Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Blood ; 132(19): 2053-2066, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30213875

ABSTRACT

Although the pathogenesis of primary myelofibrosis (PMF) and other myeloproliferative neoplasms (MPNs) is linked to constitutive activation of the JAK-STAT pathway, JAK inhibitors have neither curative nor MPN-stem cell-eradicating potential, indicating that other targetable mechanisms are contributing to the pathophysiology of MPNs. We previously demonstrated that Abelson interactor 1 (Abi-1), a negative regulator of Abelson kinase 1, functions as a tumor suppressor. Here we present data showing that bone marrow-specific deletion of Abi1 in a novel mouse model leads to development of an MPN-like phenotype resembling human PMF. Abi1 loss resulted in a significant increase in the activity of the Src family kinases (SFKs), STAT3, and NF-κB signaling. We also observed impairment of hematopoietic stem cell self-renewal and fitness, as evidenced in noncompetitive and competitive bone marrow transplant experiments. CD34+ hematopoietic progenitors and granulocytes from patients with PMF showed decreased levels of ABI1 transcript as well as increased activity of SFKs, STAT3, and NF-κB. In aggregate, our data link the loss of Abi-1 function to hyperactive SFKs/STAT3/NF-κB signaling and suggest that this signaling axis may represent a regulatory module involved in the molecular pathophysiology of PMF.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Bone Marrow/pathology , Cytoskeletal Proteins/genetics , Gene Deletion , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Animals , Bone Marrow/metabolism , Cell Self Renewal , Cells, Cultured , Down-Regulation , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Primary Myelofibrosis/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , src-Family Kinases/metabolism
2.
Blood ; 127(10): 1249-59, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26787733

ABSTRACT

Megakaryopoiesis is a complex, stepwise process that takes place largely in the bone marrow. At the apex of the hierarchy, hematopoietic stem cells undergo a number of lineage commitment decisions that ultimately lead to the production of polyploid megakaryocytes. On average, megakaryocytes release 10(11) platelets per day into the blood that repair vascular injuries and prevent excessive bleeding. This differentiation process is tightly controlled by exogenous and endogenous factors, which have been the topics of intense research in the hematopoietic field. Indeed, a skewing of megakaryocyte commitment and differentiation may entail the onset of myeloproliferative neoplasms and other preleukemic disorders together with acute megakaryoblastic leukemia, whereas quantitative or qualitative defects in platelet production can lead to inherited platelet disorders. The recent advent of next-generation sequencing has prompted mapping of the genomic landscape of these conditions to provide an accurate view of the underlying lesions. The aims of this review are to introduce the physiological pathways of megakaryopoiesis and to present landmark studies on acquired and inherited disorders that target them. These studies have not only introduced a new era in the fields of molecular medicine and targeted therapies but may also provide us with a better understanding of the mechanisms underlying normal megakaryopoiesis and thrombopoiesis that can inform efforts to create alternative sources of megakaryocytes and platelets.


Subject(s)
Blood Platelet Disorders , Blood Platelets , Genetic Diseases, Inborn , Genome, Human , Megakaryocytes , Thrombopoiesis/genetics , Animals , Blood Platelet Disorders/genetics , Blood Platelet Disorders/metabolism , Blood Platelet Disorders/pathology , Blood Platelets/metabolism , Blood Platelets/pathology , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , High-Throughput Nucleotide Sequencing , Humans , Megakaryocytes/metabolism , Megakaryocytes/pathology
3.
Mol Med ; 23: 235-246, 2017 10.
Article in English | MEDLINE | ID: mdl-28805233

ABSTRACT

Adoptive immunotherapy with Cytokine Induced Killer (CIK) cells has shown antitumor activity against several kinds of cancers in preclinical models and clinical trials. CIK cells are a subset of ex vivo expanded T lymphocytes with T-NK phenotype and MHC-unrestricted antitumor activity. Literature provides scanty information on cytokines, chemokines and growth factors secreted by CIK cells. Therefore, we investigated the secretory profile of CIK cells generated from tumor patients. The secretome analysis was performed at specific time points (day 1, day 14 and day 21) of CIK cells expansion. Mature CIK cells (day 21) produce a great variety of interleukins and secreted proteins that can be divided into 3 groups based on their secretion quantity: high (IL-13, RANTES, MIP-1α and 1ß), medium (IL-1Ra, IL-5, IL-8, IL-10, IL-17, IP-10, INF-γ, VEGF and GMCSF) and low (IL-1ß, IL-4, IL-6, IL-7, IL-9, IL-12, IL-15, Eotaxin, PDGF-bb, FGF basic, G-CSF and MCP-1) secreted. Moreover, comparing PBMC (day 1) and mature CIK cells (day 14 and 21) secretome, we observed that IL-5, IL-10, IL-13, GM-CSF, VEGF resulted greatly up-regulated, while IL-1ß, IL-6, IL-8, IL-15, IL-17, eotaxin, MCP-1, and RANTES were down-regulated. We also performed a gene expression profile analysis of patient-derived CIK cells showing that mRNA for the different cytokines and secreted proteins were modulated during PBMC to CIK differentiation. We highlighted previously unknown secretory properties and provided for the first time a comprehensive molecular characterization of CIK cells. Our findings provide rationale to explore the functional implications and possible therapeutic modulation of CIK secretome.


Subject(s)
Cytokine-Induced Killer Cells/metabolism , Cytokines/metabolism , Gastrointestinal Stromal Tumors/metabolism , Aged , Cell Proliferation , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Transcriptome
4.
Int J Mol Sci ; 18(1)2017 Jan 13.
Article in English | MEDLINE | ID: mdl-28098757

ABSTRACT

Primary Myelofibrosis (PMF) is a chronic Philadelphia-negative myeloproliferative neoplasm characterized by a skewed megakaryopoiesis and an overproduction of proinflammatory and profibrotic mediators that lead to the development of bone marrow (BM) fibrosis. Since we recently uncovered the upregulation of miR-34a-5p in PMF CD34+ hematopoietic progenitor cells (HPCs), in order to elucidate its role in PMF pathogenesis here we unravelled the effects of miR-34a-5p overexpression in HPCs. We showed that enforced expression of miR-34a-5p partially constrains proliferation and favours the megakaryocyte and monocyte/macrophage commitment of HPCs. Interestingly, we identified lymphoid enhancer-binding factor 1 (LEF1) and nuclear receptor subfamily 4, group A, member 2 (NR4A2) transcripts as miR-34a-5p-targets downregulated after miR-34a-5p overexpression in HPCs as well as in PMF CD34+ cells. Remarkably, the knockdown of NR4A2 in HPCs mimicked the antiproliferative effects of miR-34a-5p overexpression, while the silencing of LEF1 phenocopied the effects of miR-34a-5p overexpression on HPCs lineage choice, by favouring the megakaryocyte and monocyte/macrophage commitment. Collectively our data unravel the role of miR-34a-5p in HPCs fate decision and suggest that the increased expression of miR-34a-5p in PMF HPCs could be important for the skewing of megakaryopoiesis and the production of monocytes, that are key players in BM fibrosis in PMF patients.


Subject(s)
Cell Lineage , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , MicroRNAs/metabolism , Primary Myelofibrosis/pathology , Antigens, CD34/metabolism , Case-Control Studies , Cell Differentiation , Cell Proliferation , Clone Cells , Down-Regulation/genetics , Gene Expression Profiling , Gene Silencing , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Macrophages/metabolism , Macrophages/pathology , Megakaryocytes/metabolism , Megakaryocytes/pathology , MicroRNAs/genetics , Models, Biological , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Primary Myelofibrosis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Int J Cancer ; 138(7): 1657-69, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26547506

ABSTRACT

Primary myelofibrosis (PMF) is a Myeloproliferative Neoplasm (MPN) characterized by megakaryocyte hyperplasia, progressive bone marrow fibrosis, extramedullary hematopoiesis and transformation to Acute Myeloid Leukemia (AML). A number of phenotypic driver (JAK2, CALR, MPL) and additional subclonal mutations have been described in PMF, pointing to a complex genomic landscape. To discover novel genomic lesions that can contribute to disease phenotype and/or development, gene expression and copy number signals were integrated and several genomic abnormalities leading to a concordant alteration in gene expression levels were identified. In particular, copy number gain in the polyamine oxidase (PAOX) gene locus was accompanied by a coordinated transcriptional up-regulation in PMF patients. PAOX inhibition resulted in rapid cell death of PMF progenitor cells, while sparing normal cells, suggesting that PAOX inhibition could represent a therapeutic strategy to selectively target PMF cells without affecting normal hematopoietic cells' survival. Moreover, copy number loss in the chromatin modifier HMGXB4 gene correlates with a concomitant transcriptional down-regulation in PMF patients. Interestingly, silencing of HMGXB4 induces megakaryocyte differentiation, while inhibiting erythroid development, in human hematopoietic stem/progenitor cells. These results highlight a previously un-reported, yet potentially interesting role of HMGXB4 in the hematopoietic system and suggest that genomic and transcriptional imbalances of HMGXB4 could contribute to the aberrant expansion of the megakaryocytic lineage that characterizes PMF patients.


Subject(s)
Gene Dosage , HMGB2 Protein/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Primary Myelofibrosis/genetics , Chromosome Aberrations , Electroporation , Humans , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome , Polyamine Oxidase
6.
Blood ; 124(13): e21-32, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25097177

ABSTRACT

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by megakaryocyte (MK) hyperplasia, bone marrow fibrosis, and abnormal stem cell trafficking. PMF may be associated with somatic mutations in JAK2, MPL, or CALR. Previous studies have shown that abnormal MKs play a central role in the pathophysiology of PMF. In this work, we studied both gene and microRNA (miRNA) expression profiles in CD34(+) cells from PMF patients. We identified several biomarkers and putative molecular targets such as FGR, LCN2, and OLFM4. By means of miRNA-gene expression integrative analysis, we found different regulatory networks involved in the dysregulation of transcriptional control and chromatin remodeling. In particular, we identified a network gathering several miRNAs with oncogenic potential (eg, miR-155-5p) and targeted genes whose abnormal function has been previously associated with myeloid neoplasms, including JARID2, NR4A3, CDC42, and HMGB3. Because the validation of miRNA-target interactions unveiled JARID2/miR-155-5p as the strongest relationship in the network, we studied the function of this axis in normal and PMF CD34(+) cells. We showed that JARID2 downregulation mediated by miR-155-5p overexpression leads to increased in vitro formation of CD41(+) MK precursors. These findings suggest that overexpression of miR-155-5p and the resulting downregulation of JARID2 may contribute to MK hyperplasia in PMF.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , MicroRNAs/genetics , Primary Myelofibrosis/genetics , RNA, Messenger/genetics , Antigens, CD34/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Gene Regulatory Networks , Gene Silencing , Granulocytes/metabolism , Hematopoietic Stem Cells/cytology , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Polycomb Repressive Complex 2/genetics , RNA Interference , Reproducibility of Results , Thrombopoiesis/genetics
7.
Blood ; 119(1): 217-26, 2012 Jan 05.
Article in English | MEDLINE | ID: mdl-22053107

ABSTRACT

Extracellular ATP and UTP nucleotides increase the proliferation and engraftment potential of normal human hematopoietic stem cells via the engagement of purinergic receptors (P2Rs). In the present study, we show that ATP and UTP have strikingly opposite effects on human acute myeloblastic leukemia (AML) cells. Leukemic cells express P2Rs. ATP-stimulated leukemic cells, but not normal CD34+ cells, undergo down-regulation of genes involved in cell proliferation and migration, whereas cell-cycle inhibitors are up-regulated. Functionally, ATP induced the inhibition of proliferation and accumulation of AML cells, but not of normal cells, in the G0 phase of the cell cycle. Exposure to ATP or UTP inhibited AML-cell migration in vitro. In vivo, xenotransplantation experiments demonstrated that the homing and engraftment capacity of AML blasts and CD34+CD38- cells to immunodeficient mice BM was significantly inhibited by pretreatment with nucleotides. P2R-expression analysis and pharmacologic profiling suggested that the inhibition of proliferation by ATP was mediated by the down-regulation of the P2X7R, which is up-regulated on untreated blasts, whereas the inhibition of chemotaxis was mainly mediated via P2Y2R and P2Y4R subtypes. We conclude that, unlike normal cells, P2R signaling inhibits leukemic cells and therefore its pharmacologic modulation may represent a novel therapeutic strategy.


Subject(s)
Adenosine Triphosphate/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transplantation , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Receptors, Purinergic/metabolism , Uridine Triphosphate/pharmacology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Cycle/drug effects , Cells, Cultured , Female , Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction
8.
Blood ; 116(22): e99-110, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-20686118

ABSTRACT

The c-myb transcription factor is highly expressed in immature hematopoietic cells and down-regulated during differentiation. To define its role during the hematopoietic lineage commitment, we silenced c-myb in human CD34(+) hematopoietic stem/progenitor cells. Noteworthy, c-myb silencing increased the commitment capacity toward the macrophage and megakaryocyte lineages, whereas erythroid differentiation was impaired, as demonstrated by clonogenic assay, morphologic and immunophenotypic data. Gene expression profiling and computational analysis of promoter regions of genes modulated in c-myb-silenced CD34(+) cells identified the transcription factors Kruppel-Like Factor 1 (KLF1) and LIM Domain Only 2 (LMO2) as putative targets, which can account for c-myb knockdown effects. Indeed, chromatin immunoprecipitation and luciferase reporter assay demonstrated that c-myb binds to KLF1 and LMO2 promoters and transactivates their expression. Consistently, the retroviral vector-mediated overexpression of either KLF1 or LMO2 partially rescued the defect in erythropoiesis caused by c-myb silencing, whereas only KLF1 was also able to repress the megakaryocyte differentiation enhanced in Myb-silenced CD34(+) cells. Our data collectively demonstrate that c-myb plays a pivotal role in human primary hematopoietic stem/progenitor cells lineage commitment, by enhancing erythropoiesis at the expense of megakaryocyte diffentiation. Indeed, we identified KLF1 and LMO2 transactivation as the molecular mechanism underlying Myb-driven erythroid versus megakaryocyte cell fate decision.


Subject(s)
Antigens, CD34/metabolism , DNA-Binding Proteins/metabolism , Erythropoiesis , Kruppel-Like Transcription Factors/metabolism , Metalloproteins/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Stem Cells/cytology , Adaptor Proteins, Signal Transducing , Cells, Cultured , DNA-Binding Proteins/genetics , Gene Expression Regulation , Gene Silencing , Granulocyte Precursor Cells/cytology , Granulocyte Precursor Cells/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , LIM Domain Proteins , Metalloproteins/genetics , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-myb/genetics , Stem Cells/metabolism
9.
Blood ; 114(25): 5191-200, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19855080

ABSTRACT

We show the molecular and functional characterization of a novel population of lineage-negative CD34-negative (Lin(-)CD34(-)) hematopoietic stem cells from chronic myelogenous leukemia (CML) patients at diagnosis. Molecular karyotyping and quantitative analysis of BCR-ABL transcript demonstrated that approximately one-third of CD34(-) cells are leukemic. CML Lin(-)CD34(-) cells showed kinetic quiescence and limited clonogenic capacity. However, stroma-dependent cultures induced CD34 expression on some cells and cell cycling, and increased clonogenic activity and expression of BCR-ABL transcript. Lin(-)CD34(-) cells showed hematopoietic cell engraftment rate in 2 immunodeficient mouse strains similar to Lin-CD34(+) cells, whereas endothelial cell engraftment was significantly higher. Gene expression profiling revealed the down-regulation of cell-cycle arrest genes and genes involved in antigen presentation and processing, while the expression of genes related to tumor progression, such as angiogenic factors, was strongly up-regulated compared with normal counterparts. Phenotypic analysis confirmed the significant down-regulation of HLA class I and II molecules in CML Lin(-)CD34(-) cells. Imatinib mesylate did not reduce fusion transcript levels, BCR-ABL kinase activity, and clonogenic efficiency of CML Lin(-)CD34(-) cells in vitro. Moreover, leukemic CD34(-) cells survived exposure to BCR-ABL inhibitors in vivo. Thus, we identified a novel CD34(-) leukemic stem cell subset in CML with peculiar molecular and functional characteristics.


Subject(s)
Antigens, CD34/metabolism , Drug Resistance, Neoplasm , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Neoplastic Stem Cells/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Benzamides , Bone Marrow Cells/metabolism , Cells, Cultured , Cluster Analysis , Flow Cytometry , Fusion Proteins, bcr-abl/genetics , Gene Expression Profiling , Humans , Imatinib Mesylate , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Karyotyping , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/transplantation , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics
10.
Stem Cells ; 28(5): 965-73, 2010 May.
Article in English | MEDLINE | ID: mdl-20333749

ABSTRACT

Several reports showed that hematopoietic stem cells (HSCs) participate in muscle regeneration, raising hope for their therapeutic potential for degenerative muscle diseases. However, proof that HSCs are able to reprogram their fate and enter a myogenic pathway, remains elusive. We demonstrate that murine bone marrow (BM)-derived hematopoietic cells, carrying reporter genes controlled by muscle-specific regulatory elements from the Myf5, myosin light chain (MLC3F), or MCK genes, are induced by myoblasts to activate muscle-specific genes. This potential resides in the more undifferentiated progenitors, expressing surface markers typical of HSCs. Comparative gene expression profiling of CD45(+)/Sca1(+) cells isolated from muscle or BM shows that hematopoietic cells participate to muscle regeneration, by undergoing a profound although incomplete myogenic reprogramming on interaction with the muscle microenviroment. These cells undergo specification and differentiation independently from Pax7 and MyoD, and lack Pax7-associated properties, such as self-renewal and proliferation, distinguishing from satellite cells. Our findings indicate that hematopoietic cells, on seeding in the muscle, become a distinct cell population endowed with myogenic potential.


Subject(s)
Bone Marrow Cells/physiology , Cell Differentiation/genetics , Hematopoietic Stem Cells/physiology , Muscle Development/genetics , Myoblasts/physiology , PAX7 Transcription Factor/physiology , Animals , Bone Marrow Cells/cytology , Cell Lineage , Cells, Cultured , Gene Expression Regulation/genetics , Hematopoietic Stem Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/cytology , PAX7 Transcription Factor/deficiency , PAX7 Transcription Factor/genetics
11.
Blood Adv ; 5(5): 1452-1462, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33666652

ABSTRACT

Myelofibrosis (MF) belongs to the family of classic Philadelphia-negative myeloproliferative neoplasms (MPNs). It can be primary myelofibrosis (PMF) or secondary myelofibrosis (SMF) evolving from polycythemia vera (PV) or essential thrombocythemia (ET). Despite the differences, PMF and SMF patients are currently managed in the same way, and prediction of survival is based on the same clinical and genetic features. In the last few years, interest has grown concerning the ability of gene expression profiles (GEPs) to provide valuable prognostic information. Here, we studied the GEPs of granulocytes from 114 patients with MF, using a microarray platform to identify correlations with patient characteristics and outcomes. Cox regression analysis led to the identification of 201 survival-related transcripts characterizing patients who are at high risk for death. High-risk patients identified by this gene signature displayed an inferior overall survival and leukemia-free survival, together with clinical and molecular detrimental features included in contemporary prognostic models, such as the presence of high molecular risk mutations. The high-risk group was enriched in post-PV and post-ET MF and JAK2V617F homozygous patients, whereas pre-PMF was more frequent in the low-risk group. These results demonstrate that GEPs in MF patients correlate with their molecular and clinical features, particularly their survival, and represent the proof of concept that GEPs might provide complementary prognostic information to be applied in clinical decision making.


Subject(s)
Myeloproliferative Disorders , Polycythemia Vera , Primary Myelofibrosis , Thrombocythemia, Essential , Humans , Polycythemia Vera/diagnosis , Polycythemia Vera/genetics , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/genetics , Thrombocythemia, Essential/diagnosis , Thrombocythemia, Essential/genetics , Transcriptome
12.
Sci Rep ; 10(1): 13841, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796906

ABSTRACT

Corneal endothelial (CE) dysfunction is the main indication for corneal transplantation, an invasive procedure with several limitations. Developing novel strategies to re-activate CE regenerative capacity is, therefore, of fundamental importance. This goal has proved to be challenging as corneal endothelial cells (CEnC) are blocked in the G0/G1 phase of the cell cycle in vivo and, albeit retaining proliferative capacity in vitro, this is further hindered by endothelial-to-mesenchymal transition. Herein we investigated the mechanisms regulating CEnC proliferation in vitro. Comparing the proteome of non-proliferating (in vivo-G0/G1) and proliferating (in vitro-G2/M) rabbit CEnC (rCEnC), 77 proteins, out of 3,328 identified, were differentially expressed in the two groups (p < 0.005). Literature and Gene Ontology analysis revealed ß-catenin and transforming growth factor (TGF-ß) pathways to be correlated with the identified proteins. Treatment of rCEnC with a ß-catenin activator and inhibitor showed that ß-catenin activation was necessary during rCEnC proliferation, but not sufficient for its induction. Furthermore, both pro-proliferative activity of basic fibroblast growth factor and anti-proliferative effects of TGF-ß were regulated through ß-catenin. Overall, these results provide novel insights into the molecular basis underlying the proliferation process that CEnC re-activate in vitro, consolidating the role of ß-catenin and TGF-ß.


Subject(s)
Cell Proliferation/genetics , Cell Proliferation/physiology , Endothelial Cells/physiology , Endothelium, Corneal/cytology , Proteomics/methods , beta Catenin/metabolism , Animals , Cells, Cultured , Epithelial-Mesenchymal Transition , Rabbits , Resting Phase, Cell Cycle , Signal Transduction , Transforming Growth Factor beta/metabolism
13.
Stem Cells ; 26(4): 950-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18192237

ABSTRACT

CD34 is a transmembrane protein that is strongly expressed on hematopoietic stem/progenitor cells (HSCs); despite its importance as a marker of HSCs, its function is still poorly understood, although a role in cell adhesion has been demonstrated. To characterize the function of CD34 antigen on human HSCs, we examined, by both inhibition and overexpression, the role of CD34 in the regulation of HSC lineage differentiation. Our results demonstrate that CD34 silencing enhances HSC granulocyte and megakaryocyte differentiation and reduces erythroid maturation. In agreement with these results, the gene expression profile of these cells reveals the upregulation of genes involved in granulocyte and megakaryocyte differentiation and the downregulation of erythroid genes. Consistently, retroviral-mediated CD34 overexpression leads to a remarkable increase in erythroid progenitors and a dramatic decrease in granulocyte progenitors, as evaluated by clonogenic assay. Together, these data indicate that the CD34 molecule promotes the differentiation of CD34+ hematopoietic progenitors toward the erythroid lineage, which is achieved, at least in part, at the expense of granulocyte and megakaryocyte lineages.


Subject(s)
Antigens, CD34/physiology , Cell Differentiation/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Antigens, CD34/genetics , Cells, Cultured , Clone Cells , Gene Expression Profiling , Gene Silencing/physiology , Hematopoietic Stem Cells/metabolism , Humans , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/physiology , Oligonucleotide Array Sequence Analysis
14.
Sci Rep ; 9(1): 10558, 2019 07 22.
Article in English | MEDLINE | ID: mdl-31332222

ABSTRACT

Somatic mutations of calreticulin (CALR) have been described in approximately 60-80% of JAK2 and MPL unmutated Essential Thrombocythemia and Primary Myelofibrosis patients. CALR is an endoplasmic reticulum (ER) chaperone responsible for proper protein folding and calcium retention. Recent data demonstrated that the TPO receptor (MPL) is essential for the development of CALR mutant-driven Myeloproliferative Neoplasms (MPNs). However, the precise mechanism of action of CALR mutants haven't been fully unraveled. In this study, we showed that CALR mutants impair the ability to respond to the ER stress and reduce the activation of the pro-apoptotic pathway of the unfolded protein response (UPR). Moreover, our data demonstrated that CALR mutations induce increased sensitivity to oxidative stress, leading to increase oxidative DNA damage. We finally demonstrated that the downmodulation of OXR1 in CALR-mutated cells could be one of the molecular mechanisms responsible for the increased sensitivity to oxidative stress mediated by mutant CALR. Altogether, our data identify novel mechanisms collaborating with MPL activation in CALR-mediated cellular transformation. CALR mutants negatively impact on the capability of cells to respond to oxidative stress leading to genomic instability and on the ability to react to ER stress, causing resistance to UPR-induced apoptosis.


Subject(s)
Calreticulin/genetics , Calreticulin/metabolism , INDEL Mutation , Oxidative Stress/genetics , Unfolded Protein Response/genetics , Cell Transformation, Neoplastic/genetics , DNA Repair/genetics , Down-Regulation , Endoplasmic Reticulum Stress/genetics , Gene Knockdown Techniques , Humans , K562 Cells , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phenanthrenes/pharmacology , Primary Myelofibrosis/genetics , Primary Myelofibrosis/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Superoxide Dismutase/metabolism , Thrombocythemia, Essential/genetics , Thrombocythemia, Essential/metabolism , Transcriptome
15.
Exp Hematol ; 35(11): 1708-18, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17976522

ABSTRACT

OBJECTIVE: Expression profiling of microRNA (miRNA) was performed in granulocytes isolated from patients with primary myelofibrosis (PMF), with the aim of identifying abnormally expressed miRNAs in comparison with normal subjects or patients with polycythemia vera (PV) or essential thrombocythemia (ET). PATIENTS AND METHODS: Using stem loop-primed reverse transcription and TaqMan quantitative real-time polymerase chain reaction, the expression of 156 mature miRNAs was evaluated using pooled granulocytes from PMF patients, either wild-type or JAK2(617V>F) mutant with >51% allele burden, and control subjects. Differentially expressed miRNAs were then validated on additional control and PMF samples, and also on PV or ET granulocytes. RESULTS: There was a global downregulation of miRNA expression in PMF granulocytes; 60 miRNAs, of 128 called present, displayed differential expression compared to normal samples. Twelve miRNAs, which had been selected based on statistically different expression level, were finally validated. In PMF granulocytes, levels of miR-31, -150, and -95 were significantly lower, while those of miR-190 significantly greater, than control and PV or ET samples; on the other hand, miR-34a, -342, -326, -105, -149, and -147 were similarly reduced in patients with PMF, PV, or ET compared to controls. Increased expression of miR-182 and -183 correlated with JAK2(617V>F) allele burden. Three in silico-predicted putative target genes (DTR, HMGA2, and MYB), showed deregulated expression in PMF granulocytes that correlated with expression level of regulatory miRNA. CONCLUSIONS: A defined miRNA profile distinguishes PMF granulocytes from those of normal subjects and, partially, also from PV or ET patients.


Subject(s)
Gene Expression Profiling/methods , Granulocytes/pathology , MicroRNAs/genetics , Primary Myelofibrosis/genetics , Case-Control Studies , Gene Expression Regulation , Humans , Janus Kinase 2/genetics , MicroRNAs/analysis , Primary Myelofibrosis/pathology
16.
Front Pharmacol ; 9: 196, 2018.
Article in English | MEDLINE | ID: mdl-29731713

ABSTRACT

Cytokine-induced killer (CIK) cells, a heterogeneous T cell population obtained by in vitro differentiation of peripheral blood mononuclear cells (PBMC), represent a promising immunological approach in cancer. Numerous studies have explored the role of CD38, CD39, CD203a/PC-1, and CD73 in generating extracellular adenosine (ADO) and thus in shaping the tumor niche in favor of proliferation. The findings shown here reveal that CIK cells are able to produce extracellular ADO via traditional (CD39/CD73) and/or alternative (CD38/CD203a/CD73 or CD203a/CD73) pathways. Transcriptome analysis showed the mRNA expression of these molecules and their modulation during PBMC to CIK differentiation. When PBMC from normal subjects or cancer bearing patients were differentiated into CIK cells under normoxic conditions, CD38 and CD39 were greatly up-regulated while the number of CD203a, and CD73 positive cells underwent minor changes. Since hypoxic conditions are often found in tumors, we asked whether CD39, CD38, CD203a, and CD73 expressed by CIK cells were modulated by hypoxia. PBMC isolated from cancer patients and differentiated into CIK cells in hypoxic conditions did not show relevant changes in CD38, CD39, CD73, CD203a, and CD26. CIK cells also expressed A1, A2A, and A2B ADO receptors and they only underwent minor changes as a consequence of hypoxia. The present study sheds light on a previously unknown functional aspect of CIK cells, opening the possibility of pharmacologically modulated ADO-generating ectoezymes to improve CIK cells performance.

17.
Mol Oncol ; 12(12): 2102-2123, 2018 12.
Article in English | MEDLINE | ID: mdl-30259659

ABSTRACT

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by an excessive production of pro-inflammatory cytokines resulting in chronic inflammation and genomic instability. Besides the driver mutations in JAK2, MPL, and CALR genes, the deregulation of miRNA expression may also contribute to the pathogenesis of PMF. To this end, we recently reported the upregulation of miR-382-5p in PMF CD34+ cells. In order to unveil the mechanistic details of the role of miR-382-5p in pathogenesis of PMF, we performed gene expression profiling of CD34+ cells overexpressing miR-382-5p. Among the downregulated genes, we identified superoxide dismutase 2 (SOD2), which is a predicted target of miR-382-5p. Subsequently, we confirmed miR-382-5p/SOD2 interaction by luciferase assay and we showed that miR-382-5p overexpression in CD34+ cells causes the decrease in SOD2 activity leading to reactive oxygen species (ROS) accumulation and oxidative DNA damage. In addition, our data indicate that inhibition of miR-382-5p in PMF CD34+ cells restores SOD2 function, induces ROS disposal, and reduces DNA oxidation. Since the pro-inflammatory cytokine transforming growth factor-ß1 (TGF-ß1) is a key player in PMF pathogenesis, we further investigated the effect of TGF-ß1 on ROS and miR-382-5p levels. Our data showed that TGF-ß1 treatment enhances miR-382-5p expression and reduces SOD2 activity leading to ROS accumulation. Finally, inhibition of TGF-ß1 signaling in PMF CD34+ cells by galunisertib significantly reduced miR-382-5p expression and ROS accumulation and restored SOD2 activity. As a whole, this study reports that TGF-ß1/miR-382-5p/SOD2 axis deregulation in PMF cells is linked to ROS overproduction that may contribute to enhanced oxidative stress and inflammation. Our results suggest that galunisertib may represent an effective drug reducing abnormal oxidative stress induced by TGF-ß1 in PMF patients. DATABASE LINKING: GEO: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE103464.


Subject(s)
Antigens, CD34/metabolism , MicroRNAs/metabolism , Oxidative Stress , Primary Myelofibrosis/metabolism , Signal Transduction , Superoxide Dismutase/metabolism , Transforming Growth Factor beta1/metabolism , Antigens, CD34/analysis , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Humans , MicroRNAs/genetics , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Superoxide Dismutase/genetics , Transcriptome
18.
Stem Cells Dev ; 27(4): 225-236, 2018 Feb 15.
Article in English | MEDLINE | ID: mdl-29258411

ABSTRACT

Calreticulin (CALR) is a chaperone protein that localizes primarily to the endoplasmic reticulum (ER) lumen where it is responsible for the control of proper folding of neo-synthesized glycoproteins and the retention of calcium. Recently, mutations affecting exon 9 of the CALR gene have been described in approximately 40% of patients with myeloproliferative neoplasms (MPNs). Although the role of mutated CALR in the development of MPNs has begun to be clarified, there are still no data available on the function of wild-type (WT) CALR during physiological hematopoiesis. To shed light on the role of WT CALR during normal hematopoiesis, we performed gene silencing and overexpression experiments in hematopoietic stem progenitor cells (HSPCs). Our results showed that CALR overexpression is able to affect physiological hematopoiesis by enhancing both erythroid and megakaryocytic (MK) differentiation. In agreement with overexpression data, CALR silencing caused a significant decrease in both erythroid and MK differentiation of human HSPCs. Gene expression profiling (GEP) analysis showed that CALR is able to affect the expression of several genes involved in HSPC differentiation toward both the erythroid and MK lineages. Moreover, GEP data also highlighted the modulation of several genes involved in ER stress response, unfolded protein response (UPR), and DNA repair, and of several genes already described to play a role in MPN development, such as proinflammatory cytokines and hematological neoplasm-related markers. Altogether, our data unraveled a new and unexpected role for CALR in the regulation of normal hematopoietic differentiation. Moreover, by showing the impact of CALR on the expression of genes involved in several biological processes already described in cellular transformation, our data strongly suggest a more complex role for CALR in MPN development that goes beyond the activation of the THPO receptor and involves ER stress response, UPR, and DNA repair.

19.
BMC Genomics ; 8: 264, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17683550

ABSTRACT

BACKGROUND: Human myelopoiesis is an exciting biological model for cellular differentiation since it represents a plastic process where multipotent stem cells gradually limit their differentiation potential, generating different precursor cells which finally evolve into distinct terminally differentiated cells. This study aimed at investigating the genomic expression during myeloid differentiation through a computational approach that integrates gene expression profiles with functional information and genome organization. RESULTS: Gene expression data from 24 experiments for 8 different cell types of the human myelopoietic lineage were used to generate an integrated myelopoiesis dataset of 9,425 genes, each reliably associated to a unique genomic position and chromosomal coordinate. Lists of genes constitutively expressed or silent during myelopoiesis and of genes differentially expressed in commitment phase of myelopoiesis were first identified using a classical data analysis procedure. Then, the genomic distribution of myelopoiesis genes was investigated integrating transcriptional and functional characteristics of genes. This approach allowed identifying specific chromosomal regions significantly highly or weakly expressed, and clusters of differentially expressed genes and of transcripts related to specific functional modules. CONCLUSION: The analysis of genomic expression during human myelopoiesis using an integrative computational approach allowed discovering important relationships between genomic position, biological function and expression patterns and highlighting chromatin domains, including genes with coordinated expression and lineage-specific functions.


Subject(s)
Gene Expression , Genome, Human , Genomics , Myeloid Cells/metabolism , Myelopoiesis/genetics , Antigens, CD34/genetics , Antigens, CD34/metabolism , Cell Differentiation , Cell Lineage , Chromosomes, Human , Cluster Analysis , Computational Biology , Eosinophils/cytology , Eosinophils/metabolism , Erythroblasts/cytology , Erythroblasts/metabolism , Fetal Blood/cytology , Fetal Blood/metabolism , Gene Expression Profiling , Granulocyte Precursor Cells/cytology , Granulocyte Precursor Cells/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Models, Biological , Monocytes/cytology , Monocytes/metabolism , Myeloid Cells/cytology , Neutrophils/cytology , Neutrophils/metabolism , Oligonucleotide Array Sequence Analysis , Software
20.
Haematologica ; 92(10): 1311-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17768116

ABSTRACT

BACKGROUND AND OBJECTIVES: Traditionally eosinophils have been considered terminally differentiated cells that play a role in host protection against parasites. However, there is some evidence showing that eosinophils are, in fact, multifunctional leukocytes involved in inflammatory responses, as well as in tissue homeostasis. We characterized the transcriptome profile of human eosinophils, and, for the purpose of comparison, the transcriptome profile of neutrophils, monocytes and hematopoietic progenitor cells. Moreover, we studied the activation of selected cellular processes for which a significant differential expression was demonstrated. DESIGN AND METHODS: We profiled gene expression using Affymetrix GeneChips. DNA repair capacity was tested using the comet assay. Nucleoli and their activity were characterized by transmission electron microscopy analysis, silver staining of nucleolus regions (AgNOR) and RNA staining. RESULTS: Gene expression profiling showed that eosinophils appear hierarchically closer to monocytes than to neutrophils. Gene ontology mapping of differentially expressed genes revealed that eosinophils express categories very similar to those expressed by monocytes, related to DNA repair and nucleolar functions. Moreover, our data show that eosinophils and monocytes maintain the ability to repair both double and single strand DNA breaks, whereas neutrophils lack this capacity. Furthermore, eosinophils exhibit nucleolar activity, which is lacking in neutrophils, but resembles that in monocytes. INTERPRETATION AND CONCLUSIONS: The presence of large, active nucleoli in eosinophils, coupled to marked activity of DNA repair systems, suggests that eosinophils are not terminally differentiated cells. Indeed, their transcriptome profile and functional properties are more similar to those of non-terminally differentiated cells such as monocytes, rather than to neutrophils.


Subject(s)
Cell Nucleolus/genetics , DNA Repair/genetics , DNA/genetics , Eosinophils/metabolism , Antigens, CD34/metabolism , Biomarkers , Eosinophils/ultrastructure , Gene Expression Profiling , Gene Expression Regulation , Humans , Microscopy, Electron, Transmission , Monocytes/metabolism , Neutrophils/metabolism , Phenotype , RNA, Messenger/genetics , Receptors, IgG/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL