Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Cancer Gene Ther ; 29(8-9): 1263-1275, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35194200

RESUMO

DNA methylation, a major biological process regulating the transcription, contributes to the pathophysiology of hematologic malignancies, and hypomethylating agents are commonly used to treat myelodysplastic syndromes (MDS) and acute myeloid leukemias (AML). In these diseases, bone marrow mesenchymal stromal cells (MSCs) play a key supportive role through the production of various signals and interactions. The DNA methylation status of MSCs, likely to reflect their functionality, might be relevant to understand their contribution to the pathophysiology of these diseases. Consequently, the aim of our study was to analyze the modifications of DNA methylation profiles of MSCs induced by MDS or AML. MSCs from MDS/AML patients were characterized via 5-methylcytosine quantification, gene expression profiles of key regulators of DNA methylation, identification of differentially methylated regions (DMRs) by methylome array, and quantification of DMR-coupled genes expression. MDS and AML-MSCs displayed global hypomethylation and under-expression of DNMT1 and UHRF1. Methylome analysis revealed aberrant methylation profiles in all MDS and in a subgroup of AML-MSCs. This aberrant methylation was preferentially found in the sequence of homeobox genes, especially from the HOX family (HOXA1, HOXA4, HOXA5, HOXA9, HOXA10, HOXA11, HOXB5, HOXC4, and HOXC6), and impacted on their expression. These results highlight modifications of DNA methylation in MDS/AML-MSCs, both at global and focal levels dysregulating the expression of HOX genes well known for their involvement in leukemogenesis. Such DNA methylation in MSCs could be the consequence of the malignant disease or could participate in its development through defective functionality or exosomal transfer of HOX transcription factors from MSCs to hematopoietic cells.


Assuntos
Leucemia Mieloide Aguda , Células-Tronco Mesenquimais , Síndromes Mielodisplásicas , Medula Óssea/patologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Metilação de DNA , Genes Homeobox/genética , Humanos , Leucemia Mieloide Aguda/patologia , Células-Tronco Mesenquimais/metabolismo , Síndromes Mielodisplásicas/genética , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/metabolismo
2.
Cancers (Basel) ; 13(22)2021 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-34830855

RESUMO

(1) Background: The impact of occupational exposure to high doses of pesticides on hematologic disorders is widely studied. Yet, lifelong exposure to low doses of pesticides, and more particularly their cocktail effect, although poorly known, could also participate to the development of such hematological diseases as myelodysplastic syndrome (MDS) in elderly patients. (2) Methods: In this study, a cocktail of seven pesticides frequently present in water and food (maneb, mancozeb, iprodione, imazalil, chlorpyrifos ethyl, diazinon and dimethoate), as determined by the European Food Safety Authority, were selected. Their in vitro effects at low-doses on primary BM-MSCs from healthy volunteers were examined. (3) Results: Exposure of normal BM-MSCs to pesticides for 21 days inhibited cell proliferation and promoted DNA damage and senescence. Concomitantly, these cells presented a decrease in aldehyde dehydrogenase 2 (ALDH2: mRNA, protein and enzymatic activity) and an increase in acetaldehyde levels. Pharmacological inhibition of ALDH2 with disulfiram recapitulated the alterations induced by exposure to low doses of pesticides. Moreover, BM-MSCs capacity to support primitive hematopoiesis was significantly altered. Similar biological abnormalities were found in primary BM-MSCs derived from MDS patients. (4) Conclusions: these results suggest that ALDH2 could participate in the pathophysiology of MDS in elderly people long exposed to low doses of pesticides.

3.
Int J Mol Sci ; 21(22)2020 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-33202543

RESUMO

The bone marrow (BM) microenvironment plays a crucial role in the development and progression of leukemia (AML). Intracellular reactive oxygen species (ROS) are involved in the regulation of the biology of leukemia-initiating cells, where the antioxidant enzyme GPx-3 could be involved as a determinant of cellular self-renewal. Little is known however about the role of the microenvironment in the control of the oxidative metabolism of AML cells. In the present study, a coculture model of BM mesenchymal stromal cells (MSCs) and AML cells (KG1a cell-line and primary BM blasts) was used to explore this metabolic pathway. MSC-contact, rather than culture with MSC-conditioned medium, decreases ROS levels and inhibits the Nrf-2 pathway through overexpression of GPx3 in AML cells. The decrease of ROS levels also inactivates p38MAPK and reduces the proliferation of AML cells. Conversely, contact with AML cells modifies MSCs in that they display an increased oxidative stress and Nrf-2 activation, together with a concomitant lowered expression of GPx-3. Altogether, these experiments suggest that a reciprocal control of oxidative metabolism is initiated by direct cell-cell contact between MSCs and AML cells. GPx-3 expression appears to play a crucial role in this cross-talk and could be involved in the regulation of leukemogenesis.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glutationa Peroxidase/biossíntese , Leucemia Mieloide Aguda/enzimologia , Proteínas de Neoplasias/biossíntese , Microambiente Tumoral , Linhagem Celular Tumoral , Técnicas de Cocultura , Humanos , Leucemia Mieloide Aguda/patologia , Células-Tronco Mesenquimais/enzimologia , Células-Tronco Mesenquimais/patologia , Oxirredução
4.
PLoS One ; 15(6): e0225485, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32484831

RESUMO

Mesenchymal stem cells (MSC)-spheroid models favor maintenance of stemness, ex vivo expansion and transplantation efficacy. Spheroids may also be considered as useful surrogate models of the hematopoietic niche. However, accessibility to primary cells, from bone marrow (BM) or adipose tissues, may limit their experimental use and the lack of consistency in methods to form spheroids may affect data interpretation. In this study, we aimed to create a simple model by examining the ability of cell lines, from human (HS-27a and HS-5) and murine (MS-5) BM origins, to form spheroids, compared to primary human MSCs (hMSCs). Our protocol efficiently allowed the spheroid formation from all cell types within 24 hours. Whilst hMSC-spheroids began to shrink after 24 hours, the size of spheroids from cell lines remained constant during three weeks. The difference was partially explained by the balance between proliferation and cell death, which could be triggered by hypoxia and induced oxidative stress. Our results demonstrate that, like hMSCs, MSC cell lines make reproductible spheroids that are easily handled. Thus, this model could help in understanding mechanisms involved in MSC functions and may provide a simple model by which to study cell interactions in the BM niche.


Assuntos
Células-Tronco Mesenquimais/citologia , Esferoides Celulares/citologia , Animais , Agregação Celular , Morte Celular , Desdiferenciação Celular , Hipóxia Celular , Linhagem Celular , Proliferação de Células , Humanos , Camundongos , Estresse Oxidativo
5.
Oncogene ; 39(10): 2227, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31700154

RESUMO

The original version of this Article omitted the following from the Acknowledgements: This research was also supported by grants to KZ (UL and L-CNRS). This has now been corrected in both the PDF and HTML versions of the Article.

6.
Oncogene ; 39(6): 1198-1212, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31649334

RESUMO

The bone marrow (BM) niche impacts the progression of acute myeloid leukemia (AML) by favoring the chemoresistance of AML cells. Intimate interactions between leukemic cells and BM mesenchymal stromal cells (BM-MSCs) play key roles in this process. Direct intercellular communications between hematopoietic cells and BM-MSCs involve connexins, components of gap junctions. We postulated that blocking gap junction assembly could modify cell-cell interactions in the leukemic niche and consequently the chemoresistance. The comparison of BM-MSCs from AML patients and healthy donors revealed a specific profile of connexins in BM-MSCs of the leukemic niche and the effects of carbenoxolone (CBX), a gap junction disruptor, were evaluated on AML cells. CBX presents an antileukemic effect without affecting normal BM-CD34+ progenitor cells. The proapoptotic effect of CBX on AML cells is in line with the extinction of energy metabolism. CBX acts synergistically with cytarabine (Ara-C) in vitro and in vivo. Coculture experiments of AML cells with BM-MSCs revealed that CBX neutralizes the protective effect of the niche against the Ara-C-induced apoptosis of leukemic cells. Altogether, these results suggest that CBX could be of therapeutic interest to reduce the chemoresistance favored by the leukemic niche, by targeting gap junctions, without affecting normal hematopoiesis.


Assuntos
Carbenoxolona/farmacologia , Citarabina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Junções Comunicantes/efeitos dos fármacos , Leucemia Mieloide Aguda/tratamento farmacológico , Células-Tronco Mesenquimais/citologia , Microambiente Tumoral/efeitos dos fármacos , Animais , Antiulcerosos/farmacologia , Antimetabólitos Antineoplásicos/farmacologia , Apoptose , Proliferação de Células , Quimioterapia Combinada , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Blood Adv ; 3(24): 4271-4279, 2019 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-31869414

RESUMO

Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal stem cell disorders with an inherent tendency for transformation in secondary acute myeloid leukemia. This study focused on the redox metabolism of bone marrow (BM) cells from 97 patients compared with 25 healthy controls. The level of reactive oxygen species (ROS) was quantified by flow cytometry in BM cell subsets as well as the expression level of 28 transcripts encoding for major enzymes involved in the antioxidant cellular response. Our results highlight increased ROS levels in BM nonlymphoid cells and especially in primitive CD34posCD38low progenitor cells. Moreover, we identified a specific antioxidant signature, dubbed "antioxidogram," for the different MDS subgroups or secondary acute myeloblastic leukemia (sAML). Our results suggest that progression from MDS toward sAML could be characterized by 3 successive molecular steps: (1) overexpression of enzymes reducing proteic disulfide bonds (MDS with <5% BM blasts [GLRX family]); (2) increased expression of enzymes detoxifying H2O2 (MDS with 5% to 19% BM blasts [PRDX and GPX families]); and finally (3) decreased expression of these enzymes in sAML. The antioxidant score (AO-Score) defined by logistic regression from the expression levels of transcripts made it possible to stage disease progression and, interestingly, this AO-Score was independent of the revised International Scoring System. Altogether, this study demonstrates that MDS and sAML present an important disturbance of redox metabolism, especially in BM stem and progenitor cells and that the specific molecular antioxidant response parameters (antioxidogram, AO-Score) could be considered as useful biomarkers for disease diagnosis and follow-up.


Assuntos
Antioxidantes/metabolismo , Células da Medula Óssea/metabolismo , Medula Óssea/metabolismo , Síndromes Mielodisplásicas/metabolismo , Estresse Oxidativo , Medula Óssea/patologia , Células da Medula Óssea/patologia , Estudos de Casos e Controles , Células Cultivadas , Progressão da Doença , Regulação Enzimológica da Expressão Gênica , Humanos , Imunofenotipagem , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Metabolômica/métodos , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/patologia , Espécies Reativas de Oxigênio/metabolismo
8.
Ann Hematol ; 98(10): 2367-2377, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31455988

RESUMO

The coexistence of dual hematological neoplasms is very rare. Sequential or synchronous neoplasms in hematology are an uncommon and complex clinical situation. The aim of the Hemo2 study was to describe the clinical characteristics and analyze the outcome of these patients. We performed a retrospective review of all patients diagnosed with sequential or synchronous hematological malignancies in the university hospital of Tours, between 2007 and 2018. We identified 49 patients in our study, with a prevalence of 0.89%. Sequential and synchronous combinations were found in 36 (73%) and 13 (27%) patients, respectively. One patient presented three sequential neoplasms. The median cumulative incidence was 6 years (95% CI 3-7). Among all neoplasms diagnosed (n = 99), we found 79 lymphoid neoplasms (LNs) (80%) and 20 myeloid neoplasms (MNs) (20%). Sex ratio was 1.88 with 65% of males and 35% of females. The most common LNs were Hodgkin lymphoma (n = 16; 16%) and multiple myeloma (n = 11; 11%). The most frequent MN was essential thrombocythemia (n = 5; 5%). The most common combination was Hodgkin lymphoma and follicular lymphoma in five (10%) patients. The overall survival from the first diagnosis (OS1) at 5 years was 82.4% (95% CI 72.1-94.3). The median overall survival from the second diagnosis (OS2) was 98 months (95% CI 44-NR) and 5-year OS2 was 58.7% (95% CI 45.5-75.7). Median progression-free survival from the second diagnosis (PFS) was 47 months (95% CI 27-NR) with 5-year PFS of 49% (95% CI 35.9-67). OS and PFS did not statistically differ between synchronous and sequential dual neoplasms. In this cohort, that the death relative risk (RR) was significantly lower if the second neoplasm appeared after more than 4 years following the first diagnosis (OR 0.37 (95% CI 0.16-0.90)). The Hemo2study confirmed the rarity of dual hematological neoplasms. In this cohort, HL and FL were the most frequent combinations. Our results may support that synchronous and sequential dual neoplasms bear the same prognosis. Further studies are needed to better characterize these uncommon clinical situations.


Assuntos
Neoplasias Hematológicas , Segunda Neoplasia Primária , Idoso , Idoso de 80 Anos ou mais , Intervalo Livre de Doença , Feminino , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/mortalidade , Neoplasias Hematológicas/patologia , Neoplasias Hematológicas/terapia , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Segunda Neoplasia Primária/diagnóstico , Segunda Neoplasia Primária/mortalidade , Segunda Neoplasia Primária/patologia , Segunda Neoplasia Primária/terapia , Prevalência , Estudos Retrospectivos , Taxa de Sobrevida
9.
Pharmacol Res ; 136: 45-55, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30142422

RESUMO

Acute Myeloid Leukemia (AML) remains a therapeutic challenge and improvements in chemotherapy are needed. n-3 polyunsaturated fatty acids (PUFAs), present in fish oil (FO) at high concentrations, have antitumoral properties in various cancer models. We investigated the effects of two n-3 PUFAs, docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), in AML cell lines and primary AML blasts. EPA and DHA induced a dose-dependent decrease in cell viability in five AML cell lines, which was also observed with FO, but not SO (devoid of n-3 PUFAs) in cell lines and primary leucoblasts. Mitochondrial energy metabolism shifted from oxidative respiration to glycolytic metabolism in the U937, MOLM-13, and HL-60 cell lines. This phenomenon was associated with major disorganization of the mitochondrial network and mitochondrial swelling. Transcriptomic analysis after 6 h and 24 h of exposure to FO revealed a Nrf2 activation signature, which was confirmed by evidence of Nrf2 nuclear translocation in response to oxidative stress, but insufficient to prevent cell death following prolonged exposure. Apoptosis studies showed consistent phosphatidylserine exposition among the AML cell lines tested and a reduced mitochondrial membrane potential. The cell-killing effect of FO was additive with that of cytarabine (AraC), by the Chou and Talalay method, and this combination effect could be reproduced in primary AML blasts. Altogether, our results show deleterious effects of n-3 PUFAs on mitochondrial metabolism of AML cells, associated with oxidative stress and Nrf2 response, leading to cell death. These observations support further investigation of n-3 PUFA addition to standard chemotherapy in AML.


Assuntos
Antineoplásicos/farmacologia , Citarabina/farmacologia , Ácidos Graxos Ômega-3/farmacologia , Óleos de Peixe/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Metabolismo Energético/efeitos dos fármacos , Glicólise , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fator 2 Relacionado a NF-E2/genética , Transdução de Sinais/efeitos dos fármacos
10.
Artigo em Inglês | MEDLINE | ID: mdl-29695069

RESUMO

Exergames, more specifically console-based exergames, are generally enjoyed by adolescents and known to increase physical activity. Nevertheless, they have a reduced usage over time and demonstrate little effectiveness over the long term. In order to increase playing time, mobile exergames may increase potential playing time, but need to be engaging and integrated in everyday life. The goal of the present study was to examine the context of gameplay for mobile exergaming in adolescents’ everyday life to inform game design and the integration of gameplay into everyday life. Eight focus groups were conducted with 49 Flemish adolescents (11 to 17 years of age). The focus groups were audiotaped, transcribed, and analyzed by means of thematic analysis via Nvivo 11 software (QSR International Pty Ltd., Victoria, Australia). The adolescents indicated leisure time and travel time to and from school as suitable timeframes for playing a mobile exergame. Outdoor gameplay should be restricted to the personal living environment of adolescents. Besides outdoor locations, the game should also be adaptable to at-home activities. Activities could vary from running outside to fitness exercises inside. Furthermore, the social context of the game was important, e.g., playing in teams or meeting at (virtual) meeting points. Physical activity tracking via smart clothing was identified as a motivator for gameplay. By means of this study, game developers may be better equipped to develop mobile exergames that embed gameplay in adolescents’ everyday life.


Assuntos
Exercício Físico/fisiologia , Jogos Recreativos , Estilo de Vida Saudável , Jogos de Vídeo , Adolescente , Saúde do Adolescente , Bélgica , Criança , Exercício Físico/psicologia , Feminino , Jogos Recreativos/psicologia , Humanos , Masculino , Desenvolvimento de Programas , Avaliação de Programas e Projetos de Saúde , Comportamento Sedentário , Jogos de Vídeo/psicologia
11.
J Am Acad Dermatol ; 78(5): 973-982.e8, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29273486

RESUMO

BACKGROUND: Merkel cell carcinoma (MCC) is a rare tumor of the skin that has an aggressive behavior. Immunity is the main regulator of MCC development, and many interactions between lymphocytes and tumor cells have been proven. However, the impact of tumor-infiltrating myeloid cells needs better characterization. OBJECTIVE: To characterize tumor-infiltrating myeloid cells in MCC and their association with other immune effectors and patient outcome. METHODS: MCC cases were reviewed from an ongoing prospective cohort study. In all, 103 triplicate tumor samples were included in a tissue microarray. Macrophages, neutrophils, and myeloid-derived suppressor cells were characterized by the following markers: CD68, CD33, CD163, CD15, CD33, and human leukocyte antigen-DR. Associations of these cell populations with programmed cell death ligand 1 expression, CD8 infiltrates, and vascular density were assessed. Impact on survival was analyzed by log-rank tests and a Cox multivariate model. RESULTS: The median density of macrophages was 216 cells/mm2. CD68+ and CD33+ macrophage densities were associated with CD8+ T-cell infiltrates and programmed cell death ligand 1 expression. In addition, MCC harboring CD8+ T cell infiltrates and brisk CD33+ myeloid cell infiltrates were significantly and independently associated with improved outcomes (recurrence-free and overall survival). LIMITATIONS: Sampling bias and the retrospective design were potential study limitations. CONCLUSION: Infiltration of CD33+ myeloid cells and CD8+ T lymphocytes defines a subset of MCC associated with improved outcome.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Carcinoma de Célula de Merkel/mortalidade , Carcinoma de Célula de Merkel/patologia , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Neoplasias Cutâneas/mortalidade , Neoplasias Cutâneas/patologia , Biomarcadores Tumorais/metabolismo , Biópsia por Agulha , Linfócitos T CD8-Positivos/metabolismo , Carcinoma de Célula de Merkel/terapia , Causas de Morte , Estudos de Coortes , Progressão da Doença , Intervalo Livre de Doença , Feminino , Humanos , Imuno-Histoquímica , Masculino , Análise Multivariada , Células Mieloides/imunologia , Células Mieloides/patologia , Invasividade Neoplásica/patologia , Estadiamento de Neoplasias , Prognóstico , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Medição de Risco , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Neoplasias Cutâneas/terapia , Estatísticas não Paramétricas , Análise de Sobrevida , Fatores de Tempo
12.
Leuk Res ; 60: 94-102, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28772207

RESUMO

Although it has been well-demonstrated that bone marrow mesenchymal stromal cells (MSCs) from CML patients do not belong to the Ph1-positive clone, there is growing evidence that they could play a role in the leukemogenesis process or the protection of leukemic stem cells from the effects of tyrosine kinase inhibitors (TKIs). The aim of the present study was to identify genes differentially expressed in MSCs isolated from CML patients at diagnosis (CML-MSCs) as compared to MSCs from healthy controls. Using a custom gene-profiling assay, we identified six genes over-expressed in CML-MSCs (BMP1, FOXO3, MET, MITF, NANOG, PDPN), with the two highest levels being documented for PDPN (PODOPLANIN) and NANOG. To determine whether this aberrant signature persisted in patients in deep molecular response induced by TKIs, we analyzed MSCs derived from such patients (MR-MSCs). This analysis showed that, despite the deep molecular responses, BMP1, MET, MITF, NANOG, and PDPN mRNA were upregulated in MR-MSCs. Moreover, BMP1, MITF, and NANOG mRNA expressions in MR-MSCs were found to be intermediate between control MSCs and CML-MSCs. These results suggest that CML-MSCs exhibit an abnormal gene expression pattern which might have been established during the leukemogenic process and persist in patients in deep molecular response.


Assuntos
Medula Óssea/patologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Células-Tronco Mesenquimais/patologia , Animais , Transformação Celular Neoplásica/genética , Perfilação da Expressão Gênica , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Camundongos , Inibidores de Proteínas Quinases , Proteínas Tirosina Quinases/antagonistas & inibidores , RNA Mensageiro/análise , Indução de Remissão , Regulação para Cima
13.
Stem Cells Dev ; 26(10): 709-722, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28394200

RESUMO

Bone marrow (BM)-derived mesenchymal stromal cells (MSCs) frequently display alterations in several hematologic disorders, such as acute lymphoid leukemia, acute myeloid leukemia (AML), and myelodysplastic syndromes. In acute leukemias, it is not clear whether MSC alterations contribute to the development of the malignant clone or whether they are simply the effect of tumor expansion on the microenvironment. We extensively investigated the characteristics of MSCs isolated from the BM of patients with de novo AML at diagnosis (L-MSCs) in terms of phenotype (gene and protein expression, apoptosis and senescence levels, DNA double-strand break formation) and functions (proliferation and clonogenic potentials, normal and leukemic hematopoiesis-supporting activity). We found that L-MSCs show reduced proliferation capacity and increased apoptosis levels compared with MSCs from healthy controls. Longer population doubling time in L-MSCs was not related to the AML characteristics at diagnosis (French-American-British type, cytogenetics, or tumor burden), but was related to patient age and independently associated with poorer patient outcome, as was cytogenetic prognostic feature. Analyzing, among others, the expression of 93 genes, we found that proliferative deficiency of L-MSCs was associated with a perivascular feature at the expense of the osteo-chondroblastic lineage with lower expression of several niche factors, such as KITLG, THPO, and ANGPT1 genes, the cell adhesion molecule VCAM1, and the developmental/embryonic genes, BMI1 and DICER1. L-MSC proliferative capacity was correlated positively with CXCL12, THPO, and ANGPT1 expression and negatively with JAG1 expression. Anyway, these changes did not affect their in vitro capacity to support normal hematopoiesis and to modify leukemic cell behavior (protection from apoptosis and quiescence induction). Our findings indicate that BM-derived MSCs from patients with newly diagnosed AML display phenotypic and functional alterations such as proliferative deficiency that could be attributed to tumor progression, but does not seem to play a special role in the leukemic process.


Assuntos
Biomarcadores Tumorais/genética , Leucemia Mieloide Aguda/patologia , Células-Tronco Mesenquimais/metabolismo , Fenótipo , Biomarcadores Tumorais/metabolismo , Estudos de Casos e Controles , Proliferação de Células , Quebras de DNA de Cadeia Dupla , Feminino , Hematopoese , Humanos , Masculino , Células-Tronco Mesenquimais/patologia , Células-Tronco Mesenquimais/fisiologia , Pessoa de Meia-Idade , Microambiente Tumoral
14.
Oncotarget ; 8(26): 41876-41889, 2017 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-27566554

RESUMO

STAT5 transcription factors are frequently activated in hematopoietic neoplasms and are targets of various tyrosine kinase oncogenes. Evidences for a crosstalk between STAT5 and reactive oxygen species (ROS) metabolism have recently emerged but mechanisms involved in STAT5-mediated regulation of ROS still remain elusive. We demonstrate that sustained activation of STAT5 induced by Bcr-Abl in chronic myeloid leukemia (CML) cells promotes ROS production by repressing expression of two antioxidant enzymes, catalase and glutaredoxin-1(Glrx1). Downregulation of catalase and Glrx1 expression was also observed in primary cells from CML patients. Catalase was shown not only to reduce ROS levels but also, to induce quiescence in Bcr-Abl-positive leukemia cells. Furthermore, reduction of STAT5 phosphorylation and upregulation of catalase and Glrx1 were also evidenced in leukemia cells co-cultured with bone marrow stromal cells to mimic a leukemic niche. This caused downregulation of ROS levels and enhancement of leukemic cell quiescence. These data support a role of persistent STAT5 signaling in the regulation of ROS production in myeloid leukemias and highlight the repression of antioxidant defenses as an important regulatory mechanism.


Assuntos
Antioxidantes/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Estresse Oxidativo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Catalase/metabolismo , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Glutarredoxinas/genética , Glutarredoxinas/metabolismo , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Modelos Biológicos , Espécies Reativas de Oxigênio/metabolismo
15.
Int Orthop ; 38(9): 1845-53, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24728310

RESUMO

PURPOSE: Tibial fractures are the most common lower limb fractures. Some criteria such as open fractures and increasing open stage are known to be associated with high delayed union and pseudarthrosis rate. In cases of delayed or nonunion, classical treatment is autologous cancelous bone graft which is associated with high morbidity rate. The ideal treatment would be a percutaneous harvesting and grafting technique. As bone marrow autologous concentrate (BMAC) presents both advantages, we evaluated this technique from 2002 to 2007. METHODS: This was a retrospective study of 43 cases of open tibial fractures with initial surgical treatment. The criteria of inclusion were open fracture and nonunion, delayed union or suspicion of delayed union. RESULTS: In 23 cases (53.5 %) BMAC was successful. The success group had received significantly more CFU-F than the failure group (469 vs 153.10(3), p = 0.013). A threshold of 360.10(3) CFU-F grafted could be established over which there was 100 % success. BMAC done before 110 days after fracture had 47 % success and BMAC done since 110 days after fracture had 73 % success. BMAC success rate decreased with increasing initial fracture skin open stage. There was no BMAC success in cases of a fracture with a remaining gap of more than 4 mm. We had no complications with the technique at the iliac harvesting zone and tibia injection point. CONCLUSION: BMAC is a technique that should be considered as one of the different alternatives for management of long-bone delayed and nonunion because of its effectiveness, low complication rate, preservation of bone stock and low cost.


Assuntos
Transplante de Medula Óssea/métodos , Transplante Ósseo/métodos , Fraturas Mal-Unidas/cirurgia , Fraturas da Tíbia/cirurgia , Administração Cutânea , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Ílio/citologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
16.
Vet Immunol Immunopathol ; 153(1-2): 57-69, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23477930

RESUMO

Toxoplasmosis is a worldwide epizootic disease of mammals. Chickens, albeit being less susceptible, can be contaminated in free-range flocks and may have an important role in parasite transmission. Plastic adherence selection of chicken spleen cells enriched 8F2+ (putative chicken CD11c) MHC II+ cells of the myeloid type; however, we did not succeed to separate dendritic cells from macrophages using their feature to become loosely adherent after culture as in mammals. Still we clearly identified dendritic-like cells being morphologically distinguishable from macrophages in the KUL01 (macrophage marker) negative fraction, exhibiting responsiveness to LPS and parasite extracts by developing characteristic cellular protrusions as well as a minor phagocytic incorporation of dead parasites. Live T. gondii tachyzoites were able to invade the two different types of myeloid adherent cells, to replicate, and to induce an overall decrease in the expression of MHC II and co-stimulatory molecules, CD80 and CD40. Our data indicate that dendritic cells in addition to macrophages may have a role in hiding viable replicating T. gondii tachyzoites from the immune system and in shuttling them to different organs in the chicken as previously described for different Apicomplexa infecting mammals.


Assuntos
Galinhas/imunologia , Células Dendríticas/imunologia , Baço/imunologia , Toxoplasma/imunologia , Toxoplasmose Animal/imunologia , Animais , Adesão Celular , Macrófagos/imunologia , Fagocitose , Baço/citologia
17.
Stem Cells Dev ; 21(17): 3162-72, 2012 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-22651889

RESUMO

Human hematopoietic stem/progenitor cells (HSPCs) can be mobilized into the circulation using granulocyte-colony stimulating factor (G-CSF), for graft collection in view of hematopoietic transplantation. This process has been related to bone marrow (BM) release of serine proteases and of the matrix metalloproteinase-9 (MMP-9). Yet, the role of these mediators in HSC egress from their niches remains questionable, because they are produced by nonstromal cells (mainly neutrophils and monocytes/macrophages) that are not a part of the niche. We show here that the G-CSF receptor (G-CSFR) is expressed by human BM mesenchymal stromal/stem cells (MSCs), and that G-CSF prestimulation of MSCs enhances the in vitro trans-stromal migration of CD34+ cells. Zymography analysis indicates that pro-MMP-2 (but not pro-MMP-9) is expressed in MSCs, and that G-CSF treatment increases its expression and induces its activation at the cell membrane. We further demonstrate that G-CSF-stimulated migration depends on G-CSFR expression and is mediated by a mechanism that involves MMPs. These results suggest a molecular model whereby G-CSF infusion may drive, by the direct action on MSCs, HSPC egress from BM niches via synthesis and activation of MMPs. In this model, MMP-2 instead of MMP-9 is implicated, which constitutes a major difference with mouse mobilization models.


Assuntos
Antígenos CD34/metabolismo , Células da Medula Óssea/citologia , Movimento Celular , Fator Estimulador de Colônias de Granulócitos/farmacologia , Metaloproteinase 2 da Matriz/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Antígenos CD34/genética , Células da Medula Óssea/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/enzimologia , Membrana Celular/metabolismo , Ensaios de Migração Celular , Células Cultivadas , Ativação Enzimática , Ensaios Enzimáticos , Regulação Enzimológica da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos/metabolismo , Humanos , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/enzimologia , Modelos Biológicos , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Fator Estimulador de Colônias de Granulócitos/genética , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo
18.
Stem Cell Res Ther ; 3(1): 6, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22333342

RESUMO

INTRODUCTION: Mesenchymal stem cells (MSCs) are multipotent cells able to differentiate into several mesenchymal lineages, classically derived from bone marrow (BM) but potentially from umbilical cord blood (UCB). Although they are becoming a good tool for regenerative medicine, they usually need to be expanded in fetal bovine serum (FBS)-supplemented media. Human platelet lysate (HPL) has recently been proposed as substitute for safety reasons, but it is not yet clear how this supplement influences the properties of expanded MSCs. METHODS: In the present study, we compared the effect of various media combining autologous HPL with or without FBS on phenotypic, proliferative and functional (differentiation, cytokine secretion profile) characteristics of human BM-derived MSCs. RESULTS: Despite less expression of adipogenic and osteogenic markers, MSCs cultured in HPL-supplemented media fully differentiated along osteoblastic, adipogenic, chondrogenic and vascular smooth muscle lineages. The analyses of particular specific proteins expressed during osteogenic differentiation (calcium-sensing receptor (CaSR) and parathormone receptor (PTHR)) showed their decrease at D0 before any induction for MSC cultured with HPL mostly at high percentage (10%HPL). The cytokine dosage showed a clear increase of proliferation capacity and interleukin (IL)-6 and IL-8 secretion. CONCLUSIONS: This study shows that MSCs can be expanded in media supplemented with HPL that can totally replace FBS. HPL-supplemented media not only preserves their phenotype as well as their differentiation capacity, but also shortens culture time by increasing their growth rate.


Assuntos
Plaquetas/citologia , Extratos Celulares/farmacologia , Células-Tronco Mesenquimais/citologia , Animais , Células da Medula Óssea/citologia , Bovinos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura/farmacologia , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Fenótipo , Receptores de Detecção de Cálcio/metabolismo , Receptores de Hormônios Paratireóideos/metabolismo
19.
Stem Cell Rev Rep ; 7(4): 847-59, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21472453

RESUMO

Engineering living, multilayered blood vessels to form in vivo arteries is a promising alternative to peripheral artery bypass using acellular grafts restricted by thrombosis and occlusion at long term. Bone Morphogenetic Protein 2 (BMP2) is a growth factor determining in the early vascular embryonic development. The aim of the present study was evaluate the collaborative effect of recombinant human--BMP2 and Bone marrow--Mesenchymal stem cells (BM-MSCs) seeded on vascular patch to regenerate a vascular arterial wall in a rat model. BM-MSCs expressing green fluorescent protein (GFP) seeded on vascular patch were cultured in presence of recombinant human-BMP2 [100 ng/mL] during 1 week before their implantation on the abdominal aorta of Wistar rats. We observed after 2 weeks under physiological arterial flow a regeneration of a three layers adult-like arterial wall with a middle layer expressing smooth muscle proteins and a border layer expressing endothelial marker. In vitro study, using Matrigel assay and co-culture of BM-MSCs with endothelial cells demonstrated that rh-BMP2 promoted tube-like formation even at long term (90 days) allowing the organization of thick rails. We demonstrated using inhibitors and siRNAs that rh-BMP2 enhanced the expression of HIF-1α and Id1 through, at least in part, the stimulation of JAK2/STAT3/STAT5 signaling pathways. Rh-BMP2 by mimicking embryological conditions allowed vascular BM-MSCs differentiation.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteína 1 Inibidora de Diferenciação/metabolismo , Células-Tronco Mesenquimais/citologia , Regeneração , Animais , Aorta Abdominal/citologia , Aorta Abdominal/metabolismo , Aorta Abdominal/transplante , Biomarcadores/metabolismo , Prótese Vascular , Diferenciação Celular , Técnicas de Cocultura , Proteínas do Citoesqueleto/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Proteína 1 Inibidora de Diferenciação/genética , Janus Quinases/genética , Janus Quinases/metabolismo , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos dos fármacos , Modelos Animais , Proteínas Musculares/metabolismo , Neovascularização Fisiológica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Fatores de Tempo , Transfecção/métodos
20.
HIV AIDS (Auckl) ; 2: 203-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-22096399

RESUMO

BACKGROUND: An interesting finding in the epidemiology of human immunodeficiency virus (HIV) infection is that certain mutations in genes coding for chemokines, and their receptors and ligands, may confer resistance or susceptibility to HIV-1 infection and acquired immunodeficiency syndrome (AIDS) progression. The mutation most frequently studied is stromal cell-derived factor (SDF)1-3'A, a single nucleotide polymorphism in the 3' untranslated region at the 801 position of the SDF1 gene, which seems to be associated with susceptibility or resistance to diseases, including AIDS. We examined the frequency of the above polymorphisms in the Tunisian population, and evaluated their contribution to a protective genetic background against HIV infection and progression. METHODS AND MATERIALS: One hundred forty blood samples from HIV-infected patients from the Cellular Immunology Research Laboratory at the National Blood Transfusion Center were compared with those of 164 random blood donors from the same center. Genotyping was initially performed by polymerase chain reaction (PCR) analysis. SDF1 PCR product genomic regions were further subjected to restriction fragment length polymorphism analysis for genotype determination. Screening for the SDF1 polymorphism in the HIV-infected population yielded 56 heterozygous (40%), 52 mutation homozygous (37.1%), and 32 wild-type homozygous (22.8%) subjects. In contrast, in our healthy population, we found 70/164 heterozygous (42.6%), nine mutation homozygous (5.4%), and 85 wild-type homozygous (51.8%) subjects. The allele frequencies in the HIV-infected and healthy populations were f(SD1 3'A) = 57.1%, f(SDF1) = 42.8%, f(SDF1 3'A) = 26.8%, and f(SDF1) = 73.1%, respectively. The allelic and genotypic frequencies of the SDF1 3'A in our population show significantly higher distribution profiles compared with those observed in other Caucasian, European, and African American populations. Our results were examined by χ(2) test and appear to confirm an association between polymorphism and AIDS progression. A higher odds ratio (>1) was found for the SDF1-3'A allele than for the wild-type allele (<1). CONCLUSION: This result seems to confirm that the SDF1-3'A allele is associated with acceleration and progression from HIV infection to AIDS in the Tunisian population.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...