Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Stem Cells ; 37(10): 1357-1368, 2019 10.
Article in English | MEDLINE | ID: mdl-31184411

ABSTRACT

Mesenchymal stromal cells (MSC) may exert their functions by the release of extracellular vesicles (EV). Our aim was to analyze changes induced in CD34+ cells after the incorporation of MSC-EV. MSC-EV were characterized by flow cytometry (FC), Western blot, electron microscopy, and nanoparticle tracking analysis. EV incorporation into CD34+ cells was confirmed by FC and confocal microscopy, and then reverse transcription polymerase chain reaction and arrays were performed in modified CD34+ cells. Apoptosis and cell cycle were also evaluated by FC, phosphorylation of signal activator of transcription 5 (STAT5) by WES Simple, and clonal growth by clonogenic assays. Human engraftment was analyzed 4 weeks after CD34+ cell transplantation in nonobese diabetic/severe combined immunodeficient mice. Our results showed that MSC-EV incorporation induced a downregulation of proapoptotic genes, an overexpression of genes involved in colony formation, and an activation of the Janus kinase (JAK)-STAT pathway in CD34+ cells. A significant decrease in apoptosis and an increased CD44 expression were confirmed by FC, and increased levels of phospho-STAT5 were confirmed by WES Simple in CD34+ cells with MSC-EV. In addition, these cells displayed a higher colony-forming unit granulocyte/macrophage clonogenic potential. Finally, the in vivo bone marrow lodging ability of human CD34+ cells with MSC-EV was significantly increased in the injected femurs. In summary, the incorporation of MSC-EV induces genomic and functional changes in CD34+ cells, increasing their clonogenic capacity and their bone marrow lodging ability. Stem Cells 2019;37:1357-1368.


Subject(s)
Antigens, CD34/metabolism , Bone Marrow Cells/metabolism , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Humans , Mice
2.
Br J Haematol ; 187(1): 93-104, 2019 10.
Article in English | MEDLINE | ID: mdl-31172513

ABSTRACT

Patients with low-risk myelodysplastic syndromes (MDS) usually develop iron overload. This leads to a high level of oxidative stress in the bone marrow (BM) and increases haematopoietic cell dysfunction. Our objective was to analyse whether chelation with deferasirox (DFX) alleviates the consequences of oxidative stress and improves BM cell functionality. We analysed 13 iron-overloaded MDS patients' samples before and 4-10 months after treatment with DFX. Using multiparametric flow cytometry analysis, we measured intracellular reactive oxygen species (ROS), DNA oxidation and double strand breaks. Haematopoietic differentiation capacity was analysed by colony-forming unit (CFU) assays. Compared to healthy donors, MDS showed a higher level of intracellular ROS and DNA oxidative damage in BM cells. DNA oxidative damage decreased following DFX treatment. Furthermore, the clonogenic assays carried out before treatment suggest an impaired haematopoietic differentiation. DFX seems to improve this capacity, as illustrated by a decreased cluster/CFU ratio, which reached values similar to controls. We conclude that BM cells from MDS are subject to higher oxidative stress conditions and show an impaired haematopoietic differentiation. These adverse features seem to be partially rectified after DFX treatment.


Subject(s)
DNA Damage/drug effects , Deferasirox/therapeutic use , Iron Chelating Agents/therapeutic use , Myelodysplastic Syndromes/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Case-Control Studies , Cell Differentiation/drug effects , Cell Differentiation/physiology , Deferasirox/pharmacology , Humans , Iron Chelating Agents/pharmacology , Iron Overload/drug therapy , Iron Overload/etiology , Iron Overload/genetics , Iron Overload/metabolism , Middle Aged , Myelodysplastic Syndromes/complications , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Oxidative Stress/physiology , Prospective Studies , Reactive Oxygen Species/metabolism , Stem Cells/drug effects , Stem Cells/physiology , Young Adult
3.
Br J Haematol ; 181(3): 350-359, 2018 05.
Article in English | MEDLINE | ID: mdl-29611196

ABSTRACT

Treatment with azacitidine (AZA) has been suggested to be of benefit for higher-risk myelodysplastic syndrome (HR-MDS) patients with chromosome 7 abnormalities (Abn 7). This retrospective study of 235 HR-MDS patients with Abn 7 treated with AZA (n = 115) versus best supportive care (BSC; n = 120), assessed AZA treatment as a time-varying variable in multivariable analysis. A Cox Regression model with time-interaction terms of overall survival (OS) at different time points confirmed that, while chromosome 7 cytogenetic categories (complex karyotype [CK] versus non-CK) and International Prognostic Scoring System risk (high versus intermediate-2) retained poor prognosis over time, AZA treatment had a favourable impact on OS during the first 3 years of treatment compared to BSC (Hazard ratio [HR] 0·5 P < 0·001 at 1 year, 0·7 P = 0·019 at 2 years; 0·73 P = 0·029 at 3 years). This benefit was present in all chromosome 7 categories, but tended to be greater in patients with CK (risk reduction of 82%, 68% and 53% at 1, 3 and 6 months in CK patients; 79% at 1 month in non-CK patients, P < 0·05 for all). AZA also significantly improved progression-free survival (P < 0·01). This study confirms a time-dependent benefit of AZA on outcome in patients with HR-MDS and cytogenetic abnormalities involving chromosome 7, especially for those with CK.


Subject(s)
Azacitidine/administration & dosage , Chromosomes, Human, Pair 7/genetics , Myelodysplastic Syndromes , Registries , Aged , Chromosome Aberrations , Disease-Free Survival , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , Retrospective Studies , Risk Factors , Survival Rate
4.
PLoS One ; 12(8): e0182470, 2017.
Article in English | MEDLINE | ID: mdl-28796790

ABSTRACT

There is evidence of continuous bidirectional cross-talk between malignant cells and bone marrow-derived mesenchymal stromal cells (BM-MSC), which favors the emergence and progression of myeloproliferative neoplastic (MPN) diseases. In the current work we have compared the function and gene expression profile of BM-MSC from healthy donors (HD-MSC) and patients with MPN (JAK2V617F), showing no differences in the morphology, proliferation and differentiation capacity between both groups. However, BM-MSC from MPN expressed higher mean fluorescence intensity (MIF) of CD73, CD44 and CD90, whereas CD105 was lower when compared to controls. Gene expression profile of BM-MSC showed a total of 169 genes that were differentially expressed in BM-MSC from MPN patients compared to HD-MSC. In addition, we studied the ability of BM-MSC to support the growth and survival of hematopoietic stem/progenitor cells (HSPC), showing a significant increase in the number of CFU-GM colonies when MPN-HSPC were co-cultured with MPN-MSC. Furthermore, MPN-MSC showed alteration in the expression of genes associated to the maintenance of hematopoiesis, with an overexpression of SPP1 and NF-kB, and a downregulation of ANGPT1 and THPO. Our results suggest that BM-MSC from JAK2+ patients differ from their normal counterparts and favor the maintenance of malignant clonal hematopoietic cells.


Subject(s)
Hematologic Neoplasms/pathology , Janus Kinase 2/metabolism , Mesenchymal Stem Cells/physiology , Adult , Aged , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle , Cell Line, Tumor , Coculture Techniques , Female , Gene Expression , Hematologic Neoplasms/blood , Hematologic Neoplasms/enzymology , Hematopoiesis , Humans , Male , Middle Aged
5.
Am J Hematol ; 92(9): E534-E541, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28612357

ABSTRACT

The International Prognostic Scoring System and its revised form (IPSS-R) are the most widely used indices for prognostic assessment of patients with myelodysplastic syndromes (MDS), but can only partially account for the observed variation in patient outcomes. This study aimed to evaluate the relative contribution of patient condition and mutational status in peripheral blood when added to the IPSS-R, for estimating overall survival and the risk of leukemic transformation in patients with MDS. A prospective cohort (2006-2015) of 200 consecutive patients with MDS were included in the study series and categorized according to the IPSS-R. Patients were further stratified according to patient condition (assessed using the multidimensional Lee index for older adults) and genetic mutations (peripheral blood samples screened using next-generation sequencing). The change in likelihood-ratio was tested in Cox models after adding individual covariates. The addition of the Lee index to the IPSS-R significantly improved prediction of overall survival [hazard ratio (HR) 3.02, 95% confidence interval (CI) 1.96-4.66, P < 0.001), and mutational analysis significantly improved prediction of leukemic evolution (HR 2.64, 1.56-4.46, P < 0.001). Non-leukemic death was strongly linked to patient condition (HR 2.71, 1.72-4.25, P < 0.001), but not to IPSS-R score (P = 0.35) or mutational status (P = 0.75). Adjustment for exposure to disease-modifying therapy, evaluated as a time-dependent covariate, had no effect on the proposed model's predictive ability. In conclusion, patient condition, assessed by the multidimensional Lee index and patient mutational status can improve the prediction of clinical outcomes of patients with MDS already stratified by IPSS-R.


Subject(s)
Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , Survival Rate , Disease-Free Survival , Female , Follow-Up Studies , Humans , Male , Prospective Studies , Spain/epidemiology
6.
J Hematol Oncol ; 10(1): 131, 2017 06 26.
Article in English | MEDLINE | ID: mdl-28651604

ABSTRACT

BACKGROUND: Particularly since the advent of lenalidomide, lower-risk myelodysplastic syndromes (MDS) patients with del(5q) have been the focus of many studies; however, the impact of age on disease characteristics and response to lenalidomide has not been analyzed. METHODS: We assessed the effect of age on clinical characteristics and outcomes in 286 lenalidomide-treated MDS patients with del(5q) from two multicenter trials. RESULTS: A total of 33.9, 34.3, and 31.8% patients were aged <65 years, ≥65 to <75 years, and ≥75 years, respectively. Age <65 years was associated with less favorable International Prognostic Scoring System (IPSS) risk and additional cytopenias at baseline versus older age groups, significantly lower cytogenetic response rates (p = 0.022 vs. ≥65 to <75 years; p = 0.047 vs. ≥75 years), and higher rates of acute myeloid leukemia (AML) progression (Gray's test, p = 0.013). Lenalidomide was equally well tolerated across age groups, producing consistently high rates of red blood cell transfusion independence ≥26 weeks. CONCLUSIONS: Baseline disease characteristics and AML progression appear to be more severe in younger lower-risk MDS patients with del(5q), whereas older age does not seem to compromise the response to lenalidomide. TRIAL REGISTRATION: ClinicalTrials.gov NCT00065156 and NCT00179621.


Subject(s)
Immunologic Factors/therapeutic use , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/pathology , Thalidomide/analogs & derivatives , Adult , Age Factors , Aged , Aged, 80 and over , Chromosomes, Human, Pair 5/genetics , Cytogenetics , Disease Progression , Erythrocyte Transfusion , Female , Humans , Lenalidomide , Leukemia, Myeloid, Acute/etiology , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Prognosis , Risk Factors , Thalidomide/therapeutic use , Treatment Outcome
7.
Oncotarget ; 8(17): 28187-28202, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28390197

ABSTRACT

Histone deacetylases (HDACs) are involved in epigenetic modulation and their aberrant expression has been demonstrated in myeloproliferative neoplasms (MPN). HDAC8 inhibition has been shown to inhibit JAK2/STAT5 signaling in hematopoietic cells from MPN. Nevertheless, the role of HDAC8 expression in bone marrow-mesenchymal stromal cells (BM-MSC) has not been assessed. In the current work we describe that HDAC8 is significantly over-expressed in MSC from in JAK-2 positive MPN compared to those from healthy-donors (HD-MSC). Using a selective HDAC8 inhibitor (PCI34051), we verified that the subsequent decrease in the protein and mRNA expression of HDAC8 is linked with an increased apoptosis of malignant MSC whereas it has no effects on normal MSC. In addition, HDAC8 inhibition in MPN-MSC also decreased their capacity to maintain neoplastic hematopoiesis, by increasing the apoptosis, cell-cycle arrest and colony formation of JAK2+-hematopoietic cells. Mechanistic studies using different MPN cell lines revealed that PCI34051 induced their apoptosis, which is enhanced when were co-cultured with JAK2V617F-MSC, decreased their colony formation and the phosphorylation of STAT3 and STAT5. In summary, we show for the first time that the inhibition of HDAC8 in MSC from JAK2+ MPN patients selectively decreases their hematopoietic-supporting ability, suggesting that HDAC8 may be a potential therapeutic target in this setting by acting not only on hematopoietic cells but also on the malignant microenvironment.


Subject(s)
Histone Deacetylases/genetics , Janus Kinase 2/metabolism , Mesenchymal Stem Cells/metabolism , Myeloproliferative Disorders/genetics , Repressor Proteins/genetics , Apoptosis/drug effects , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Gene Expression , Hematopoiesis/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Janus Kinase 2/genetics , Mesenchymal Stem Cells/drug effects , Molecular Targeted Therapy , Mutation , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism
8.
Leuk Res ; 56: 82-87, 2017 05.
Article in English | MEDLINE | ID: mdl-28222336

ABSTRACT

Our study aimed to analyze the presence of mutations in SF3B1 and other spliceosome-related genes in myelodysplastic syndromes with ringed sideroblasts (MDS-RS) by combining conventional Sanger and next-generation sequencing (NGS) methods, and to determine the feasibility of this approach in a clinical setting. 122 bone marrow samples from MDS-RS patients were studied. Initially, exons 14 and 15 of the SF3B1 gene were analyzed by Sanger sequencing. Secondly, they were studied by NGS covering besides SF3B1, SRSF2, U2AF1 and ZRSR2 genes. An 86% of all patients showed mutations in the SF3B1 gene. Six of them, which were not identifiable by conventional sequencing in the first diagnostic step, were revealed by NGS. In addition, 19.5% of cases showed mutations in other splicing genes: SRSF2, U2AF1, and ZRSR2. Furthermore, 8.7% of patients had two mutations in SF3B1, SF3B1 and SRSF2, and SF3B1 and U2AF1, while 5.7% showed no mutations in the four spliceosome-related genes analyzed. The combined use of conventional Sanger and NGS allows the identification of mutations in spliceosome-related genes in almost all MDS patients with RS. This two-step approach is affordable and could be useful as a complementary technique in cases with an unclear diagnosis.


Subject(s)
Anemia, Sideroblastic/genetics , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Spliceosomes/genetics , Anemia, Sideroblastic/diagnosis , Bone Marrow , Humans , Methods , Mutation , Phosphoproteins/genetics , RNA Splicing Factors/genetics , Sequence Analysis, DNA , Serine-Arginine Splicing Factors/genetics , Splicing Factor U2AF/genetics
9.
BMC Genomics ; 17(1): 944, 2016 Nov 21.
Article in English | MEDLINE | ID: mdl-27871224

ABSTRACT

BACKGROUND: Mesenchymal Stromal/Stem Cells (MSCs), isolated under the criteria established by the ISCT, still have a poorly characterized phenotype that is difficult to distinguish from similar cell populations. Although the field of transcriptomics and functional genomics has quickly grown in the last decade, a deep comparative analysis of human MSCs expression profiles in a meaningful cellular context has not been yet performed. There is also a need to find a well-defined MSCs gene-signature because many recent biomedical studies show that key cellular interaction processes (i.e. inmuno-modulation, cellular cross-talk, cellular maintenance, differentiation, epithelial-mesenchymal transition) are dependent on the mesenchymal stem cells within the stromal niche. RESULTS: In this work we define a core mesenchymal lineage signature of 489 genes based on a deep comparative analysis of multiple transcriptomic expression data series that comprise: (i) MSCs of different tissue origins; (ii) MSCs in different states of commitment; (iii) other related non-mesenchymal human cell types. The work integrates several public datasets, as well as de-novo produced microarray and RNA-Seq datasets. The results present tissue-specific signatures for adipose tissue, chorionic placenta, and bone marrow MSCs, as well as for dermal fibroblasts; providing a better definition of the relationship between fibroblasts and MSCs. Finally, novel CD marker patterns and cytokine-receptor profiles are unravelled, especially for BM-MSCs; with MCAM (CD146) revealed as a prevalent marker in this subtype of MSCs. CONCLUSIONS: The improved biomolecular characterization and the released genome-wide expression signatures of human MSCs provide a comprehensive new resource that can drive further functional studies and redesigned cell therapy applications.


Subject(s)
Mesenchymal Stem Cells/metabolism , Transcriptome , Adipose Tissue/cytology , Biomarkers , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cluster Analysis , Computational Biology/methods , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Humans , Mesenchymal Stem Cells/cytology , Organ Specificity/genetics , Placenta/cytology , Pregnancy
10.
J Hematol Oncol ; 9(1): 113, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27765055

ABSTRACT

BACKGROUND: Graft-versus-host disease (GvHD) remains the major obstacle to successful allogeneic hematopoietic stem cell transplantation, despite of the immunosuppressive regimens administered to control T cell alloreactivity. PI3K/AKT/mTOR pathway is crucial in T cell activation and function and, therefore, represents an attractive therapeutic target to prevent GvHD development. Recently, numerous PI3K inhibitors have been developed for cancer therapy. However, few studies have explored their immunosuppressive effect. METHODS: The effects of a selective PI3K inhibitor (BKM120) and a dual PI3K/mTOR inhibitor (BEZ235) on human T cell proliferation, expression of activation-related molecules, and phosphorylation of PI3K/AKT/mTOR pathway proteins were analyzed. Besides, the ability of BEZ235 to prevent GvHD development in mice was evaluated. RESULTS: Simultaneous inhibition of PI3K and mTOR was efficient at lower concentrations than PI3K specific targeting. Importantly, BEZ235 prevented naïve T cell activation and induced tolerance of alloreactive T cells, while maintaining an adequate response against cytomegalovirus, more efficiently than BKM120. Finally, BEZ235 treatment significantly improved the survival and decreased the GvHD development in mice. CONCLUSIONS: These results support the use of PI3K inhibitors to control T cell responses and show the potential utility of the dual PI3K/mTOR inhibitor BEZ235 in GvHD prophylaxis.


Subject(s)
Graft vs Host Disease/prevention & control , Lymphocyte Activation/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Animals , Cell Proliferation/drug effects , Imidazoles/pharmacology , Imidazoles/therapeutic use , Mice , Morpholines/pharmacology , Morpholines/therapeutic use , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Quinolines/pharmacology , Quinolines/therapeutic use , Signal Transduction/drug effects , T-Lymphocytes/immunology , TOR Serine-Threonine Kinases/antagonists & inhibitors
11.
Cytotherapy ; 18(10): 1297-311, 2016 10.
Article in English | MEDLINE | ID: mdl-27637760

ABSTRACT

BACKGROUND AIMS: The immunomodulatory properties of mesenchymal stromal cells (MSCs), together with their tissue regenerative potential, make them interesting candidates for clinical application. METHODS: In the current study, we analyzed the in vitro immunomodulatory effects of MSCs derived from bone marrow (BM-MSCs) and from adipose tissue (AT-MSCs) obtained from the same donor on both innate and acquired immunity cells. BM-MSCs and AT-MSCs were expanded to fourth or fifth passage and co-cultured with T cells, monocytes or natural killer (NK) cells isolated from human peripheral blood and stimulated in vitro. The possible differing impact of MSCs obtained from distinct sources on phenotype, cell proliferation and differentiation, cytokine production and function of these immune cells was comparatively analyzed. RESULTS: BM-MSCs and AT-MSCs induced a similar decrease in NK-cell proliferation, cytokine secretion and expression of both activating receptors and cytotoxic molecules. However, only BM-MSCs significantly reduced NK-cell cytotoxic activity, although both MSC populations showed the same susceptibility to NK-cell-mediated lysis. AT-MSCs were more potent in inhibiting dendritic-cell (DC) differentiation than BM-MSC, but both MSC populations similarly reduced the ability of DCs to induce CD4(+) T-cell proliferation and cytokine production. BM-MSCs and AT-MSCs induced a similar decrease in T-cell proliferation and production of inflammatory cytokines after activation. CONCLUSIONS: AT-MSCs and BM-MSCs from the same donor had similar immunomodulatory capacity on both innate and acquired immunity cells. Thus, other variables, such as accessibility of samples or the frequency of MSCs in the tissue should be considered to select the source of MSC for cell therapy.


Subject(s)
Adipose Tissue/cytology , Bone Marrow Cells/physiology , Immunomodulation/physiology , Mesenchymal Stem Cells/physiology , T-Lymphocytes/immunology , Adult , Aged , Bone Marrow Cells/cytology , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic , Female , Humans , Killer Cells, Natural/immunology , Lymphocyte Activation , Male , Mesenchymal Stem Cells/cytology , Middle Aged , Tissue Donors
12.
J Clin Oncol ; 34(25): 2988-96, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27354480

ABSTRACT

PURPOSE: This international phase III, randomized, placebo-controlled, double-blind study assessed the efficacy and safety of lenalidomide in RBC transfusion-dependent patients with International Prognostic Scoring System lower-risk non-del(5q) myelodysplastic syndromes ineligible for or refractory to erythropoiesis-stimulating agents. PATIENTS AND METHODS: In total, 239 patients were randomly assigned (2:1) to treatment with lenalidomide (n = 160) or placebo (n = 79) once per day (on 28-day cycles). The primary end point was the rate of RBC transfusion independence (TI) ≥ 8 weeks. Secondary end points were RBC-TI ≥ 24 weeks, duration of RBC-TI, erythroid response, health-related quality of life (HRQoL), and safety. RESULTS: RBC-TI ≥ 8 weeks was achieved in 26.9% and 2.5% of patients in the lenalidomide and placebo groups, respectively (P < .001). Ninety percent of patients achieving RBC-TI responded within 16 weeks of treatment. Median duration of RBC-TI with lenalidomide was 30.9 weeks (95% CI, 20.7 to 59.1). Transfusion reduction of ≥ 4 units packed RBCs, on the basis of a 112-day assessment, was 21.8% in the lenalidomide group and 0% in the placebo group. Higher response rates were observed in patients with lower baseline endogenous erythropoietin ≤ 500 mU/mL (34.0% v 15.5% for > 500 mU/mL). At week 12, mean changes in HRQoL scores from baseline did not differ significantly between treatment groups, which suggests that lenalidomide did not adversely affect HRQoL. Achievement of RBC-TI ≥ 8 weeks was associated with significant improvements in HRQoL (P < .01). The most common treatment-emergent adverse events were neutropenia and thrombocytopenia. CONCLUSION: Lenalidomide yields sustained RBC-TI in 26.9% of RBC transfusion-dependent patients with lower-risk non-del(5q) myelodysplastic syndromes ineligible for or refractory to erythropoiesis-stimulating agents. Response to lenalidomide was associated with improved HRQoL. Treatment-emergent adverse event data were consistent with the known safety profile of lenalidomide.


Subject(s)
Erythrocyte Transfusion , Myelodysplastic Syndromes/therapy , Thalidomide/analogs & derivatives , Adult , Aged , Aged, 80 and over , DNA Mutational Analysis , Double-Blind Method , Drug Resistance , Erythropoiesis/drug effects , Female , Gene Expression , Humans , Lenalidomide , Male , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Quality of Life , Thalidomide/adverse effects , Thalidomide/therapeutic use
13.
Oncotarget ; 7(21): 30492-503, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27127180

ABSTRACT

The biological and molecular events that underlie bone marrow fibrosis in patients with myelodysplastic syndromes are poorly understood, and its prognostic role in the era of the Revised International Prognostic Scoring System (IPSS-R) is not yet fully determined. We have evaluated the clinical and biological events that underlie bone marrow fibrotic changes, as well as its prognostic role, in a well-characterized prospective patient cohort (n=77) of primary MDS patients. The degree of marrow fibrosis was linked to parameters of erythropoietic failure, marrow cellularity, p53 protein accumulation, WT1 gene expression, and serum levels of CXCL9 and CXCL10, but not to other covariates including the IPSS-R score. The presence of bone marrow fibrosis grade 2 or higher was associated with the presence of mutations in cohesin complex genes (31.5% vs. 5.4%, p=0.006). By contrast, mutations in CALR, JAK2, PDGFRA, PDGFRB,and TP53 were very rare. Survival analysis showed that marrow fibrosis grade 2 or higher was a relevant significant predictor for of overall survival, and independent of age, performance status, and IPSS-R score in multivariate analysis.


Subject(s)
Bone Marrow/metabolism , DNA Mutational Analysis/methods , Mutation , Myelodysplastic Syndromes/genetics , Adult , Aged , Aged, 80 and over , Bone Marrow/pathology , Chemokine CXCL10/blood , Chemokine CXCL9/blood , Female , Fibrosis , Humans , Male , Middle Aged , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Prognosis , Prospective Studies , Survival Analysis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism
14.
Eur J Haematol ; 97(6): 528-537, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27118602

ABSTRACT

INTRODUCTION: The ability of mesenchymal stromal cells (MSC) to suppress T-cell function has prompted their therapeutic use for graft-versus-host disease (GVHD) control. However, as MSC also modulate the activity of NK cells, which play an important role in graft-versus-leukemia (GVL) reaction, their administration could hamper this beneficial effect of allogeneic hematopoietic stem cell transplantation. MSC can be expanded from several sources, especially bone marrow and fat, but it is not well established if the cell source makes a difference in their immunoregulatory capacity. OBJECTIVE: The aim of this study was to compare the immunomodulatory effect of MSC derived from bone marrow (BM-CSM) or adipose tissue (AT-MSC) on NK cells, to determine whether the use of MSC from one or the other origin could be more favorable to preserve NK cell activity and, therefore, GVL. METHODS: Human NK cells were stimulated with IL-15 in the presence of BM-MSC or AT-MSC. The effect of both MSC populations on NK cell proliferation, cell cycle progression, and CD56 expression was analyzed by flow cytometry. Cytokine secretion was measured by ELISA, and cytotoxic activity was assessed by calcein release assays. RESULTS: Although both BM-MSC and AT-MSC induced a similar inhibition of NK cell proliferation, only BM-MSC decreased significantly NK cell cytotoxic activity and showed a trend for a higher reduction of IFN-γ secretion. CONCLUSION: These results suggest that, in the context of GVHD inhibition, the use of AT-MSC rather than BM-MSC could further preserve NK cell activity and, thus, favor GVL.


Subject(s)
Adipose Tissue/cytology , Cell Communication/immunology , Immunomodulation , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mesenchymal Stem Cells/metabolism , Adult , Aged , Biomarkers , Cell Cycle , Cell Line, Tumor , Cytokines/metabolism , Cytotoxicity, Immunologic , Female , Humans , Immunophenotyping , Lymphocyte Activation , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Middle Aged , Phenotype
15.
Br J Haematol ; 173(5): 754-68, 2016 06.
Article in English | MEDLINE | ID: mdl-26914848

ABSTRACT

The mechanistic target of rapamycin (mTOR) pathway is crucial for the activation and function of T cells, which play an essential role in the development of graft-versus-host disease (GvHD). Despite its partial ability to block mTOR pathway, the mTORC1 inhibitor rapamycin has shown encouraging results in the control of GvHD. Therefore, we considered that simultaneous targeting of both mTORC1 and mTORC2 complexes could exert a more potent inhibition of T cell activation and, thus, could have utility in GvHD control. To assess this assumption, we have used the dual mTORC1/mTORC2 inhibitors CC214-1 and CC214-2. In vitro studies confirmed the superior ability of CC214-1 versus rapamycin to block mTORC1 and mTORC2 activity and to reduce T cell proliferation. Both drugs induced a similar decrease in Th1/Th2 cytokine secretion, but CC214-1 was more efficient in inhibiting naïve T cell activation and the expression of T-cell activation markers. In addition, CC214-1 induced specific tolerance against alloantigens, while preserving anti-cytomegalovirus response. Finally, in a mouse model of GvHD, the administration of CC214-2 significantly improved mice survival and decreased GvHD-induced damages. In conclusion, the current study shows, for the first time, the immunosuppressive ability of CC214-1 on T lymphocytes and illustrates the role of CC214-2 in the allogeneic transplantation setting as a possible GvHD prophylaxis agent.


Subject(s)
Graft vs Host Disease/prevention & control , Multiprotein Complexes/antagonists & inhibitors , T-Lymphocytes/immunology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cells, Cultured , Female , Graft vs Host Disease/drug therapy , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Immune Tolerance/drug effects , Lymphocyte Activation/drug effects , Male , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pyrazines/pharmacology , Pyrazines/therapeutic use , T-Lymphocytes/drug effects
16.
Cell Commun Signal ; 14: 2, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26754424

ABSTRACT

BACKGROUND: Human mesenchymal stromal cells (hMSC) are multipotent cells with both regenerative and immunomodulatory activities making them an attractive tool for cellular therapy. In the last few years it has been shown that the beneficial effects of hMSC may be due to paracrine effects and, at least in part, mediated by extracellular vesicles (EV). EV have emerged as important mediators of cell-to-cell communication. Flow cytometry (FCM) is a routine technology used in most clinical laboratories and could be used as a methodology for hMSC-EV characterization. Although several reports have characterized EV by FCM, a specific panel and protocol for hMSC-derived EV is lacking. The main objective of our study was the characterization of hMSC-EV using a standard flow cytometer. METHODS: Human MSC from bone marrow of healthy donors, mesenchymal cell lines (HS-5 and hTERT) and a leukemic cell line (K562 cells) were used to obtain EV for FCM characterization. EV released from the different cell lines were isolated by ultracentrifugation and were characterized, using a multi-parametric analysis, in a conventional flow cytometer. EV characterization by transmission electron microscopy (TEM), western blot (WB) and Nano-particle tracking analysis (NTA) was also performed. RESULTS: EV membranes are constituted by the combination of specific cell surface molecules depending on their cell of origin, together with specific proteins like tetraspanins (e.g. CD63). We have characterized by FCM the EV released from BM-hMSC, that were defined as particles less than 0.9 µm, positive for the hMSC markers (CD90, CD44 and CD73) and negative for CD34 and CD45 (hematopoietic markers). In addition, hMSC-derived EV were also positive for CD63 and CD81, the two characteristic markers of EV. To validate our characterization strategy, EV from mesenchymal cell lines (hTERT/HS-5) were also studied, using the leukemia cell line (K562) as a negative control. EV released from mesenchymal cell lines displayed the same immunophenotypic profile as the EV from primary BM-hMSC, while the EV derived from K562 cells did not show hMSC markers. We further validated the panel using EV from hMSC transduced with GFP. Finally, EV derived from the different sources (hMSC, hTERT/HS-5 and K562) were also characterized by WB, TEM and NTA, demonstrating the expression by WB of the exosomal markers CD63 and CD81, as well as CD73 in those from MSC origin. EV morphology and size/concentration was confirmed by TEM and NTA, respectively. CONCLUSION: We described a strategy that allows the identification and characterization by flow cytometry of hMSC-derived EV that can be routinely used in most laboratories with a standard flow cytometry facility.


Subject(s)
5'-Nucleotidase/analysis , Extracellular Vesicles/chemistry , Flow Cytometry/methods , Hyaluronan Receptors/analysis , Mesenchymal Stem Cells/cytology , Thy-1 Antigens/analysis , Adult , Cell Line , Cells, Cultured , Female , Humans , Male , Mesenchymal Stem Cells/chemistry , Middle Aged , Young Adult
17.
Lancet Haematol ; 2(6): e260-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26688236

ABSTRACT

BACKGROUND: We aimed to compare the ability of recently developed prognostic indices for myelodysplastic syndromes to identify patients with poor prognoses within the lower-risk (low and intermediate-1) categories defined by the International Prognosis Scoring System (IPSS). METHODS: We included patients with de-novo myelodysplastic syndromes diagnosed between Nov 29, 1972, and Dec 15, 2011, who had low or intermediate-1 IPSS scores and were in the Spanish Registry of Myelodysplastic Syndromes. We reclassified these patients with the new prognostic indices (revised IPSS [IPSS-R], revised WHO-based Prognostic Scoring System [WPSS-R], Lower Risk Scoring System [LRSS], and the Grupo Español de Síndromes Mielodisplásicos [Spanish Group of Myelodysplastic Syndromes; GESMD]) and calculated the overall survival of the different risk groups within each prognostic index to identify the groups of patients with overall poor prognoses (defined as an expected overall survival <30 months). We calculated overall survival with the Kaplan-Meier method. FINDINGS: We identified 2373 patients. None of the prognostic indices could be used to identify a population with poor prognoses (median overall survival <30 months) for the patients with low IPSS scores (1290 individuals). In the group with intermediate-1 scores (1083 individuals), between 17% and 47% of patients were identified as having poor prognoses with the new prognostic indices. The LRSS had the best model fit with the lowest value in the Akaike information criteria test, whereas the IPSS-R identified the largest proportion of patients with poor prognoses (47%). Patients with intermediate-1 scores who were classified as having poor prognoses by one or more prognostic index (646 [60%] individuals) had worse median overall survival (33·1 months, 95% CI 28·4-37·9) than did patients who were classified as having low risk by all prognostic indices (63·7 months, 49·5-78·0], HR 1·9, 95% CI 1·6-2·3, p<0·0001) INTERPRETATION: Recently proposed prognostic indices for myelodysplastic syndromes can be used to improve identification of patients with poor prognoses in the group of patients with intermediate-1 IPSS scores, who could potentially benefit from a high-risk treatment approach. FUNDING: None.


Subject(s)
Myelodysplastic Syndromes/diagnosis , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/classification , Prognosis , Registries , Retrospective Studies , Risk Factors , Survival Analysis
18.
Clin Case Rep ; 3(6): 370-5, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26185631

ABSTRACT

Sixty-seven-year-old patient, diagnosed with multiple myeloma who had received autologous stem cell transplantation, following bortezomib, dexamethasone and thalidomide conventional regimen, achieving complete response, developed rash, diarrhea, and severe respiratory failure, 80 days after the transplantation procedure. He was diagnosed with graft-versus-host disease and bronchiolitis obliterans syndrome.

19.
Leuk Res ; 39(8): 921-4, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26105049

ABSTRACT

The expression of BCR-ABL in hematopoietic stem cells is a well-defined primary event in chronic myeloid leukemia (CML). Some reports have described the presence of BCR-ABL on endothelial cells from CML patients, suggesting the origin of the disease in a primitive hemangioblastic cell. On the other hand, extracellular vesicles (EVs) released by CML leukemic cells are involved in the angiogenesis modulation process. In the current work we hypothesized that EVs released from BCR-ABL(+) cells may carry inside the oncogene that can be transferred to endothelial cells leading to the expression of both BCR-ABL transcript and the oncoprotein. EVs from K562 cells and plasma of newly diagnosed CML patients were isolated by ultracentrifugation. RT-PCR analysis detected the presence of BCR-ABL RNA in the EVs isolated from both K562 cells and plasma of CML patients. The incorporation of these EVs into endothelial cells was demonstrated by flow cytometry and fluorescence microscopy showed that after 24h of incubation most EVs were incorporated. BCR-ABL transcripts were detected in all experiments on endothelial cells incubated with EVs from both sources. The presence of BCR-ABL on endothelial cells incubated with Philadelphia(+) EVs was also confirmed by Western blot assays. In summary, endothelial cells acquire BCR-ABL RNA and the oncoprotein after incubation with EVs released from Ph(+) positive cells (either from K562 cells or from plasma of newly diagnosed CML patients). This results challenge the hypothesis that endothelial cells may be part of the Philadelphia(+) clone in CML.


Subject(s)
Hematopoietic Stem Cells/physiology , Human Umbilical Vein Endothelial Cells/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/pathology , Secretory Vesicles/physiology , Cells, Cultured , Clone Cells/metabolism , Clone Cells/pathology , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Hematopoietic Stem Cells/pathology , Human Umbilical Vein Endothelial Cells/pathology , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
20.
Cell Transplant ; 24(12): 2423-33, 2015.
Article in English | MEDLINE | ID: mdl-25695936

ABSTRACT

Clinical trials have assessed the use of human bone marrow stromal cells (hBMSCs) for the treatment of immune-related disorders such as graft-versus-host disease (GVHD). In the current study, we show that GFP(+)-transduced hBMSCs generated from bone marrow migrate and differentiate into corneal tissue after subconjunctival injection in mice. Interestingly, these hBMSCs display morphological features of epithelial, stromal, and endothelial cells and appear at different layers and with different morphologies depending on their position within the epithelium. Furthermore, these cells display ultrastructural properties, such as bundles of intermediate filaments, interdigitations, and desmosomes with GFP(-) cells, which confirms their differentiation into corneal tissues. GFP(+)-transduced hBMSCs were injected at different time points into the right eye of lethally irradiated mice undergoing bone marrow transplantation, which developed ocular GVHD (oGVHD). Remarkably, hBMSCs massively migrate to corneal tissues after subconjunctival injection. Both macroscopic and histopathological examination showed minimal or no evidence of GVHD in the right eye, while the left eye, where no hBMSCs were injected, displayed features of GVHD. Thus, in the current study, we confirm that hBMSCs may induce their therapeutic effect at least in part by differentiation and regeneration of damaged tissues in the host. Our results provide experimental evidence that hBMSCs represent a potential cellular therapy to attenuate oGVHD.


Subject(s)
Bone Marrow Cells/cytology , Cornea/cytology , Corneal Transplantation/adverse effects , Graft vs Host Disease/prevention & control , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Adult , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Cell- and Tissue-Based Therapy/methods , Extracellular Matrix Proteins/metabolism , Female , Graft vs Host Disease/therapy , Green Fluorescent Proteins , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...