Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Cell Mol Med ; 24(18): 10978-10986, 2020 09.
Article in English | MEDLINE | ID: mdl-32790151

ABSTRACT

Myeloproliferative neoplasms are divided into essential thrombocythemia (ET), polycythemia vera (PV) and primary myelofibrosis (PMF). Although ruxolitinib was proven to be effective in reducing symptoms, patients rarely achieve complete molecular remission. Therefore, it is relevant to identify new therapeutic targets to improve the clinical outcome of patients. Bcl-xL protein, the long isoform encoded by alternative splicing of the Bcl-x gene, acts as an anti-apoptotic regulator. Our study investigated the role of Bcl-xL as a marker of severity of MPN and the possibility to target Bcl-xL in patients. 129 MPN patients and 21 healthy patients were enrolled in the study. We analysed Bcl-xL expression in leucocytes and in enriched CD34+ and CD235a+ cells. Furthermore, ABT-737, a Bcl-xL inhibitor, was tested in HEL cells and in leucocytes from MPN patients. Bcl-xL was found progressively over-expressed in cells from ET, PV and PMF patients, independently by JAK2 mutational status. Moreover, our data indicated that the combination of ABT-737 and ruxolitinib resulted in a significantly higher apoptotic rate than the individual drug. Our study suggests that Bcl-xL plays an important role in MPN independently from JAK2 V617F mutation. Furthermore, data demonstrate that targeting simultaneously JAK2 and Bcl-xL might represent an interesting new approach.


Subject(s)
Molecular Targeted Therapy , Myeloproliferative Disorders/drug therapy , Neoplasm Proteins/antagonists & inhibitors , bcl-X Protein/antagonists & inhibitors , Alternative Splicing , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Biomarkers, Tumor , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/pharmacology , Cell Division/drug effects , Cell Line, Tumor , Drug Synergism , Hematopoietic Stem Cells/metabolism , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Leukocytes/metabolism , Mutation, Missense , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Neoplasm Proteins/genetics , Nitriles , Nitrophenols/administration & dosage , Nitrophenols/pharmacology , Philadelphia Chromosome , Piperazines/administration & dosage , Piperazines/pharmacology , Protein Isoforms/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Pyrimidines , Severity of Illness Index , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , bcl-X Protein/genetics
2.
Int J Mol Sci ; 21(20)2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33081324

ABSTRACT

Iron is crucial to satisfy several mitochondrial functions including energy metabolism and oxidative phosphorylation. Patients affected by Myelodysplastic Syndromes (MDS) and acute myeloid leukemia (AML) are frequently characterized by iron overload (IOL), due to continuous red blood cell (RBC) transfusions. This event impacts the overall survival (OS) and it is associated with increased mortality in lower-risk MDS patients. Accordingly, the oral iron chelator Deferasirox (DFX) has been reported to improve the OS and delay leukemic transformation. However, the molecular players and the biological mechanisms laying behind remain currently mostly undefined. The aim of this study has been to investigate the potential anti-leukemic effect of DFX, by functionally and molecularly analyzing its effects in three different leukemia cell lines, harboring or not p53 mutations, and in human primary cells derived from 15 MDS/AML patients. Our findings indicated that DFX can lead to apoptosis, impairment of cell growth only in a context of IOL, and can induce a significant alteration of mitochondria network, with a sharp reduction in mitochondrial activity. Moreover, through a remarkable reduction of Murine Double Minute 2 (MDM2), known to regulate the stability of p53 and p73 proteins, we observed an enhancement of p53 transcriptional activity after DFX. Interestingly, this iron depletion-triggered signaling is enabled by p73, in the absence of p53, or in the presence of a p53 mutant form. In conclusion, we propose a mechanism by which the increased p53 family transcriptional activity and protein stability could explain the potential benefits of iron chelation therapy in terms of improving OS and delaying leukemic transformation.


Subject(s)
Deferasirox/pharmacology , Iron Chelating Agents/pharmacology , Leukemia, Myeloid, Acute/metabolism , Mitochondria/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Humans , Iron/metabolism , Mitochondria/drug effects , Protein Stability , Proto-Oncogene Proteins c-mdm2/metabolism
3.
Pediatr Blood Cancer ; 66(5): e27635, 2019 05.
Article in English | MEDLINE | ID: mdl-30688024

ABSTRACT

BACKGROUND: Neuroblastoma (NB), a pediatric tumor of the sympathetic nervous system, is characterized by very frequent chromosomal aberrations at the onset of the disease. Identification of further risk factors for relapse, which could lead to increased survival and potentially reduced late effects among survivors, is still urgently needed. Segmental chromosome aberrations (SCA) are associated with poor prognosis, whereas numerical whole-chromosome aberrations (NCA) are found in patients with a good prognosis; however, a small percentage of the latter patients (10%-15%) subsequently relapse and/or die of disease. PROCEDURE: DNA copy-number data from 174 NB patients with an NCA genomic profile were analyzed. Association between NCA and event-free survival (EFS) was investigated by the Kaplan-Meier estimator and prognostic decision tree (DT). RESULTS: DT identified 65 patients with normal chromosome X and an excellent five-year EFS (100%) independently from the stage at diagnosis. The association between poor EFS and whole chromosome X alterations was confirmed after stratification into two groups of different expected prognosis and by internal validation via bootstrap analysis. Furthermore, the association was also observed in an independent cohort of NB patients extracted from the data set of the National Cancer Institute TARGET Project for Neuroblastoma, but sample size was small (n = 75) and statistical significance was not achieved. CONCLUSIONS: Loss of whole chromosome X may represent a new prognostic marker for NB patients with an NCA genomic profile. If confirmed by further studies, this finding could indicate that such patients should be reclassified as intermediate risk and treated accordingly.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, X/genetics , Genomics/methods , Neuroblastoma/genetics , Neuroblastoma/pathology , Adolescent , Child , Child, Preschool , Cohort Studies , Disease Progression , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Male , Prognosis , Survival Rate
4.
Cell Mol Life Sci ; 75(5): 889-903, 2018 03.
Article in English | MEDLINE | ID: mdl-28975370

ABSTRACT

We evaluated the energy metabolism of human mesenchymal stem cells (MSC) isolated from umbilical cord (UC) of preterm (< 37 weeks of gestational age) and term (≥ 37 weeks of gestational age) newborns, using MSC from adult bone marrow as control. A metabolic switch has been observed around the 34th week of gestational age from a prevalently anaerobic glycolysis to the oxidative phosphorylation. This metabolic change is associated with the organization of mitochondria reticulum: preterm MSCs presented a scarcely organized mitochondrial reticulum and low expression of proteins involved in the mitochondrial fission/fusion, compared to term MSCs. These changes seem governed by the expression of CLUH, a cytosolic messenger RNA-binding protein involved in the mitochondria biogenesis and distribution inside the cell; in fact, CLUH silencing in term MSC determined a metabolic fingerprint similar to that of preterm MSC. Our study discloses novel information on the production of energy and mitochondrial organization and function, during the passage from fetal to adult life, providing useful information for the management of preterm birth.


Subject(s)
Energy Metabolism/physiology , Glycolysis/physiology , Mesenchymal Stem Cells/metabolism , Oxidative Phosphorylation , Premature Birth/metabolism , Term Birth/metabolism , Anaerobiosis , Cells, Cultured , Humans , Infant, Newborn , Infant, Premature , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Umbilical Cord/metabolism
5.
Blood ; 125(26): 4095-102, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-25957389

ABSTRACT

Mechanisms of hematopoietic reconstitution after bone marrow (BM) transplantation remain largely unknown. We applied a computational quantification software application to hybrid 18F-fluorodeoxyglucose positron emission tomography (PET)/computed tomography (CT) images to assess activity and distribution of the hematopoietic system throughout the whole skeleton of recently transplanted patients. Thirty-four patients underwent PET/CT 30 days after either adult stem cell transplantation (allogeneic cell transplantation [ACT]; n = 18) or cord blood transplantation (CBT; n = 16). Our software automatically recognized compact bone volume and trabecular bone volume (IBV) in CT slices. Within IBV, coregistered PET data were extracted to identify the active BM (ABM) from the inactive tissue. Patients were compared with 34 matched controls chosen among a published normalcy database. Whole body ABM increased in ACT and CBT when compared with controls (12.4 ± 3 and 12.8 ± 6.8 vs 8.1 ± 2.6 mL/kg of ideal body weight [IBW], P < .001). In long bones, ABM increased three- and sixfold in CBT and ACT, respectively, compared with controls (0.9 ± 0.9 and 1.7 ± 2.5 vs 0.3 ± 0.3 mL/kg IBW, P < .01). These data document an unexpected distribution of transplanted BM into previously abandoned BM sites.


Subject(s)
Adult Stem Cells/transplantation , Bone Marrow Transplantation , Bone Marrow/diagnostic imaging , Cord Blood Stem Cell Transplantation , Hematopoietic Stem Cell Transplantation , Adolescent , Adult , Aged , Allografts , Female , Fluorodeoxyglucose F18 , Humans , Male , Middle Aged , Multimodal Imaging , Positron-Emission Tomography/methods , Radiopharmaceuticals , Tomography, X-Ray Computed/methods , Young Adult
6.
FASEB J ; 30(4): 1416-24, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26655706

ABSTRACT

Exosomes are secreted nanovesicles that are able to transfer RNA and proteins to target cells. The emerging role of mesenchymal stem cell (MSC) exosomes as promoters of aerobic ATP synthesis restoration in damaged cells, prompted us to assess whether they contain an extramitochondrial aerobic respiration capacity. Exosomes were isolated from culture medium of human MSCs from umbilical cord of ≥37-wk-old newborns or between 28- to 30-wk-old newborns (i.e.,term or preterm infants). Characterization of samples was conducted by cytofluorometry. Oxidative phosphorylation capacity was assessed by Western blot analysis, oximetry, and luminometric and fluorometric analyses. MSC exosomes express functional respiratory complexes I, IV, and V, consuming oxygen. ATP synthesis was only detectable in exosomes from term newborns, suggestive of a specific mechanism that is not completed at an early gestational age. Activities are outward facing and comparable to those detected in mitochondria isolated from term MSCs. MSC exosomes display an unsuspected aerobic respiratory ability independent of whole mitochondria. This may be relevant for their ability to rescue cell bioenergetics. The differential oxidative metabolism of pretermvs.term exosomes sheds new light on the preterm newborn's clinical vulnerability. A reduced ability to repair damaged tissue and an increased capability to cope with anoxic environment for preterm infants can be envisaged.-Panfoli, I., Ravera, S., Podestà, M., Cossu, C., Santucci, L., Bartolucci, M., Bruschi, M., Calzia, D., Sabatini, F., Bruschettini, M., Ramenghi, L. A., Romantsik, O., Marimpietri, D., Pistoia, V., Ghiggeri, G., Frassoni, F., Candiano, G. Exosomes from human mesenchymal stem cells conduct aerobic metabolism in term and preterm newborn infants.


Subject(s)
Energy Metabolism , Exosomes/metabolism , Infant, Premature/metabolism , Mesenchymal Stem Cells/metabolism , Term Birth/metabolism , Adenosine Triphosphate/biosynthesis , Blotting, Western , Cells, Cultured , Electron Transport Complex I/metabolism , Electron Transport Complex IV/metabolism , Humans , Infant, Newborn , Infant, Premature/blood , Mitochondrial Proton-Translocating ATPases/metabolism , Oxidative Phosphorylation , Oximetry , Oxygen Consumption , Term Birth/blood
7.
Ann Hematol ; 95(6): 967-72, 2016 May.
Article in English | MEDLINE | ID: mdl-26994010

ABSTRACT

In the context of discussions on the reproducibility of clinical studies, we reanalyzed a prospective randomized study on the role of splenic irradiation as adjunct to the conditioning for hematopoietic stem cell transplantation (HSCT) for chronic myeloid leukemia (CML). Between 1986 and 1989, a total of 229 patients with CML were randomized; of these, 225 (98 %; 112 with, 113 without splenic irradiation) could be identified in the database and their survival updated. Results confirmed the early findings with no significant differences in all measured endpoints (overall survival at 25 years: 42.7 %, 32.0-52.4 % vs 52.9 %, 43.2-62.6 %; p = 0.355, log rank test). Additional splenic irradiation failed to reduce relapse incidence. It did not increase non-relapse mortality nor the risk of late secondary malignancies. Comforting are the long-term results from this predefined consecutive cohort of patients: more than 60 % were alive at plus 25 years when they were transplanted with a low European Society for Blood and Marrow Transplantation (EBMT) risk sore. This needs to be considered today when treatment options are discussed for patients who failed initial tyrosine kinase inhibitor therapy and have an available low risk HLA-identical donor.


Subject(s)
Hematopoietic Stem Cell Transplantation/trends , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/radiotherapy , Spleen/radiation effects , Transplantation Conditioning/trends , Adolescent , Adult , Child , Cohort Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prospective Studies , Time Factors , Transplantation Conditioning/methods , Young Adult
8.
J Immunol ; 192(4): 1471-9, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24442432

ABSTRACT

NK cells are the first lymphoid population recovering after allogeneic hematopoietic stem cell transplantation and play a crucial role in early immunity after the graft. Recently, it has been shown that human CMV (HCMV) infection/reactivation can deeply influence NK cell reconstitution after umbilical cord blood transplantation by accelerating the differentiation of mature NKG2A(-) killer Ig-like receptor (KIR)(+) NK cells characterized by the expression of the NKG2C-activating receptor. In view of the hypothesis that NKG2C could be directly involved in NK cell maturation driven by HCMV infection, we analyzed the maturation and function of NK cells developing in three patients with hematological malignancies given umbilical cord blood transplantation from donors carrying a homozygous deletion of the NKG2C gene. We show that HCMV infection can drive rapid NK maturation, characterized by the expansion of CD56(dim)NKG2A(-)KIR(+) cells, even in the absence of NKG2C expression. Interestingly, this expanded mature NK cell subset expressed surface-activating KIR that could trigger NK cell cytotoxicity, degranulation, and IFN-γ release. Given the absence of NKG2C, it is conceivable that activating KIRs may play a role in the HCMV-driven NK cell maturation and that NK cells expressing activating KIRs might contribute, at least in part, to the control of infections after transplantation.


Subject(s)
Cytomegalovirus/immunology , Fetal Blood/transplantation , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Adult , CD56 Antigen/metabolism , Cell Differentiation , Child , Cytomegalovirus Infections/immunology , Hematopoietic Stem Cell Transplantation , Humans , Interferon-gamma/metabolism , NK Cell Lectin-Like Receptor Subfamily C/genetics , Receptors, KIR/metabolism , Receptors, KIR2DL2/metabolism , Receptors, KIR2DL3/metabolism , Receptors, KIR3DL1/metabolism , Receptors, KIR3DS1/metabolism
9.
Cytotherapy ; 17(6): 730-738, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25778757

ABSTRACT

The outcome of umbilical cord blood transplantation for adult patients with hematologic malignancies now rivals that of matched unrelated donor transplantation. However, relatively low lymphocyte and hematopoietic stem and progenitor cell dose is a source of significant morbidity and mortality. Multiple strategies are now being studied to overcome these limitations. One strategy involves ex vivo expansion of the umbilical cord blood unit before transplantation. Ex vivo expansion has the potential to increase the number of lymphocytes, committed progenitors and long-term repopulating hematopoietic stem cells. Increasing the numbers of lymphocytes and committed progenitor cells will address the issue of delayed hematopoietic recovery after umbilical cord blood transplantation. Increasing the hematopoietic stem cell content will improve the availability of adequately sized and matched cord blood units for transplantation. It may also eliminate the need for dual umbilical cord blood transplantation for those without an adequately sized single umbilical cord blood graft. The second strategy involves exposure of the umbilical cord blood graft to compounds aimed at improving homing and engraftment following transplantation. Such a strategy may also address the problem of slow hematopoietic recovery as well as the increased risk of graft failure. Many of these strategies are now being tested in late-phase multi-center clinical trials. If proven cost-effective and efficacious, they may alter the landscape of donor options for allogeneic stem cell transplantation.


Subject(s)
Cord Blood Stem Cell Transplantation , Fetal Blood/cytology , Adult , Cell Proliferation/drug effects , Chelating Agents/pharmacology , Coculture Techniques , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Receptors, Notch/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Treatment Outcome
10.
BMC Immunol ; 15: 44, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25277788

ABSTRACT

BACKGROUND: In former studies we showed in a rat model of renal transplantation that Mesenchymal Stromal Cells (MSC) prevent acute rejection in an independent way of their endowing in the graft. In this study we investigated whether MSC operate by resetting cytokine network and Scatter Factor systems, i.e. Hepatocyte Growth Factor (HGF), Macrophage Stimulating Protein (MSP) and their receptors Met and RON, respectively. METHODS: MSC were injected into the renal artery soon after reperfusion. Controls were grafted untreated and normal rats. Rats were sacrificed 7 days after grafting. Serum and renal tissue levels of IFN-γ, IL-1, IL-2, IL-4, IL-6, IL-10, MSP/RON, HGF/Met systems, Treg lymphocytes were investigated. RESULTS: In grafted untreated rats IFN-γ increased in serum and renal tissue and IL-6 rose in serum. MSC prevented both the phenomena, increased IL-10 serum levels and Treg number in the graft. Furthermore MSC increased serum and tissue HGF levels, Met tubular expression and prevented the suppression of tubular MSP/RON expression. CONCLUSIONS: Our results demonstrate that MSC modify cytokine network to a tolerogenic setting, they suppress Th1 cells, inactivate monocytes/macrophage, recruit Tregs. In addition, MSC sustain the expression of the Scatter Factor systems expression, i.e. systems that are committed to defend survival and stimulate regeneration of tubular cells.


Subject(s)
Cytokines/metabolism , Hepatocyte Growth Factor/metabolism , Kidney Transplantation , Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Allografts , Animals , Cell Proliferation , Cytokines/blood , Forkhead Transcription Factors/metabolism , Hepatocyte Growth Factor/blood , Hepatocyte Growth Factor/genetics , Kidney Tubules/pathology , Monocytes/metabolism , Necrosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-met/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Receptor Protein-Tyrosine Kinases/genetics
11.
Radiology ; 271(3): 805-13, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24592961

ABSTRACT

PURPOSE: To assess the presence of alteration of bone structure and bone marrow metabolism in adult patients who were suspected of having advanced chronic lymphocytic leukemia (ACLL) by using a computational prognostic model that was based on computational analysis of positron emission tomography (PET)/computed tomography (CT) images. MATERIALS AND METHODS: In this retrospective study, all patients signed written informed consent as a requisite to undergo PET/CT examination. However, due to its observational nature, approval from the ethical committee was not deemed necessary. Twenty-two previously untreated chronic lymphocytic leukemia patients underwent PET/CT for disease progression. PET/CT images were analyzed by using dedicated software, capable of recognizing an external 2-pixel bone ring whose Hounsfield coefficient served as cutoff to recognize trabecular and compact bone. PET/CT data from 22 age- and sex-matched control subjects were used as comparison. All data are reported as means ± standard deviations. The Student t test, log-rank, or Cox proportional hazards model were used as appropriate, considering a difference with a P value of less than .05 as significant. RESULTS: Trabecular bone was expanded in ACLL patients and occupied a larger fraction of the skeleton with respect to control subjects (mean, 39% ± 5 [standard deviation] vs 31% ± 7; ie, 32 of 81 mL/kg of ideal body weight vs 27 of 86 mL/kg of ideal body weight, respectively; P < .001). After stratification according to median value, patients with a ratio of trabecular to skeletal bone volume of more than 37.3% showed an actuarial 2-year survival of 18%, compared with 82% for those with a ratio of less than 37.3% (P < .001), independent from age, sex, biological markers, and disease duration. CONCLUSION: These data suggest that computational assessment of skeletal alterations might represent a new window for prediction of the clinical course of the disease.


Subject(s)
Bone and Bones/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Multimodal Imaging , Adolescent , Adult , Aged , Aged, 80 and over , Algorithms , Bone and Bones/diagnostic imaging , Female , Fluorodeoxyglucose F18 , Humans , Image Interpretation, Computer-Assisted , Leukemia, Lymphocytic, Chronic, B-Cell/diagnostic imaging , Male , Middle Aged , Positron-Emission Tomography/methods , Radiopharmaceuticals , Retrospective Studies , Tomography, X-Ray Computed/methods , Whole Body Imaging/methods
12.
Blood ; 119(2): 399-410, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22096237

ABSTRACT

Natural killer (NK) cells play a crucial role in early immunity after hematopoietic stem cell transplantation because they are the first lymphocyte subset recovering after the allograft. In this study, we analyzed the development of NK cells after intrabone umbilical cord blood (CB) transplantation in 18 adult patients with hematologic malignancies. Our data indicate that, also in this transplantation setting, NK cells are the first lymphoid population detectable in peripheral blood. However, different patterns of NK-cell development could be identified. Indeed, in a group of patients, a relevant fraction of NK cells expressed a mature phenotype characterized by the KIR(+)NKG2A(-) signature 3-6 months after transplantation. In other patients, most NK cells maintained an immature phenotype even after 12 months. A possible role for cytomegalovirus in the promotion of NK-cell development was suggested by the observation that a more rapid NK-cell maturation together with expansion of NKG2C(+) NK cells was confined to patients experiencing cytomegalovirus reactivation. In a fraction of these patients, an aberrant and hyporesponsive CD56(-)CD16(+)p75/AIRM1(-) NK-cell subset (mostly KIR(+)NKG2A(-)) reminiscent of that described in patients with viremic HIV was detected. Our data support the concept that cytomegalovirus infection may drive NK-cell development after umbilical CB transplantation.


Subject(s)
Cord Blood Stem Cell Transplantation , Cytomegalovirus Infections/immunology , Fetal Blood/cytology , Graft vs Leukemia Effect/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Leukemia, Myeloid, Acute/therapy , Adult , Antigens, CD/immunology , Antigens, CD/metabolism , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/microbiology , Cytomegalovirus Infections/pathology , Female , Fetal Blood/metabolism , Humans , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/microbiology , Male , Middle Aged , Phenotype , Young Adult
13.
Proc Natl Acad Sci U S A ; 108(42): 17384-9, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-21960443

ABSTRACT

Dendritic cells (DC) are highly specialized antigen-presenting cells characterized by the ability to prime T-cell responses. Mesenchymal stem cells (MSC) are adult stromal progenitor cells displaying immunomodulatory activities including inhibition of DC maturation in vitro. However, the specific impact of MSC on DC functions, upon in vivo administration, has never been elucidated. Here we show that murine MSC impair Toll-like receptor-4 induced activation of DC resulting in the inhibition of cytokines secretion, down-regulation of molecules involved in the migration to the lymph nodes, antigen presentation to CD4(+) T cells, and cross-presentation to CD8(+) T cells. These effects are associated with the inhibition of phosphorylation of intracellular mitogen-activated protein kinases. Intravenous administration of MSC decreased the number of CCR7 and CD49dß1 expressing CFSE-labeled DC in the draining lymph nodes and hindered local antigen priming of DO11.10 ovalbumin-specific CD4(+) T cells. Upon labeling of DC with technetium-99m hexamethylpropylene amine oxime to follow their in vivo biodistribution, we demonstrated that intravenous injection of MSC blocks, almost instantaneously, the migration of subcutaneously administered ovalbumin-pulsed DC to the draining lymph nodes. These findings indicate that MSC significantly affect DC ability to prime T cells in vivo because of their inability to home to the draining lymph nodes and further confirm MSC potentiality as therapy for immune-mediated diseases.


Subject(s)
Dendritic Cells/immunology , Mesenchymal Stem Cells/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Movement/immunology , Coculture Techniques , Cytokines/genetics , Dendritic Cells/cytology , Dendritic Cells/physiology , Dendritic Cells/transplantation , Gene Expression , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
15.
Br J Haematol ; 163(4): 501-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23992039

ABSTRACT

Mesenchymal stromal cell (MSC) infusions have been reported to be effective in patients with steroid-refractory, acute graft-versus-host disease (aGvHD) but comprehensive data on paediatric patients are limited. We retrospectively analysed a cohort of 37 children (aged 3 months-17 years) treated with MSCs for steroid-refractory grade III-IV aGvHD. All patients but three received multiple MSC infusions. Complete response (CR) was observed in 24 children (65%), while 13 children had either partial (n = 8) or no response (n = 5). Cumulative incidence of transplantation-related mortality (TRM) in patients who did or did not achieve CR was 17% and 69%, respectively (P = 0.001). After a median follow-up of 2.9 years, overall survival (OS) was 37%; it was 65% vs. 0% in patients who did or did not achieve CR, respectively (P = 0.001). The median time from starting steroids for GvHD treatment to first MSC infusion was 13 d (range 5-85). Children treated between 5 and 12 d after steroid initiation showed a trend for better OS (56%) and lower TRM (17%) as compared with patients receiving MSCs 13-85 d after steroids (25% and 53%, respectively; P = 0.22 and 0.06, respectively). Multiple MSC infusions are safe and effective for children with steroid-refractory aGvHD, especially when employed early in the disease course.


Subject(s)
Graft vs Host Disease/surgery , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Acute Disease , Adolescent , Child , Child, Preschool , Cohort Studies , Female , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Hematologic Neoplasms/immunology , Hematologic Neoplasms/surgery , Humans , Infant , Male , Neoplasm Grading , Remission Induction , Steroids/administration & dosage
16.
Haematologica ; 98(4): 555-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23242593

ABSTRACT

Several studies have established an association between iron chelation therapy with deferasirox and hematopoietic improvement in patients with myelodysplastic syndromes. There are no data from patients with ß-thalassemia major. In a cross-sectional study, we evaluated the absolute number of several hematopoietic peripheral progenitors (colony-forming unit-granulocyte/macrophage, erythroid burst-forming units, colony-forming unit-granulocyte/erythrocyte/macrophage/megakaryocyte, and long-term culture-initiating cells) in 30 patients with ß-thalassemia major (median age 29.5 years, 40% males) and 12 age-matched controls. For the ß-thalassemia major patients, data on splenectomy status, the type of iron chelator used, and serum ferritin levels reflecting changes in iron status on the chelator were also retrieved. All patients had to be using the same iron chelator for at least 6 months with >80% compliance. The absolute number of all hematopoietic peripheral progenitors was higher in ß-thalassemia major patients than in controls, and varied between splenectomized and non-splenectomized patients (lower number of erythroid burst-forming units and higher numbers of colony-forming unit-granulocyte/macrophage, colony-forming unit-granulocyte/erythrocyte/macrophage/megakaryocyte, and long-term culture-initiating cells). The number of erythroid burst-forming units was significantly higher in patients taking deferasirox (n=10) than in those taking either deferoxamine (n=10) or deferiprone (n=10) (P<0.05). After adjusting for age, sex, splenectomy status, and serum ferritin changes, the association between a higher absolute number of erythroid burst-forming units in deferasirox-treated patients than in patients taking deferoxamine or deferiprone remained statistically significant (P=0.011). In conclusion, in ß-thalassemia major patients, compared with other iron chelators, deferasirox therapy is associated with higher levels of circulating erythroid burst-forming units. This variation is independent of iron status changes and is more likely to be due to the type of chelator.


Subject(s)
Chelation Therapy/methods , Hematopoietic Stem Cells/drug effects , Iron Chelating Agents/therapeutic use , beta-Thalassemia/drug therapy , Adult , Benzoates/therapeutic use , Blood Cell Count , Colony-Forming Units Assay , Cross-Sectional Studies , Deferasirox , Deferiprone , Deferoxamine/therapeutic use , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Female , Ferritins/blood , Flow Cytometry , Hematopoietic Stem Cells/cytology , Humans , Linear Models , Male , Multivariate Analysis , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/drug effects , Pyridones/therapeutic use , Splenectomy , Triazoles/therapeutic use , Young Adult , beta-Thalassemia/blood
17.
BMC Cancer ; 13: 348, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23865766

ABSTRACT

BACKGROUND: Mutation(s) of the JAK2 gene (V617F) has been described in a significant proportion of Philadelphia negative Myeloproliferative Neoplasms (MPN) patients and its detection is now a cornerstone in the diagnostic algorithm. METHODS: We developed a novel assay based on peptide nucleic acid (PNA) technology coupled to immuno-fluorescence microscopy (PNA-FISH) for the specific detection at a single cell level of JAK2-mutation thus improving both the diagnostic resolution and the study of clonal prevalence. RESULTS: Using this assay we found a percentage of mutated CD34+ cells ranging from 40% to 100% in Polycythemia Vera patients, from 15% to 80% in Essential Thrombocythemia and from 25% to 100% in Primary Myelofibrosis. This method allows to distinguish, with a high degree of specificity, at single cell level, between CD34+ progenitor stem cells harbouring the mutated or the wild type form of JAK2 in NPM patients. CONCLUSIONS: This method allows to identify multiple gene abnormalities which will be of paramount relevance to understand the pathophysiology and the evolution of any type of cancer.


Subject(s)
Janus Kinase 2/genetics , Microscopy, Fluorescence/methods , Mutation , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Peptide Nucleic Acids , Cell Separation , Flow Cytometry , Humans , Polymerase Chain Reaction
18.
Biol Blood Marrow Transplant ; 18(1): 92-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21689774

ABSTRACT

Cytomegalovirus (CMV) infection and disease are important complications after hematopoietic stem cell transplant, particularly after transplant from alternative donors. Allogeneic cord blood transplantation (CBT) is being increasingly used, but immune recovery may be delayed. The aim of this study was to compare CMV infection in CBT with transplants from unrelated or mismatched related donors, from now on defined as alternative donors. A total of 165 consecutive transplants were divided in 2 groups: (1) alternative donors transplants (n = 85) and (2) CBT recipients (n = 80). Donor and recipient (D/R) CMV serostatus were recorded. The incidence of CMV infection, its severity, timing, and outcome were compared. Median follow-up was 257 days (1-1328). CMV infection was monitored by CMV antigenemia and expressed as CMV Ag positive cell/2 × 10(5) polymorphonuclear blood cells. There was a trend toward a higher cumulative incidence of CMV infection among CBT than alternative donor transplant recipients (64% vs 51%, P = .12). The median time to CMV reactivation was 35 days, and was comparable in the 2 groups (P = .8). The maximum number of CMV-positive cells was similar in the 2 groups (11 versus 16, P = .2). The time interval between the first and the last positive CMV antigenemia was almost 4 times longer in CBT compared with alternative donor transplants (109 vs 29 days, respectively, P = .008). The incidence of late CMV infection was also higher in CBT (62% vs 24%, P < .001). The incidence of early and late CMV infection in CBT was similar to D-/R+ alternative transplants, and higher than in D+/R+ alternative transplants: early infection, 72% in CBT versus 69% in D-/R+ alternative versus 55% in D+/R+ alternative (P = .21); and late infection, 67% in CBT versus 60% in D-/R+ alternative versus 7% in D+/R+ alternative (P < .001). Transplant-related mortality and overall survival were similar between the groups: 34% versus 36% (P = .6) and 54% versus 46% (P = .3) for alternative transplant and CBT, respectively. Longer duration and higher incidence of late CMV infection was seen in CBT patients, when compared with alternative donor transplants, whereas no difference in mortality was observed. The duration and incidence of late CMV infection were similar when D-/R+ CBT were compared with D-/R+ alternative donor transplants.


Subject(s)
Cord Blood Stem Cell Transplantation/adverse effects , Cytomegalovirus Infections/etiology , Tissue Donors , Adolescent , Adult , Aged , Cord Blood Stem Cell Transplantation/methods , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/immunology , Female , Fetal Blood/virology , HLA-A Antigens/immunology , HLA-B Antigens/immunology , HLA-DR Antigens/immunology , Humans , Incidence , Male , Middle Aged , Retrospective Studies , Survival Rate , Transplantation, Homologous , Young Adult
19.
Blood ; 116(1): 122-8, 2010 Jul 08.
Article in English | MEDLINE | ID: mdl-20395416

ABSTRACT

Human Development Index (HDI) is used by the United Nations Organization to measure socioeconomic achievements of countries. We evaluated the association of HDI with rates and outcomes of hematopoietic stem cell transplantation (HSCT) for patients with acute leukemia. For the analysis of HSCT rates, all adults with acute leukemia (n = 16 403) treated in 30 European countries, between 2001 and 2005, were included. Association of HDI with the outcome was analyzed for 2015 patients with acute myeloid leukemia treated with myeloablative allotransplantation. Countries were classified according to HDI quintiles. Highly significant correlation was found for HDI and the total number of HSCT per population (R = 0.78; P < .001), as well as separately for sibling HSCT (R = 0.84; P < .001), unrelated HSCT (R = 0.66; P < .001), and autologous HSCT (R = 0.43; P = .02). The probabilities of leukemia-free survival for 5 consecutive groups of countries with increasing HDI were: 56%, 59%, 63%, 58%, and 68% (P = .01). In a multivariate analysis, transplantations performed in countries belonging to the upper HDI category were associated with higher leukemia-free survival compared with the remaining ones (HR = 1.36, P = .008), which resulted mainly from reduced risk of relapse (HR = 0.72, P = .04). We conclude that, in Europe, the HDI is associated with both rates and results of HSCT for acute leukemia.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Leukemia/surgery , Outcome Assessment, Health Care/statistics & numerical data , Acute Disease , Adolescent , Adult , Aged , Educational Status , Humans , Kaplan-Meier Estimate , Leukemia/classification , Longevity , Middle Aged , Outcome Assessment, Health Care/methods , Retrospective Studies , Social Class , Socioeconomic Factors , Transplantation, Autologous , Transplantation, Homologous , Transplantation, Isogeneic , Young Adult
20.
Eur J Nucl Med Mol Imaging ; 39(8): 1326-38, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22639281

ABSTRACT

PURPOSE: Despite their relevance in clinical medicine, the extension and activity of the bone marrow (BM) cannot be directly evaluated in vivo. We propose a new method to estimate these variables by combining structural and functional maps provided by CT and PET. METHODS: BM extension and glucose uptake were estimated in 102 patients undergoing whole-body PET/CT because of a history of nonmetastatic melanoma. Image analysis assumed that the BM is surrounded by compact bone. An iterative optimization scheme was applied to each CT slice to identify the external border of the bone. To identify compact bone, the algorithm measured the average Hounsfield coefficient within a two-pixel ring located just inside the bone contour. All intraosseous pixels with an attenuation coefficient lower than this cut-off were flagged as 1, while the remaining pixels were set at 0. Binary masks created from all CT slices were thus applied to the PET data to determine the metabolic activity of the intraosseous volume (IBV). RESULTS: Estimated whole-body IBV was 1,632 ± 587 cm(3) and was higher in men than in women (2,004 ± 498 cm(3) vs. 1,203 ± 354 cm(3), P < 0.001). Overall, it was strictly correlated with ideal body weight (r = 0.81, P = 0.001) but only loosely with measured body weight (r = 0.43, P = 0.01). The average FDG standardized uptake value (SUV) in the thoracic and lumbar vertebrae was 2.01 ± 0.36, Accordingly, intraosseous voxels with SUV ≥ 1.11 (mean spine SUV - 2.5 × SD) were considered as active "red" BM and those with SUV <1.11 as "yellow" BM. Estimated red BM volume was 541 ± 195 ml, with a higher prevalence in the axial than in the appendicular skeleton (87 ± 8 % vs. 10 ± 8 %, P < 0.001). Again, red BM volume was higher in men than in women (7.8 ± 2.2 vs. 6.7 ± 2.1 ml/kg body weight, P < 0.05), but in women it occupied a greater fraction of the IBV (32 ± 7 % vs. 36 ± 10 %, P < 0.05). Patient age modestly predicted red BM SUV, while it was robustly and inversely correlated with red BM volume. CONCLUSION: Our computational analysis of PET/CT images provides a first estimation of the extension and metabolism of the BM in a population of adult patients without haematooncological disorders. This information might represent a new window to explore pathophysiology the BM and the response of BM diseases to chemotherapy.


Subject(s)
Algorithms , Bone Marrow/metabolism , Image Processing, Computer-Assisted/methods , Multimodal Imaging , Positron-Emission Tomography , Tomography, X-Ray Computed , Age Factors , Bone Marrow/diagnostic imaging , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Databases, Factual , Female , Humans , Male , Middle Aged , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL