Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Cytotherapy ; 19(1): 61-74, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27836573

RESUMO

BACKGROUND AIMS: The biodistribution of human MSCs after systemic delivery is incompletely understood. We investigated the changes in cell size and cell surface markers of human MSCs after intravenous (IV) injection in immune competent mice. METHODS: Male human MSCs were labeled with fluorescent vital dye PKH67 and tracked after IV administration in C57/BL6 mice. MSCs were tracked in blood and different murine tissues by human SRY gene quantitative polymerase chain reaction (qPCR) analysis, flow cytometry and fluorescence microscopy. Calibrated microbeads were used to track the size of transplanted MSCs. RESULTS: The majority of injected MSCs were detected by qPCR in the lungs 5 min after transplantation, whereas <0.1% were detected in other tissues over 24 h. Flow cytometric and fluorescence microscopic analysis indicated that MSCs continuously decreased in size after transplantation and underwent fragmentation. The majority of PKH+ MSCs and their fragments were found in lungs and liver. PKH+ MSCs rapidly became positive for annexin V, propidium iodide and calreticulin, indicating loss of cell integrity. In addition, PKH+ fragments co-stained with antibodies against C3b, F4/80 and/or GR-1 indicating opsonization. Preincubation of MSCs in hyperosmolaric hydroxyethyl starch (HyperHAES) decreased MSCs size before transplantation, delayed the loss of viability markers and increased the frequency of traceable MSCs up to 24 h after transplantation. CONCLUSIONS: PKH67 labeled MSCs are fragmented after IV injection in mice, acquire apoptotic and phagocytic cell markers and accumulate in the lungs and liver.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Animais , Apoptose , Biomarcadores/análise , Tamanho Celular , Sobrevivência Celular , Citometria de Fluxo/métodos , Xenoenxertos , Humanos , Injeções Intravenosas , Camundongos Endogâmicos C57BL , Compostos Orgânicos/farmacocinética , Distribuição Tecidual
2.
Blood ; 121(10): 1814-8, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23319569

RESUMO

Bone marrow (BM) provides chemoprotection for acute lymphoblastic leukemia (ALL) cells, contributing to lack of efficacy of current therapies. Integrin alpha4 (alpha4) mediates stromal adhesion of normal and malignant B-cell precursors, and according to gene expression analyses from 207 children with minimal residual disease, is highly associated with poorest outcome. We tested whether interference with alpha4-mediated stromal adhesion might be a new ALL treatment. Two models of leukemia were used, one genetic (conditional alpha4 ablation of BCR-ABL1 [p210(+)] leukemia) and one pharmacological (anti-functional alpha4 antibody treatment of primary ALL). Conditional deletion of alpha4 sensitized leukemia cell to nilotinib. Adhesion of primary pre-B ALL cells was alpha4-dependent; alpha4 blockade sensitized primary ALL cells toward chemotherapy. Chemotherapy combined with Natalizumab prolonged survival of NOD/SCID recipients of primary ALL, suggesting adjuvant alpha4 inhibition as a novel strategy for pre-B ALL.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Resistencia a Medicamentos Antineoplásicos , Proteínas de Fusão bcr-abl/fisiologia , Integrina alfa4/química , Neoplasia Residual/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Medula Óssea/patologia , Adesão Celular , Criança , Citometria de Fluxo , Humanos , Integrases/metabolismo , Integrina alfa4/genética , Integrina alfa4/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Natalizumab , Neoplasia Residual/metabolismo , Neoplasia Residual/mortalidade , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/mortalidade , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Células Estromais/patologia
3.
Cytotherapy ; 13(4): 449-58, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21077730

RESUMO

BACKGROUND AIMS: Enumeration of CD34(+) cells in leukocyte-rich cell suspensions is important for clinical decision-making in stem cell transplantation. Single-platform flow cytometry assays offer the significant advantages of speed and reproducibility, and have therefore become the gold standard in stem cell enumeration. The clinical community has recently defined the need for stem cell enumeration kits that incorporate viability dyes. The purpose of this study was to evaluate a novel assay, BD Biosciences' (BD) stem cell enumeration kit (SCE kit(‡)), in relation to Beckman Coulter's (BC) commercially available BC Stem-Kit™. METHODS: Fresh/freeze-thawed samples from leukapheresis, bone marrow and cord blood, and fresh normal/mobilized blood, were analyzed with both assays (simultaneous detection of side/forward scatter and three fluorescence signals) on two flow cytometry platforms, BD FACSCanto II and BD FACSCalibur. Results. Results from both assays were highly congruent, with an overall r(2) ≥ 0.99 (all specimen types included), a linear correlation across all CD34(+) cell frequencies and concentrations, and an almost ideal steepness of the trend line. CONCLUSIONS: Both assays functioned reliably. Being based on single-platform International Society of Hematotherapy and Graft Engineering (ISHAGE) guidelines and similar staining methods, both assays essentially come to identical results. For most specimen types, the viability of CD34(+) cells was equal to overall leukocyte viability. In summary, in the hands of an experienced technician, the BD™ SCE kit and the BC Stem-Kit are equivalent. The infrequent user might derive benefit from the fact that counting spheres are pre-pipetted into the Trucount tube for the SCE kit, making this assay less susceptible to pipetting inaccuracy.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Contagem de Células/métodos , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Citometria de Fluxo/métodos , Células-Tronco/citologia , Células-Tronco/metabolismo , Antígenos CD34 , Humanos
4.
Exp Hematol ; 78: 35-45, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31562901

RESUMO

Endocannabinoids are lipid mediators that signal via several seven-transmembrane domain G protein-coupled receptors. The endocannabinoid receptor CB2 is expressed on blood cells, including stem cells, and mediates the effects of cannabinoids on the immune system. The role of the endocannabinoid system in immature hematopoiesis is largely elusive. Both direct effects of endocannabinoids on stem cells and indirect effects through endocannabinoid-responsive niche cells like macrophages have been reported. Using two different CB2-deficient mouse models, we studied the role of the endocannabinoid system in immature hematopoiesis. Moreover, we utilized both models to assess the specificity of putative CB2 agonists. As heterodimerization of CB2 and CXCR4, which is highly expressed on hematopoietic stem cells, has already been described, we also assessed potential consequences of CB2 loss for CXCR4/CXCL12 signaling. Overall, no differential effects were observed with any of the compounds tested; the compounds barely induced signaling by themselves, whereas they attenuated CXCL12-induced signals in both CB2-competent and CB2-deficient cells. In vivo experiments were therefore by necessity restricted to loss-of-function studies in knockout (CB2-/-) mice: Except for mild lymphocytosis and slightly elevated circulating progenitor cells, homeostatic hematopoiesis in CB2-/- mice appears to be entirely normal. Mobilization in response to pharmacological stimuli, Plerixafor or G-CSF, was equally potent in wild-type and CB2-/- mice. CB2-/- bone marrow cells reconstituted hematopoiesis in lethally irradiated recipients with engraftment kinetics indistinguishable from those of wild-type grafts. In summary, we found the endocannabinoid system to be largely dispensable for normal murine hematopoiesis.


Assuntos
Endocanabinoides/metabolismo , Regulação da Expressão Gênica , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Modelos Biológicos , Receptor CB2 de Canabinoide/biossíntese , Animais , Endocanabinoides/genética , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Knockout , Receptor CB2 de Canabinoide/genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
5.
Front Immunol ; 8: 44, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28191009

RESUMO

In the colon, a sophisticated balance between immune reaction and tolerance is absolutely required. Dysfunction may lead to pathologic phenotypes ranging from chronic inflammatory processes to cancer development. Two prominent modulators of colon inflammation are represented by the closely related cytokines interleukin (IL)-12 and IL-23, which initiate adaptive Th1 and Th17 immune responses, respectively. In this study, we investigated the impact of the NADPH oxidase protein p47phox, which negatively regulates IL-12 in dendritic cells, on colon cancer development in a colitis-associated colon cancer model. Initially, we found that IL-12-/- mice developed less severe colitis but are highly susceptible to colon cancer. By contrast, p47phox-/- mice showed lower tumor scores and fewer high grade tumors than wild-type (WT) littermates. Treatment with toll-like receptor 9 ligand CpG2216 significantly enhanced colitis in p47phox-/- mice, whereas tumor growth was simultaneously reduced. In tumor tissue of p47phox-/- mice, the IL-23/IL-17 axis was crucially hampered. IL-23p19 protein expression in tumor tissue correlated with tumor stage. Reconstitution of WT mice with IL-23p19-/- bone marrow protected these mice from colon cancer, whereas transplantation of WT hematopoiesis into IL-23p19-/- mice increased the susceptibility to tumor growth. Our study strengthens the divergent role of IL-12 and IL-23 in colon cancer development. With the characterization of p47phox as a novel modulator of both cytokines our investigation introduces a promising new target for antitumor strategies.

6.
Stem Cells Dev ; 24(6): 737-46, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25316534

RESUMO

Hematopoietic stem and progenitor cells (HSPCs) reside in bone marrow (BM) in an environment rich in CXCL12, the ligand for CXCR4, which is constitutively expressed on all immature hematopoietic cells in BM. This ligand-receptor pair critically controls HSPC retention and (relative) quiescence in BM. Interestingly, in a chemokine-abundant environment, CXCR4 surface expression and CXCL12 sensitivity of BM-residing HSPCs are continuously maintained. The mechanisms underlying this peculiar pattern of G-protein signal integration by BM-HSPCs are unknown. G-protein receptor kinases (GRKs) control receptor function by phosphorylating the intracellular domains upon ligand-induced activation, which results in receptor internalization and transient refractoriness. Using, therefore, a GRK6-deficient (GRK6(-/-)) mouse, we sought to address how perturbed ligand-induced CXCR4 (in)activation affects HSPC behavior in vitro and in vivo. In vitro, GRK6(-/-) HSPCs were characterized by hyper-responsiveness to CXCL12, as expected. In vivo, GRK6(-/-) immature hematopoiesis was characterized by a marked expansion of immature hematopoiesis in spleens and a modest repopulation defect in serial competitive transplantation. Enforced mobilization with granulocyte colony-stimulating factor (G-CSF) and AMD3100 was normal, as was hematopoietic regeneration after noncompetitive transplantation or pharmacological myelosuppression. These observations illustrate that GRK-mediated restriction of CXCR4 signal input after ligand engagement is largely dispensable for BM-resident HSPCs, which may explain how continuous CXCL12 responsiveness of BM-HSPCs can be maintained.


Assuntos
Quimiocina CXCL12/metabolismo , Quinases de Receptores Acoplados a Proteína G/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Quinases de Receptores Acoplados a Proteína G/genética , Células-Tronco Hematopoéticas/citologia , Camundongos , Fator Plaquetário 4/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa