Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cancer Invest ; 36(9-10): 475-491, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30396283

RESUMO

AS1411 is an antiproliferative DNA aptamer, which binds the ubiquitous protein, nucleolin. In this study, we show that constitutive overexpression of nucleolin confers increased sensitivity to the growth inhibitory effects of AS1411. HeLa cells overexpressing nucleolin have an increased growth rate and invasiveness relative to control cells. Nucleolin overexpressing cells demonstrate increased growth inhibition in response to the AS1411 treatment, which correlates with increased apoptosis and cell cycle arrest, when compared to non-transfected cells. AS1411 induces nucleolin expression at the RNA and protein level in HeLa cells, suggesting a feedback loop with important implications for the clinical use of AS1411.


Assuntos
Oligodesoxirribonucleotídeos/farmacologia , Fosfoproteínas/genética , Proteínas de Ligação a RNA/genética , Regulação para Cima , Neoplasias do Colo do Útero/genética , Aptâmeros de Nucleotídeos , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Retroalimentação Fisiológica , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transfecção , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/metabolismo , Nucleolina
2.
J Exp Med ; 201(3): 373-83, 2005 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-15699072

RESUMO

Bone marrow transplantation offers great promise for treating a number of disease states. However, the widespread application of this approach is dependent upon the development of less toxic methods to establish chimerism and avoid graft-versus-host disease (GVHD). CD8+/TCR- facilitating cells (FCs) have been shown to enhance engraftment of hematopoietic stem cells (HSCs) in allogeneic recipients without causing GVHD. In the present studies, we have identified the main subpopulation of FCs as plasmacytoid precursor dendritic cells (p-preDCs). FCs and p-preDCs share many phenotypic, morphological, and functional features: both produce IFN-alpha and TNF-alpha, both are activated by toll-like receptor (TLR)-9 ligand (CpG ODN) stimulation, and both expand and mature after Flt3 ligand (FL) treatment. FL-mobilized FCs, most of which express a preDC phenotype, significantly enhance engraftment of HSCs and induce donor-specific tolerance to skin allografts. However, p-preDCs alone or p-preDCs from the FC population facilitate HSC engraftment less efficiently than total FCs. Moreover, FCs depleted of preDCs completely fail to facilitate HSC engraftment. These results are the first to define a direct functional role for p-preDCs in HSC engraftment, and also suggest that p-preDCs need to be in a certain state of maturation/activation to be fully functional.


Assuntos
Células Dendríticas/imunologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Plasmócitos/imunologia , Células-Tronco/imunologia , Transplante Homólogo , Animais , Antígeno CD11c/imunologia , Linfócitos T CD8-Positivos/imunologia , Separação Celular , Citocinas/imunologia , DNA/metabolismo , Citometria de Fluxo , Sobrevivência de Enxerto , Doença Enxerto-Hospedeiro , Masculino , Camundongos , Camundongos Endogâmicos , Quimeras de Transplante
3.
Cytokine ; 56(3): 760-8, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22015108

RESUMO

Clefts of the lip and/or palate are among the most prevalent birth defects affecting approximately 7000 newborns in the United States annually. Disruption of the developmentally programmed migration of neural crest cells (NCCs) into the orofacial region is thought to be one of the major causes of orofacial clefting. Signaling of the chemokine SDF-1 (Stromal Derived Factor-1) through its specific receptor, CXCR4, is required for the migration of many stem cell and progenitor cell populations from their respective sites of emergence to the regions where they differentiate into complex cell types, tissues and organs. In the present study, "transwell" assays of chick embryo mesencephalic (cranial) NCC migration and ex ovo whole embryo "bead implantation" assays were utilized to determine whether SDF-1/CXCR4 signaling mediates mesencephalic NCC migration. Results from this study demonstrate that attenuation of SDF-1 signaling, through the use of specific CXCR4 antagonists (AMD3100 and TN14003), disrupts the migration of mesencephalic NCCs into the orofacial region, suggesting a novel role for SDF-1/CXCR4 signaling in the directed migration of mesencephalic NCCs in the early stage embryo.


Assuntos
Movimento Celular/efeitos dos fármacos , Quimiocina CXCL12/farmacologia , Mesencéfalo/citologia , Crista Neural/citologia , Crista Neural/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Embrião de Galinha , Galinhas , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Tubo Neural/citologia , Tubo Neural/efeitos dos fármacos , Peptídeos/farmacologia , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
Differentiation ; 77(5): 462-72, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19349107

RESUMO

Inhibitors of differentiation (Id) proteins are helix-loop-helix (HLH) transcription factors lacking a DNA-binding domain. Id proteins modulate cell proliferation, apoptosis and differentiation in embryonic/fetal tissue. Perturbation of any of these processes in cells of the developing orofacial region results in orofacial anomalies. Chondrogenesis, a process integral to normal orofacial ontogenesis, is known to be modulated, in part, by Id proteins. In the present study, the mRNA and protein expression patterns of Id1, Id2, Id3 and Id4 were examined in developing murine orofacial tissue in vivo, as well as in murine embryonic maxillary mesenchymal cells in vitro. The functional role of Ids during chondrogenesis was also explored in vitro. Results reveal that cells derived from developing murine orofacial tissue (1) express Id1, Id2, Id3 and Id4 mRNAs and proteins on each of gestational days 12-14, (2) express all four Id proteins in a developmentally regulated manner, (3) undergo chondrogenesis and express genes encoding various chondrogenic marker proteins (e.g. Runx2, Type X collagen, Sox9) when cultured under micromass conditions and (4) can have their chondrogenic potential regulated via alteration of Id protein function through overexpression of a basic HLH factor. In summary, results from the current report reveal for the first time the expression of all four Id proteins in cells derived from developing murine orofacial tissue, and demonstrate a functional role for the Ids in regulating the ability of these cells to undergo chondrogenesis.


Assuntos
Condrogênese/genética , Face , Regulação da Expressão Gênica no Desenvolvimento , Proteína 1 Inibidora de Diferenciação/genética , Proteínas/genética , Animais , Face/anatomia & histologia , Face/embriologia , Feminino , Proteína 1 Inibidora de Diferenciação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Gravidez , Proteínas/fisiologia , RNA Mensageiro/metabolismo
5.
PLoS One ; 14(1): e0211046, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30682194

RESUMO

BACKGROUND: Vascular endothelial growth factor (VEGF) is commonly overexpressed in a variety of tumor types including lung cancer. As a key regulator of angiogenesis, it promotes tumor survival, growth, and metastasis through the activation of the downstream protein kinase B (AKT) and extracellular signal-regulated kinase (ERK 1/2) activation. The VEGF promoter contains a 36 bp guanine-rich sequence (VEGFq) which is capable of forming quadruplex (four-stranded) DNA. This sequence has been implicated in the down-regulation of both basal and inducible VEGF expression and represents an ideal target for inhibition of VEGF expression. RESULTS: Our experiments demonstrate sequence-specific interaction between a G-rich quadruplex-forming oligonucleotide encoding a portion of the VEGFq sequence and its double stranded target sequence, suggesting that this G-rich oligonucleotide binds specifically to its complementary C-rich sequence in the genomic VEGF promoter by strand invasion. We show that treatment of A549 non-small lung cancer cells (NSCLC) with this oligonucleotide results in decreased VEGF expression and growth inhibition. The VEGFq oligonucleotide inhibits proliferation and invasion by decreasing VEGF mRNA/protein expression and subsequent ERK 1/2 and AKT activation. Furthermore, the VEGFq oligonucleotide is abundantly taken into cells, localized in the cytoplasm/nucleus, inherently stable in serum and intracellularly, and has no effect on non-transformed cells. Suppression of VEGF expression induces cytoplasmic accumulation of autophagic vacuoles and increased expression of LC3B, suggesting that VEGFq may induce autophagic cell death. CONCLUSION: Our data strongly suggest that the G-rich VEGFq oligonucleotide binds specifically to the C-rich strand of the genomic VEGF promoter, via strand invasion, stabilizing the quadruplex structure formed by the genomic G-rich sequence, resulting in transcriptional inhibition. Strand invading oligonucleotides represent a new approach to specifically inhibit VEGF expression that avoids many of the problems which have plagued the therapeutic use of oligonucleotides. This is a novel approach to specific inhibition of gene expression.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Quadruplex G , Neoplasias Pulmonares/tratamento farmacológico , Proteínas de Neoplasias/biossíntese , Oligonucleotídeos/farmacologia , Regiões Promotoras Genéticas , Fator A de Crescimento do Endotélio Vascular/biossíntese , Células A549 , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Sistemas de Liberação de Medicamentos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Proteínas de Neoplasias/genética , Oligonucleotídeos/genética , Fator A de Crescimento do Endotélio Vascular/genética
6.
PLoS One ; 14(7): e0219610, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31295307

RESUMO

Glyphosate is a broad-spectrum herbicide that is used worldwide. It represents a potential harm to surface water, and when commercially mixed with surfactants, its uptake is greatly magnified. The most well-known glyphosate-based product is Roundup. This herbicide is potentially an endocrine disruptor and many studies have shown the cytotoxicity potential of glyphosate-based herbicides. In breast cancer (BC) cell lines it has been demonstrated that glyphosate can induce cellular proliferation via estrogen receptors. Therefore, we aimed to identify gene expression changes in ER+ and ER- BC cell lines treated with Roundup and AMPA, to address changes in canonical pathways that would be related or not with the ER pathway, which we believe could interfere with cell proliferation. Using the Human Transcriptome Arrays 2.0, we identified gene expression changes in MCF-7 and MDA-MB-468 exposed to low concentrations and short exposure time to Roundup Original and AMPA. The results showed that at low concentration (0.05% Roundup) and short exposure (48h), both cell lines suffered deregulation of 11 canonical pathways, the most important being cell cycle and DNA damage repair pathways. Enrichment analysis showed similar results, except that MDA-MB-468 altered mainly metabolic processes. In contrast, 48h 10mM AMPA showed fewer differentially expressed genes, but also mainly related with metabolic processes. Our findings suggest that Roundup affects survival due to cell cycle deregulation and metabolism changes that may alter mitochondrial oxygen consumption, increase ROS levels, induce hypoxia, damage DNA repair, cause mutation accumulation and ultimately cell death. To our knowledge, this is the first study to analyze the effects of Roundup and AMPA on gene expression in triple negative BC cells. Therefore, we conclude that both compounds can cause cellular damage at low doses in a relatively short period of time in these two models, mainly affecting cell cycle and DNA repair.


Assuntos
Neoplasias da Mama/genética , Glicina/análogos & derivados , Transdução de Sinais/genética , Transcriptoma/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Receptor alfa de Estrogênio/genética , Estrogênios/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicina/farmacologia , Herbicidas/efeitos adversos , Herbicidas/farmacologia , Humanos , Células MCF-7 , Transdução de Sinais/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Glifosato
7.
Transplantation ; 85(8): 1175-84, 2008 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-18431239

RESUMO

BACKGROUND: Fms-related tyrosine kinase 3 (Flt3)-ligand (FL) promotes the proliferation, differentiation, development, and mobilization of hematopoietic cells. We previously found that FL-mobilized hematopoietic stem cells (HSC) engraft efficiently, whereas FL-expanded bone marrow HSC do not. The function of FL-mobilized c-Kit(+) Sca-1(+)Lin(-)(KSL) subpopulations has not been systematically evaluated. A precise definition of the repopulating ability is needed to define which HSC subpopulations are critical for long-term chimerism and tolerance induction. FL significantly mobilized c-Kit(hi) and c-Kit(lo) Sca-1(+)Lin(-) cells into peripheral blood (PB). Here, we evaluated the influence of Flt3 expression on long-term repopulating ability of HSC subpopulations. METHODS: c-Kit(hi) or c-Kit(lo) KSL cells were sorted from PB of FL-treated green fluorescent protein-positive donors. The function of these cells was evaluated using competitive reconstitution assays, colony-forming units spleen, and colony forming cell assays. The function of c-Kit(hi) CD34(-)Flt3(-) KSL, c-Kit CD34(+)Flt3(-) KSL, c-Kit(hi) CD34(+)Flt3(+) KSL were investigated in an in vivo transplantation model. RESULTS: Only FL-mobilized PB c-Kit(hi) KSL cells exhibited high spleen colony-forming unit activity, generated high numbers of both lymphoid and myeloid colonies in vitro, and rescued ablated recipients. FL-mobilization expanded both c-Kit(hi) CD34(+)Flt3(-) cells (short-term HSC) and c-Kit(hi) CD34(-)Flt3(-) KSL cells (long-term HSC). There was a significant decrease in c-Kit CD34Flt3 KSL late multipotent progenitors in PB. A combination of c-Kit(hi) CD34Flt3 and c-Kit CD34(+)Flt3(-) KSL cells offered the most effective rescue of ablated recipients. CONCLUSIONS: These data suggest that engraftment of purified HSC is influenced by both short- and long-term repopulating populations and that Flt3 expression may be useful for selecting the most critical HSC subpopulations for transplantation.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Tirosina Quinase 3 Semelhante a fms/fisiologia , Animais , Antígenos CD34/análise , Diferenciação Celular , Mobilização de Células-Tronco Hematopoéticas , Masculino , Proteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos C57BL
8.
Reprod Toxicol ; 67: 85-99, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27915011

RESUMO

In this study, we identify gene targets and cellular events mediating the teratogenic action(s) of 5-Aza-2'-deoxycytidine (AzaD), an inhibitor of DNA methylation, on secondary palate development. Exposure of pregnant mice (on gestation day (GD) 9.5) to AzaD for 12h resulted in the complete penetrance of cleft palate (CP) in fetuses. Analysis of cells of the embryonic first branchial arch (1-BA), in fetuses exposed to AzaD, revealed: 1) significant alteration in expression of genes encoding several morphogenetic factors, cell cycle inhibitors and regulators of apoptosis; 2) a decrease in cell proliferation; and, 3) an increase in apoptosis. Pyrosequencing of selected genes, displaying pronounced differential expression in AzaD-exposed 1-BAs, failed to reveal significant alterations in CpG methylation levels in their putative promoters or gene bodies. CpG methylation analysis suggested that the effects of AzaD on gene expression were likely indirect.


Assuntos
Azacitidina/análogos & derivados , Região Branquial/efeitos dos fármacos , Fissura Palatina/induzido quimicamente , Desenvolvimento Embrionário/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Azacitidina/toxicidade , Região Branquial/embriologia , Região Branquial/patologia , Proliferação de Células/efeitos dos fármacos , Fissura Palatina/embriologia , Fissura Palatina/genética , Fissura Palatina/patologia , Metilação de DNA/efeitos dos fármacos , Decitabina , Desenvolvimento Embrionário/genética , Feminino , Perfilação da Expressão Gênica , Idade Gestacional , Camundongos Endogâmicos ICR , Gravidez
9.
Circ Res ; 95(12): 1191-9, 2004 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-15550692

RESUMO

The concept that bone marrow (BM)-derived cells participate in cardiac regeneration remains highly controversial and the identity of the specific cell type(s) involved remains unknown. In this study, we report that the postnatal BM contains a mobile pool of cells that express early cardiac lineage markers (Nkx2.5/Csx, GATA-4, and MEF2C). These cells are present in significant amounts in BM harvested from young mice but their abundance decreases with age; in addition, the responsiveness of these cells to gradients of motomorphogens SDF-1, HGF, and LIF changes with age. FACS analysis, combined with analysis of early cardiac markers at the mRNA and protein levels, revealed that cells expressing these markers reside in the nonadherent, nonhematopoietic CXCR4+/Sca-1+/lin-/CD45- mononuclear cell (MNC) fraction in mice and in the CXCR4+/CD34+/AC133+/CD45- BMMNC fraction in humans. These cells are mobilized into the peripheral blood after myocardial infarction and chemoattracted to the infarcted myocardium in an SDF-1-CXCR4-, HGF-c-Met-, and LIF-LIF-R-dependent manner. To our knowledge, this is the first demonstration that the postnatal BM harbors a nonhematopoietic population of cells that express markers for cardiac differentiation. We propose that these potential cardiac progenitors may account for the myocardial regenerative effects of BM. The present findings provide a novel paradigm that could reconcile current controversies and a rationale for investigating the use of BM-derived cardiac progenitors for myocardial regeneration.


Assuntos
Células Sanguíneas/citologia , Células da Medula Óssea/citologia , Coração/fisiologia , Infarto do Miocárdio/patologia , Miocárdio/citologia , Regeneração/fisiologia , Células-Tronco/citologia , Idoso , Animais , Antígenos CD34/análise , Antígenos de Diferenciação/análise , Antígenos Ly/análise , Células da Medula Óssea/química , Células da Medula Óssea/efeitos dos fármacos , Linhagem da Célula , Quimiocina CXCL12 , Quimiocinas CXC/biossíntese , Quimiocinas CXC/genética , Quimiocinas CXC/fisiologia , Quimiotaxia/efeitos dos fármacos , Fator de Crescimento de Hepatócito/biossíntese , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/fisiologia , Humanos , Interleucina-6 , Fator Inibidor de Leucemia , Subunidade alfa de Receptor de Fator Inibidor de Leucemia , Antígenos Comuns de Leucócito/análise , Proteínas de Membrana/análise , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Miocárdio/química , Especificidade de Órgãos , Proteínas/genética , Proteínas/fisiologia , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/fisiologia , RNA Mensageiro/biossíntese , Receptores CXCR4/análise , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/fisiologia , Receptores de Citocinas/antagonistas & inibidores , Receptores de Citocinas/fisiologia , Receptores de OSM-LIF , Células-Tronco/química , Células-Tronco/efeitos dos fármacos , Extratos de Tecidos/farmacologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
10.
PLoS One ; 11(8): e0161588, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27551915

RESUMO

G-quadruplex forming sequences are particularly enriched in the promoter regions of eukaryotic genes, especially of oncogenes. One of the most well studied G-quadruplex forming sequences is located in the nuclease hypersensitive element (NHE) III1 of the c-MYC promoter region. The oncoprotein c-MYC regulates a large array of genes which play important roles in growth regulation and metabolism. It is dysregulated in >70% of human cancers. The silencer NHEIII1 located upstream of the P1 promoter regulates up-to 80% of c-MYC transcription and includes a G-quadruplex structure (Pu27) that is required for promoter inhibition. We have identified, for the first time, a family of seventeen G-quadruplex-forming motifs with >90% identity with Pu27, located on different chromosomes throughout the human genome, some found near or within genes involved in stem cell maintenance or neural cell development. Notably, all members of the Pu27 family interact specifically with NHEIII1 sequence, in vitro. Crosslinking studies demonstrate that Pu27 oligonucleotide binds specifically to the C-rich strand of the NHEIII1 resulting in the G-quadruplex structure stabilization. Pu27 homologous sequences (Pu27-HS) significantly inhibit leukemic cell lines proliferation in culture. Exposure of U937 cells to the Pu27-HS induces cell growth inhibition associated with cell cycle arrest that is most likely due to downregulation of c-MYC expression at the RNA and/or protein levels. Expression of SOX2, another gene containing a Pu27-HS, was affected by Pu27-HS treatment as well. Our data suggest that the oligonucleotides encoding the Pu27 family target complementary DNA sequences in the genome, including those of the c-MYC and SOX2 promoters. This effect is most likely cell type and cell growth condition dependent. The presence of genomic G-quadruplex-forming sequences homologous to Pu27 of c-MYC silencer and the fact that they interact specifically with the parent sequence suggest a common regulatory mechanism for genes whose promoters contain these sequences.


Assuntos
Epistasia Genética , Regulação da Expressão Gênica , Genoma Humano , Genômica , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/genética , Sítios de Ligação , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Bases de Dados Genéticas , Quadruplex G , Genômica/métodos , Humanos , Modelos Biológicos , Família Multigênica , Ligação Proteica , Proteínas Proto-Oncogênicas c-myc/química , Fatores de Transcrição/metabolismo
11.
Diabetes ; 53(8): 1995-2002, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15277378

RESUMO

The mechanism by which mixed chimerism reverses autoimmunity in type 1 diabetes has not been defined. NOD mice have a well-characterized defect in the production of myeloid progenitors that is believed to contribute significantly to the autoimmune process. We therefore investigated whether chimerism induces a correction of this defect. Mixed chimerism restored production of myeloid progenitors in NOD mice to normal levels. Notably, NOD bone marrow cells as well as donor bone marrow cells produced the mature myeloid progeny, and the level of donor chimerism was not correlated with the degree of restoration of the defect. Moreover, NOD bone marrow cells cultured with Flt3-ligand developed a heat-stable antigen-positive/Ly6C+ population comprised primarily of mature myeloid dendritic cells, suggesting that the underlying abnormality is not cell intrinsic but rather due to a block in development of mature myeloid progeny, including myeloid dendritic cells. Strikingly, treatment of NOD mice with Flt3-ligand significantly decreased insulitis and progression to diabetes and was associated with a significant increase in myeloid dendritic cells and in vivo induction of CD4+/CD25+ cells in the pancreatic lymph node. Therefore, Flt3-ligand treatment and/or the establishment of mixed chimerism in prediabetic candidates may provide a benign and novel approach to treat diabetes.


Assuntos
Diabetes Mellitus Tipo 1/prevenção & controle , Proteínas de Membrana/uso terapêutico , Adjuvantes Imunológicos/uso terapêutico , Animais , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Diabetes Mellitus Tipo 1/imunologia , Feminino , Ilhotas Pancreáticas/imunologia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Pâncreas/imunologia , Quimeras de Transplante , Transplante Homólogo
12.
Exp Hematol ; 32(10): 946-54, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15504550

RESUMO

OBJECTIVE: We previously demonstrated that CD8(+)/TCR(-) bone marrow cells facilitate engraftment of HSC in allogeneic recipients without causing graft-vs-host disease. Whether facilitating cells (FC) develop from T cells or represent a distinct lineage has not been determined. METHODS: In the present studies, we characterized the lineage derivation of FC, defined the role for the CD3 complex in allogeneic facilitation, and demonstrated syngeneic facilitation by FC but not T cells. RESULTS: We demonstrate for the first time that FC development and function is independent of T cells and cannot be replaced by them. Purified GFP(+) HSC transplanted in syngeneic recipients produce GFP(+) FC, which facilitate in secondary transplants, confirming that FC are derived from HSC. In addition, FC, but not T cells, potently facilitate the engraftment of suboptimal numbers of HSC in syngeneic recipients. Notably, FC contain the transcripts for CD3 epsilon and CD3 delta, but not TCR alpha or TCR beta, excluding the possibility of T-cell contamination. Genetic mutations that generate a functional deficiency in CD3 signaling significantly impair FC function in allogeneic facilitation (p=0.006). CONCLUSION: Taken together, these data clearly distinguish FC from T cells. Moreover, they indicate that FC require the CD3 epsilon gene to facilitate allogeneic HSC engraftment. The unique function(s) of FC make them an attractive focus for new cell-based therapeutic approaches to enhance HSC engraftment while reducing toxicity, especially when limiting numbers of HSC are available.


Assuntos
Complexo CD3/genética , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Subpopulações de Linfócitos , Animais , Linhagem da Célula , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Transgênicos , RNA Mensageiro/análise , Transplante Homólogo , Transplante Isogênico
13.
Transplantation ; 77(6): 819-26, 2004 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-15077020

RESUMO

BACKGROUND: In the present study we examined the effect of the timing of marrow infusion on engraftment in nonmyeloablatively conditioned mice. METHODS: B10 mice were conditioned with decreasing doses of total body irradiation (TBI) and reconstituted with bone marrow cells (BMCs) from major histocompatibility complex-disparate donor B10.BR mice at 0 or 6 hr, or on days 1, 2, 3, 4, 5, 8, and 12 with respect to TBI. RESULTS: After undergoing conditioning with 700 cGy TBI and transplantation with 15 x 10(6) BMCs, 100% of recipients engrafted if the marrow was infused between 0 and 4 days after TBI. For lower doses of TBI, a delay in infusion of the marrow after TBI conditioning was associated with a significant increase in engraftment. Significantly less engraftment was achieved in animals conditioned with 600 cGy TBI if the marrow was infused at 0 or 6 hr compared with a 1- to 4-day delay. When the TBI was decreased to 500 cGy, engraftment occurred only when the transplant was performed between days 2 and 8. The highest proportion of recipients engrafted when the marrow was infused on day 4. This enhanced engraftment after a delay in marrow infusion is associated with a significant reduction in host mixed lymphocyte reaction reactivity and is correlated inversely with serum levels of interleukin-6 in the recipient. CONCLUSIONS: These data demonstrate for the first time that a delay between conditioning and marrow infusion significantly improves allogeneic engraftment in nonmyeloablatively conditioned recipients and reduces the total conditioning required.


Assuntos
Transplante de Medula Óssea/fisiologia , Animais , Transplante de Medula Óssea/imunologia , Transplante de Medula Óssea/métodos , Rejeição de Enxerto , Ilhotas Pancreáticas/citologia , Transplante das Ilhotas Pancreáticas/métodos , Transplante das Ilhotas Pancreáticas/fisiologia , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Quimeras de Transplante , Transplante Homólogo/fisiologia , Irradiação Corporal Total
14.
Reprod Toxicol ; 37: 40-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23395981

RESUMO

Fetal alcohol syndrome (FAS), presenting with a constellation of neuro-/psychological, craniofacial and cardiac abnormalities, occurs frequently in offspring of women who consume alcohol during pregnancy, with a prevalence of 1-3 per 1000 livebirths. The present study was designed to test the hypothesis that alcohol alters global DNA methylation, and modulates expression of the DNA methyltransferases (DNMTs) and various methyl CpG-binding proteins. Murine embryonic fibroblasts (MEFs), utilized as an in vitro embryonic model system, demonstrated ∼5% reduction in global DNA methylation following exposure to 200mM ethanol. In addition, ethanol induced degradation of DNA methyltransferases (DNMT-1, DNMT-3a, and DNMT-3b), as well as the methyl CpG-binding proteins (MeCP-2, MBD-2 and MBD-3), in MEF cells by the proteasomal pathway. Such degradation could be completely rescued by pretreatment of MEF cells with the proteasomal inhibitor, MG-132. These data support a potential epigenetic molecular mechanism underlying the pathogenesis of FAS during mammalian development.


Assuntos
Metilases de Modificação do DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Etanol/toxicidade , Animais , Metilação de DNA/efeitos dos fármacos , DNA Metiltransferase 3A , Embrião de Mamíferos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Camundongos , Células NIH 3T3
15.
J Immunol ; 180(1): 49-57, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18097003

RESUMO

The use of tolerogenic cells as an approach to induce tolerance to solid organ allografts is being aggressively pursued. A major limitation to the clinical application of cell-based therapies has been the ability to obtain sufficient numbers and also preserve their tolerogenic state. We previously reported that small numbers of bone marrow-derived CD8+/TCR- graft facilitating cells (FC) significantly enhance hemopoietic stem cell (HSC) engraftment in allogeneic and syngeneic recipients. Although the majority of FC resemble precursor plasmacytoid dendritic cells (p-preDC), p-preDC do not replace FC in facilitating function. In the present studies, we investigated the mechanism of FC function. We show for the first time that FC significantly enhance HSC clonogenicity, increase the proportion of multipotent progenitors, and prevent apoptosis of HSC. These effects require direct cell:cell contact between FC and HSC. Separation of FC from HSC by transwell membranes completely abrogates the FC effect on HSC. p-preDC FC do not replace FC total in these effects on HSC function. FC produce TNF-alpha, and FC from TNF-alpha-deficient mice exhibit impaired facilitation in vivo and loss of the in vitro effects on HSC. Neutralizing TNF-alpha in FC similarly blocks the FC effect. The antiapoptotic effect of FC is associated with up-regulation of Bcl-3 transcripts in HSC and blocking of TNF-alpha is associated with abrogation of up-regulation of Bcl-3 transcripts. These data demonstrate a critical role for TNF-alpha in mediating FC function. FC may have a significant impact upon the safe use of chimerism to establish tolerance to transplanted organs and tissue.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Imunologia de Transplantes , Fator de Necrose Tumoral alfa/fisiologia , Animais , Apoptose , Proteína 3 do Linfoma de Células B , Antígenos CD8/análise , Células Dendríticas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Antígenos de Linfócitos T/análise , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética
16.
Stem Cells ; 24(4): 936-48, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16644924

RESUMO

Facilitating cells (CD8+/TCR-) (FCs) enhance engraftment of limiting numbers of hematopoietic stem cells (HSCs). The primary component of FCs is precursor-plasmacytoid dendritic cells (p-preDCs), a tolerogenic cell expanded by Flt3-ligand (FL). In this study, we evaluated the function and composition of FL-expanded FCs. FL treatment resulted in a significant increase of FCs in bone marrow (BM) and peripheral blood (PB). When FL-expanded FCs were transplanted with c-Kit+/Sca-1+/Lin- (KSL) cells into allogeneic recipients, BM-FCs exhibited significantly impaired function whereas PB-FCs were potently functional. A significant upregulation of P-selectin expression and downregulation of VCAM-1 (vascular cell adhesion molecule 1) were present on FL-expanded PB-FCs compared with FL BM-FCs. Stromal cell-derived factor-1 (SDF-1), and CXCR4 transcripts were significantly increased in FL PB-FCs and decreased in FL BM-FCs. Supernatant from FL PB-FCs primed HSC migration to SDF-1, confirming production of the protein product. The FL PB-FCs contained a predominance of p-preDCs and natural killer (NK)-FCs, and NK-FCs were lacking in FL BM-FCs. The impaired function for BM-FCs was restored within 5 days after cessation of treatment. Taken together, these data suggest that FCs may enhance HSC homing and migration via the SDF-1/CXCR4 axis and adhesion molecule modulation. These findings may have implications in development of strategies for retaining function of ex vivo manipulated FCs and HSCs.


Assuntos
Células Sanguíneas/citologia , Células Sanguíneas/efeitos dos fármacos , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Proteínas de Membrana/farmacologia , Animais , Células Sanguíneas/imunologia , Células Sanguíneas/metabolismo , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Quimiocina CXCL12 , Quimiocinas CXC/genética , Quimera , Ensaio de Unidades Formadoras de Colônias , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Tolerância Imunológica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Selectina-P/metabolismo , Transplante de Células-Tronco de Sangue Periférico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores CXCR4/genética , Proteínas Recombinantes/farmacologia , Transplante Homólogo
17.
J Immunol ; 175(6): 3753-61, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16148121

RESUMO

The role that NK cells play in the rejection of hemopoietic stem cell (HSC) and tolerance induction has remained controversial. In this study, we examined whether NK cells play a direct role in the rejection of HSC. Purified HSC from MHC class II-deficient mice engrafted readily in congenic mice, while HSC from class I-deficient donors (beta(2)-microglobulin(-/-) (beta(2)m(-/-))) failed to engraft. Recipient mice lacking CD8(+), CD4(+), or T cells also rejected HSC from class I-deficient donors, pointing directly to NK cells as the effector in rejection of HSC. Recipients, deficient in or depleted of NK cells, engrafted readily with beta(2)m(-/-) HSC. Expression of the activating Ly-49D and inhibitory Ly-49G2 receptors on recipient NK cells was significantly decreased in these beta(2)m(-/-)-->B6 chimeras, and the proportion of donor NK cells expressing Ly-49D was also significantly decreased. Notably, beta(2)m(-/-) chimeras accepted beta(2)m(-/-) HSC in second transplants, demonstrating that NK cells in the chimeras had been tolerized to beta(2)m(-/-). Taken together, our data demonstrate that NK cells play a direct role in the regulation of HSC engraftment, and down-regulation and/or deletion of specific NK subsets in mixed chimeras can contribute to the induction of NK cell tolerance in vivo. Moreover, our data show that bone marrow-derived elements significantly contribute to NK cell development and tolerance.


Assuntos
Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Tolerância Imunológica , Células Matadoras Naturais/fisiologia , Microglobulina beta-2/deficiência , Animais , Antígenos Ly/análise , Rejeição de Enxerto/imunologia , Antígenos de Histocompatibilidade Classe I , Lectinas Tipo C/análise , Subpopulações de Linfócitos , Camundongos , Camundongos Knockout , Receptores Semelhantes a Lectina de Células NK , Linfócitos T
18.
Blood ; 105(5): 2189-97, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15522953

RESUMO

Type 1 diabetes is a systemic autoimmune disease that can be cured by transplantation of hematopoietic stem cells (HSCs) from disease-resistant donors. Nonobese diabetic (NOD) mice have a number of features that distinguish them as bone marrow transplant recipients that must be understood prior to the clinical application of chimerism to induce tolerance. In the present studies, we characterized NOD HSCs, comparing their engraftment characteristics to HSCs from disease-resistant strains. Strikingly, NOD HSCs are significantly enhanced in engraftment potential compared with HSCs from disease-resistant donors. Unlike HSCs from disease-resistant strains, they do not require graft-facilitating cells to engraft in allogeneic recipients. Additionally, they exhibit a competitive advantage when coadministered with increasing numbers of syngeneic HSCs, produce significantly more spleen colony-forming units (CFU-Ss) in vivo in allogeneic recipients, and more granulocyte macrophage-colony-forming units (CFU-GMs) in vitro compared with HSCs from disease-resistant controls. NOD HSCs also exhibit significantly enhanced chemotaxis to a stromal cell-derived factor 1 (SDF-1) gradient and adhere significantly better on primary stroma. This enhanced engraftment potential maps to the insulin-dependent diabetes locus 9 (Idd9) locus, and as such the tumor necrosis factor (TNF) receptor family as well as ski/sno genes may be involved in the mechanism underlying the autonomy of NOD HSCs. These findings may have important implications to understand the evolution of autoimmune disease and impact on potential strategies for cure.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Animais , Quimiotaxia , Sobrevivência de Enxerto , Hematopoese , Camundongos , Camundongos Endogâmicos NOD , Receptores do Fator de Necrose Tumoral/genética , Transplante Homólogo
19.
Blood ; 104(3): 873-80, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15054040

RESUMO

The events that regulate engraftment and long-term repopulating ability of hematopoietic stem cells (HSCs) after transplantation are not well defined. We report for the first time that major histocompatibility complex (MHC) class I K plays a critical role in HSC engraftment via interaction with recipient natural killer (NK) cells. Durable engraftment of purified HSCs requires MHC class I K matching between HSC donor and recipient. In the absence of MHC class I K matching, HSCs exhibit impaired long-term engraftment (P =.01). Dependence on MHC class I K matching is eliminated in B6 beige mice that lack NK cell function, as well as in wild-type mice depleted of NK cells, implicating a possible regulatory role of NK cells for HSC engraftment. The coadministration of CD8+/T-cell receptor-negative (TCR-) graft facilitating cells (FCs) matched at MHC class I K to the HSC donor overcomes the requirement for MHC class I K matching between HSCs and recipient. These data demonstrate that FCs inhibit NK cell effects on the HSCs. Notably, FCs do not suppress the cytotoxic activity of activated NK cells. Enhanced green fluorescent protein-positive (EGFP+) FCs persist for one month following allogeneic transplantation, making cold target inhibition an unlikely mechanism. Therefore, MHC class I may play a critical role in the initiating events that dictate HSC engraftment and/or NK-mediated rejection following allogeneic transplantation.


Assuntos
Sobrevivência de Enxerto/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Células Matadoras Naturais/imunologia , Complexo Principal de Histocompatibilidade , Transplante de Células-Tronco/métodos , Animais , Teste de Histocompatibilidade , Camundongos , Camundongos Endogâmicos , Fatores de Tempo , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa