/niche interaction in vitro as well as in vivo.


Assuntos
Antígenos CD34/metabolismo , Células da Medula Óssea/citologia , Separação Celular/métodos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Mesenquimais/citologia , Adapaleno/metabolismo , Adipogenia/genética , Animais , Biomarcadores/metabolismo , Células da Medula Óssea/metabolismo , Proliferação de Células , Condrogênese/genética , Ensaio de Unidades Formadoras de Colônias , Feminino , Sangue Fetal/citologia , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos NOD , Camundongos SCID , Osteogênese/genética , Antígenos Embrionários Estágio-Específicos/metabolismo
15.
Stem Cells ; 29(11): 1783-91, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21898688

RESUMO

Although c-kit is expressed highly on murine hematopoietic stem cells (HSCs) and essential for bone marrow (BM) hematopoiesis, the significance of the high level of expression of c-kit on HSCs was not well determined. We show here that CD150(+) CD48(-) Lineage(-) Sca-1(+) c-kit(+) HSCs in adult BM are distributed within the range of roughly a 20-fold difference in the expression level of c-kit, and that c-kit density correlates with the cycling status of the HSC population. This predisposition is more evident in the BM of mice older than 30 weeks. The HSCs in G(0) phase express a lower level of c-kit both on the cell surface and inside the cells, which cannot be explained by ligand receptor binding and internalization. It is more likely that the low level of c-kit expression is a unique property of HSCs in G(0). Despite functional differences in the c-kit gradient, the HSCs are uniformly hypoxic and accessible to blood perfusion. Therefore, our data indicate the possibility that the hypoxic state of the HSCs is actively regulated, rather than them being passively hypoxic through a simple anatomical isolation from the circulation.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Animais , Antígenos CD/metabolismo , Benzimidazóis/farmacologia , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Antígeno CD48 , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Células Cultivadas , Citometria de Fluxo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-kit/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Superfície Celular/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária
16.
Exp Hematol ; 39(2): 203-213.e1, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21112372

RESUMO

OBJECTIVE: We have successfully identified human cord blood (CB)-derived CD34-negative (CD34(-)) severe combined immunodeficiency (SCID)-repopulating cells (SRCs) with extensive lymphomyeloid repopulating ability using the intrabone marrow injection method. In our previous study, a limiting dilution analysis demonstrated the frequency of CD34(-) SRCs in CB-derived 13lineage-negative (Lin(-)) CD34(-) cells to be approximately 1/25,000. In this study, we intended to develop a high-resolution purification method to obtain highly purified CD34(-) SRCs. MATERIALS AND METHODS: The pooled CB-derived Lin(-) cells were stained with 13 reported Lin monoclonal antibodies (mAbs) and 5 more Lin mAb, against CD11b, CD33, CD66c, CD45RA, and CD127. Then 18Lin(-)CD34(high), 18Lin(-)CD34(-), and 13Lin(-)CD34(high)CD38(-) cells were sorted by fluorescence-activated cell sorting. Stem cell characteristics of these three fractions of cells were analyzed by in vitro cultures and in vivo repopulation assays for evaluation of this new purification method. RESULTS: A limiting dilution analysis demonstrated the frequency of CD34(-) SRCs in these 18Lin(-)CD34(-) cells to be approximately 1/1,000, which is associated with a seeding efficiency 25 times greater than the previous method. All primary recipient nonobese diabetic/Shi-scid/IL-2Rγc(null) mice that received transplants of only two CD34(-) SRCs were highly engrafted with human lymphomyeloid cells at 24 weeks after primary transplantation and showed secondary multilineage repopulating abilities. CONCLUSIONS: We succeeded to highly purify the CD34(-) SRCs using 18Lin mAbs and the intrabone marrow injection technique. This newly developed high-resolution purification method is indispensable to precisely characterize a distinct class of primitive human CB-derived CD34(-) hematopoietic stem cells.


Assuntos
Antígenos CD34/metabolismo , Separação Celular/métodos , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Imunodeficiência Combinada Severa , Animais , Células Cultivadas , Feminino , Sangue Fetal/química , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/química , Humanos , Camundongos , Camundongos Endogâmicos , Camundongos SCID , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Cell Biol Int ; 35(3): 201-8, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20958269

RESUMO

OECs (outgrowth endothelial cells), also known as late-EPCs (late-endothelial progenitor cells), have a high proliferation potential in addition to in vitro tube formation capability. In ischaemic animal models, injected OECs were integrated into regenerating blood vessels and improved neovascularization. Previous reports have demonstrated the expression of CXCL8 to be up-regulated in ischaemic tissues. It has also been documented that CXCL8 stimulates the angiogenic activity of mature ECs (endothelial cells). Therefore, it has been suggested that CXCL8 plays an important role in neovascularization in ischaemic tissues. However, it is still uncertain whether CXCL8 also stimulates the angiogenic activity of OECs. This study evaluated the effects of CXCL8 on the angiogenic activity of OECs in vitro. OECs were isolated from human UCB (umbilical cord blood)-derived mononuclear cells. Phenotypes of the OECs were assessed by flow cytometry, immunostaining, and real-time RT (reverse transcription)-PCR. The effects of CXCL8 on OECs were investigated by transwell migration assay and capillary tube formation assay on Matrigel. The OEC clones isolated from UCB expressed OEC phenotypes. In addition, CXCL8 receptors (CXCR1 and CXCR2) were expressed on these OEC clones. CXCL8 significantly stimulated the transwell migration and capillary tube formation of OECs. Neutralizing antibody against CXCR2, but not CXCR1, abolished a transwell migration of OECs induced by CXCL8, suggesting the involvement of CXCL8/CXCR2 axis in transwell migration. These results demonstrate that CXCL8 stimulates the angiogenic activity of UCB-derived OECs in vitro.


Assuntos
Células Endoteliais/metabolismo , Sangue Fetal/citologia , Interleucina-8/farmacologia , Neovascularização Fisiológica , Anticorpos/imunologia , Movimento Celular , Humanos , Fenótipo , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/metabolismo
19.
J Pharmacol Sci ; 113(3): 271-5, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20647687

RESUMO

Dilazep dihydrochloride (dilazep) is used to treat ischemic dysfunction, although the mechanisms underlying the anti-inflammatory effects of the drug have not yet been elucidated. The present study evaluated the anti-inflammatory effect of dilazep. Dilazep suppressed the production of nitric oxide (NO) and the expression of TNF-alpha mRNA by lipopolysaccharide (LPS) in RAW 264 cells. However, 1400W, an inducible NO synthase inhibitor, suppressed the production of NO but did not suppress the expression of TNF-alpha mRNA following treatment with LPS. Caffeine, an adenosine antagonist, restored LPS-stimulated NO synthesis, which is suppressed by dilazep. Therefore, these observations may suggest that the suppression of NO synthesis after dilazep treatment in RAW 264 cells is caused by the inhibition of TNF-alpha expression via adenosine receptors.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Dilazep/farmacologia , Regulação para Baixo/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Linhagem Celular Transformada , Lipopolissacarídeos/toxicidade , Macrófagos/metabolismo , Camundongos , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/genética , Concentração Osmolar , Antagonistas de Receptores Purinérgicos P1 , RNA Mensageiro/metabolismo , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/genética
20.
Arch Oral Biol ; 55(5): 350-7, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20362276

RESUMO

OBJECTIVE: Tissue stem cells in dental pulp are assumed to possess differentiation potentials similar to mesenchymal stem cells (MSCs). The aim of this in vitro study is to examine the differentiation potentials of mouse dental pulp stem cells (DPSCs) and develop the appropriate differentiation assay systems for skeletal myogenic differentiation of these cells. METHODS: Dental pulps were extracted from mandible sections of C57/BL6 mice, and adherent dental pulp cells were isolated in culture. These cells were cultured in osteogenic or adipogenic induction medium to induce osteogenic and adipogenic differentiation. On the other hand, the skeletal myogenic differentiation potential of these cells was investigated using different conditions, such as serum-free medium, Myod1 overexpression, or 5-Aza-2'-deoxycytidine (5-Aza) treatment for DNA demethylation. Muscle-specific transcriptional factor expression was evaluated by RT-PCR, and myotube formation and myosin heavy chain expression were evaluated by phase-contrast microscopy and immunofluorescence staining, respectively. RESULTS: The adherent dental pulp cells exhibited a proliferative capacity and they showed osteogenic and adipogenic differentiation as seen in previous studies. Although the expression of Myod1 mRNA and myotube formation was not detected in serum-free conditions, the forced expression of Myod1 up-regulated the expression of Myogenin and Pax7 mRNA. However, myotube formation was not confirmed. Interestingly, myosin heavy chain expression and myotube formation were observed following 5-Aza treatment of these cells. CONCLUSIONS: These results demonstrated that mouse DPSCs possess MSC-like differentiation potential. DNA demethylation induced by 5-Aza treatment resulted in the skeletal muscle differentiation in mouse DPSCs, suggesting that DNA demethylation might trigger this differential induction of mouse DPSCs.


Assuntos
Azacitidina/análogos & derivados , Polpa Dentária/citologia , Inibidores Enzimáticos/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Desenvolvimento Muscular/fisiologia , Adipogenia , Animais , Azacitidina/farmacologia , Diferenciação Celular , Células Cultivadas , Metilação de DNA/efeitos dos fármacos , Metilases de Modificação do DNA/antagonistas & inibidores , Decitabina , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Proteína MyoD/biossíntese , Cadeias Pesadas de Miosina/biossíntese , Osteogênese , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA