Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Stem Cells Dev ; 23(14): 1568-81, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24617415

ABSTRACT

It has been shown that bone marrow mesenchymal stromal cells (MSCs) from patients with myelodysplastic syndromes (MDSs) display defective proliferative potential. We have probed the impaired replicative capacity of culture-expanded MSCs in MDS patients (n=30) compared with healthy subjects (n=32) by studying senescence characteristics and gene expression associated with WNT/transforming growth factor-ß1 (TGFB1) signaling pathways. We have also explored the consequences of the impaired patient MSC proliferative potential by investigating their differentiation potential and the capacity to support normal CD34(+) cell growth under coculture conditions. Patient MSCs displayed decreased gene expression of the senescence-associated cyclin-dependent kinase inhibitors CDKN1A, CDKN2A, and CDKN2B, along with PARG1, whereas the mean telomere length was upregulated in patient MSCs. MDS-derived MSCs exhibited impaired capacity to support normal CD34(+) myeloid and erythroid colony formation. No significant changes were observed between patients and controls in gene expression related to TGFB1 pathway. Patient MSCs displayed upregulated non-canonical WNT expression, combined with downregulated canonical WNT expression and upregulated canonical WNT inhibitors. MDS-derived MSCs displayed defective osteogenic and adipogenic lineage priming under non-differentiating culture conditions. Pharmacological activation of canonical WNT signaling in patient MDSs led to an increase in cell proliferation and upregulation in the expression of early osteogenesis-related genes. This study indicates abnormal WNT signaling in MSCs of MDS patients and supports the concept of a primary MSC defect that might have a contributory effect in MDS natural history.


Subject(s)
Bone Marrow Cells/pathology , Mesenchymal Stem Cells/pathology , Myelodysplastic Syndromes/genetics , Wnt Signaling Pathway/genetics , Adipogenesis/genetics , Aged , Aged, 80 and over , Cell Differentiation/genetics , Cell Lineage , Cell Proliferation/genetics , Female , Gene Expression Regulation , Humans , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Osteogenesis/genetics
2.
Curr Stem Cell Res Ther ; 8(2): 144-55, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23279098

ABSTRACT

In recent years there seems to be an unbounded interest concerning mesenchymal stem cells (MSCs). This is mainly attributed to their exciting characteristics including long-term ex vivo proliferation, multilineage potential and immunomodulatory properties. In this regard MSCs emerge as attractive candidates for various therapeutic applications. MSCs were originally isolated from the bone marrow (BM) and this population is still considered as the gold standard for MSC applications. Nevertheless the BM has several limitations as source of MSCs, including MSC low frequency in this compartment, the painful isolation procedure and the decline in MSC characteristics with donor's age. Thus, there is accumulating interest in identifying alternative sources for MSCs. To this end MSCs obtained from the Wharton's Jelly (WJ) of umbilical cords (UC) have gained much attention over the last years since they can be easily isolated, without any ethical concerns, from a tissue which is discarded after birth. Furthermore WJ-derived MSCs represent a more primitive population than their adult counterparts, opening new perspectives for cell-based therapies. In this review we will at first give an overview of the biology of WJ-derived UC-MSCs. Then their potential application for the treatment of cancer and immune mediated disorders, such graft versus host disease (GVHD) and systemic lupus erythematosus (SLE) will be discussed, and finally their putative role as feeder layer for ex vivo hematopoietic stem cell (HSC) expansion will be pointed out.


Subject(s)
Clinical Trials as Topic , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Wharton Jelly/cytology , Animals , Humans , Killer Cells, Natural/cytology
3.
Stem Cells Dev ; 22(9): 1329-41, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23249221

ABSTRACT

The bone marrow (BM) microenvironment has clearly been implicated in the pathogenesis of B-cell chronic lymphocytic leukemia (B-CLL). However, the potential involvement of BM stromal progenitors, the mesenchymal stem cells (MSCs), in the pathophysiology of the disease has not been extensively investigated. We expanded in vitro BM-MSCs from B-CLL patients (n=11) and healthy individuals (n=16) and comparatively assessed their reserves, proliferative potential, differentiation capacity, and immunoregulatory effects on T- and B-cells. We also evaluated the anti-apoptotic effect of patient-derived MSCs on leukemic cells and studied their cytogenetic characteristics in comparison to BM hematopoietic cells. B-CLL-derived BM MSCs exhibit a similar phenotype, differentiation potential, and ability to suppress T-cell proliferative responses as compared with MSCs from normal controls. Furthermore, they do not carry the cytogenetic abnormalities of the leukemic clone, and they exert a similar anti-apoptotic effect on leukemic cells and healthy donor-derived B-cells, as their normal counterparts. On the other hand, MSCs from B-CLL patients significantly promote normal B-cell proliferation and IgG production, in contrast to healthy-donor-derived MSCs. Furthermore, they have impaired reserves, defective cellular growth due to increased apoptotic cell death and exhibit aberrant production of stromal cell-derived factor 1, B-cell activating factor, a proliferation inducing ligand, and transforming growth factor ß1, cytokines that are crucial for the survival/nourishing of the leukemic cells. We conclude that ex vivo expanded B-CLL-derived MSCs harbor intrinsic qualitative and quantitative abnormalities that may be implicated in disease development and/or progression.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mesenchymal Stem Cells/immunology , Aged , Aged, 80 and over , Apoptosis , B-Lymphocytes/physiology , Case-Control Studies , Cell Differentiation , Cell Proliferation , Cell Shape , Cell Survival , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Female , Hematopoietic Stem Cells/metabolism , Humans , Immunoglobulin G/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , T-Lymphocytes/physiology
4.
Clin Dev Immunol ; 2013: 265608, 2013.
Article in English | MEDLINE | ID: mdl-24386000

ABSTRACT

Immune-mediated bone marrow failure syndromes (BMFS) are characterized by ineffective marrow haemopoiesis and subsequent peripheral cytopenias. Ineffective haemopoiesis is the result of a complex marrow deregulation including genetic, epigenetic, and immune-mediated alterations in haemopoietic stem/progenitor cells, as well as abnormal haemopoietic-to-stromal cell interactions, with abnormal release of haemopoietic growth factors, chemokines, and inhibitors. Mesenchymal stem/stromal cells (MSCs) and their progeny (i.e., osteoblasts, adipocytes, and reticular cells) are considered as key cellular components of the bone marrow haemopoietic niche. MSCs may interfere with haemopoietic as well as immune regulation. Evidence suggests that bone marrow MSCs may be involved in immune-mediated BMFS underlying pathophysiology, harboring either native abnormalities and/or secondary defects, caused by exposure to activated marrow components. This review summarizes previous as well as more recent information related to the biologic/functional characteristics of bone marrow MSCs in myelodysplastic syndromes, acquired aplastic anemia, and chronic idiopathic neutropenia.


Subject(s)
Hemoglobinuria, Paroxysmal/immunology , Hemoglobinuria, Paroxysmal/metabolism , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Anemia, Aplastic/immunology , Anemia, Aplastic/metabolism , Bone Marrow Diseases , Bone Marrow Failure Disorders , Cell Differentiation , Humans , Mesenchymal Stem Cells/cytology , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/metabolism , Neutropenia/immunology , Neutropenia/metabolism
5.
J Inflamm (Lond) ; 9(1): 27, 2012 Jul 02.
Article in English | MEDLINE | ID: mdl-22747954

ABSTRACT

OBJECTIVE: Idiopathic Pulmonary Fibrosis and Rheumatoid Arthritis associated usual interstitial pneumonia seem to have the same poor outcome as there is not an effective treatment. The aim of the study is to explore the reparative ability of bone marrow mesenchymal stem cells by evaluating the system telomerase/telomeres and propose a novel therapeutic approach. METHODS: BM-MSCs were studied in 6 IPF patients, 7 patients with RA-UIP and 6 healthy controls. We evaluated the telomere length as well as the mRNA expression of both components of telomerase (human telomerase reverse transcriptase, h-TERT and RNA template complementary to the telomeric loss DNA, h-TERC). RESULTS: We found that BM-MSCs from IPF, RA-UIP cases do not present smaller telomere length than the controls (p = 0.170). There was no significant difference regarding the expression of both h-TERT and h-TERC genes between patients and healthy controls (p = 0.107 and p = 0.634 respectively). CONCLUSIONS: We demonstrated same telomere length and telomerase expression in BM-MSCs of both IPF and RA-UIP which could explain similarities in pathogenesis and prognosis. Maintenance of telomere length in these cells could have future implication in cell replacement treatment with stem cells of these devastating lung disorders.

6.
Haematologica ; 97(5): 743-50, 2012 May.
Article in English | MEDLINE | ID: mdl-22133767

ABSTRACT

BACKGROUND: Chronic idiopathic neutropenia is characterized by immune-mediated suppression of neutrophil production. Because patients with immune-mediated bone marrow failure syndromes display age-inappropriate telomere shortening in leukocytes, we investigated telomere lengths in peripheral blood mononuclear cells and granulocytes of patients with chronic idiopathic neutropenia. DESIGN AND METHODS: We studied 37 patients with chronic idiopathic neutropenia and 68 age- and sex-matched healthy controls. Relative telomere length and telomerase reverse transcriptase expression were assessed by a quantitative real time polymerase chain reaction. Telomerase activity was determined by a polymerase chain reaction-based immunoassay. RESULTS: The mean relative telomere values of peripheral blood mononuclear cells and granulocytes were significantly lower in patients compared to controls, and significantly lower than expected on the basis of the age-adjusted healthy control distribution. The difference in the relative telomere lengths between patients and controls in both peripheral blood mononuclear cells and granulocytes was prominent in those under the age of 50 years. Contrary to the peripheral blood mononuclear cells, in which an inverse correlation was observed between relative telomere values and age, no significant correlation was noted between granulocyte telomere values and patient age. A significant correlation was observed between individual relative telomere values and absolute neutrophil counts. There was no difference in expression of telomerase reverse transcriptase in peripheral blood mononuclear cells between patients and controls but telomerase activity was identified at a significantly higher frequency in controls than in patients. No correlation was found between telomerase activity or telomerase reverse transcriptase expression and relative telomere lengths of peripheral blood mononuclear cells. CONCLUSIONS: Patients with chronic idiopathic neutropenia display age-inappropriate telomere shortening of peripheral blood cells and low telomerase activity in peripheral blood mononuclear cells. A compensatory increased proliferation of bone marrow hematopoietic progenitor cells in association with lymphocyte replicative exhaustion probably account for these abnormalities.


Subject(s)
Granulocytes/pathology , Leukocytes, Mononuclear/pathology , Neutropenia/genetics , Neutropenia/pathology , Telomere Shortening , Adult , Aged , Case-Control Studies , Cohort Studies , DNA/genetics , Female , Humans , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Telomerase/genetics
7.
Curr Stem Cell Res Ther ; 6(2): 122-30, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20528751

ABSTRACT

Myelodysplastic syndromes comprise a heterogeneous group of clonal hematopoietic stem cell malignancies characterized by ineffective bone marrow (BM) hematopoiesis, peripheral blood cytopenias and substantial risk for progression to acute myeloid leukemia. It is generally accepted that myelodysplastic syndromes originate as a result of multistep leukemogenesis, implicating genetic, epigenetic and immune-mediated alterations of an early hematopoietic stem cell. However, alterations in the BM microenvironment in terms of abnormal hematopoietic-to-stromal cell interactions, relative deficiency of hematopoietic growth factors and aberrant release of inhibitors may also have a role in myelodysplastic syndrome (MDS) pathogenesis. The possible involvement of the BM mesenchymal stem cells (MSC) in the pathogenetic/pathophysiologic process of MDS has been recently studied but existing data on MSCs' cytogenetic and functional integrity are controversial. Notably, in our study we did not find any significant quantitative or qualitative deficits in MDS-derived MSCs. As no conclusive data on the characteristics of BM MSCs have been reported so far, future studies should aim at elucidating whether BM MSCs belong primarily to the abnormal clone or whether they are indirectly damaged and whether they might be safely used for therapeutic purposes in MDS patients. This article aims to give an overview of the current state of the art on the quantitative, functional, immunoregulatory and cytogenetic properties of BM MSCs in MDS.


Subject(s)
Bone Marrow/pathology , Mesenchymal Stem Cells/pathology , Myelodysplastic Syndromes/pathology , Animals , Humans
8.
Stem Cells Dev ; 20(8): 1309-18, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21047210

ABSTRACT

Chronic idiopathic neutropenia (CIN) is a granulopoiesis disorder associated with an inhibitory bone marrow (BM) microenvironment consisting of activated T-lymphocytes and pro-inflammatory mediators. In this study, we investigated the possible involvement of BM mesenchymal stem cells (MSCs) in the pathophysiology of CIN by assessing the frequency and function of BM MSCs in terms of the proliferative/clonogenic characteristics, the differentiation capacity, the potential to produce pro-inflammatory cytokines, and the ability to suppress T-cell proliferation. The frequency, differentiation capacity toward adipocytes, chondrocytes, or osteoblasts, and immunosuppressive potential to inhibit mitogen-induced T-cell proliferation did not differ significantly between patient (n = 14) and normal (n = 21) MSCs. Tumor necrosis factor-α, interleukin-1ß, and interleukin-6 levels in MSC supernatants did not differ significantly between patients and controls; however, transforming growth factor (TGF)-ß1 levels were significantly elevated in patients, particularly in those displaying the -509C/T TGF-ß1 polymorphism. Patient MSCs displayed defective proliferative/clonogenic potential, which could not be attributed to altered cellular survival characteristics or to increased TGF-ß1 production as TGF-ß1 neutralization did not restore the impaired colony formation by patient MSCs. We conclude that although BM MSCs do not exert a significant role in the immune deregulation associated with CIN, they contribute to the inhibitory microenvironment by overproducing TGF-ß1, at least in patients displaying the -509C/T polymorphism.


Subject(s)
Bone Marrow/immunology , Mesenchymal Stem Cells/immunology , Neutropenia/immunology , Neutropenia/metabolism , Transforming Growth Factor beta1/immunology , Adolescent , Adult , Aged , Bone Marrow/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytokines/biosynthesis , Female , Humans , Lymphocyte Activation/immunology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Middle Aged , Polymorphism, Restriction Fragment Length , Polymorphism, Single Nucleotide , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Transforming Growth Factor beta1/biosynthesis
9.
Respir Med ; 104(10): 1535-42, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20483575

ABSTRACT

BACKGROUND: Experimental data have provided evidence that progenitor cells of bone marrow (BM) origin may play a role in the fibrogenic process of the lung. OBJECTIVE: To probe the possible involvement of BM mesenchymal stem cells (MSCs) in the pathophysiology of Idiopathic Pulmonary Fibrosis (IPF) by investigating the molecular profile of these cells. DESIGN: BM MSCs were studied in 10 IPF patients and 10 healthy controls. MSCs were identified by their immunophenotypic characteristics and their potential to differentiate towards adipocytes/osteocytes/chondrocytes. We evaluated the mRNA expression of genes involved in the lung injury of IPF, namely the vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), transforming growth factor beta-1 (TGF-beta1) and the axis stromal-cell-derived factor-1 (SDF-1)/CXCR4 in BM MSCs using quantitative RT-PCR. RESULTS: The BM MSCs of IPF patients displayed normal immunophenotypic characteristics and differentiation potential. No statistically significant difference was found between patients and controls in VEGF and FGF mRNA expression. TGF-beta1 was not expressed in either patients or controls. A significant increase in SDF-1-TR1 and CXCR4 mRNA expression was detected in IPF patients (1.6 x 10(25) +/- 1.2 x 10(25) and 3.1 x 10(7) +/- 3.1 x 10(7), respectively) compared to controls (0.32 x 10(25) +/- 0.07 x 10(25) and 1.67 x 10(7) +/- 0.30 x 10(7), respectively) (p = 0.001 and p = 0.001, respectively) whereas SDF-1 levels in MSC supernatants were similar in patients and controls. CONCLUSIONS: The increased CXCR4 expression by patient MSCs suggests that the BM is probably implicated in the pathophysiology of IPF by mobilizing MSCs in response to or preceding lung injury. The potential role of BM MSCs in IPF is another interesting field for further investigation.


Subject(s)
Fibroblast Growth Factors/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Mesenchymal Stem Cells/metabolism , Adult , Aged , Female , Fibroblast Growth Factors/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Humans , Idiopathic Pulmonary Fibrosis/pathology , Male , Mesenchymal Stem Cells/cytology , Middle Aged , Phenotype , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/metabolism
10.
Stem Cells Dev ; 19(7): 1043-54, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19788374

ABSTRACT

Defective hematopoiesis supporting capacity of bone marrow (BM) stroma has been implicated in the pathophysiology of myelodysplastic syndromes (MDS). The aim of this study is to explore whether the BM stroma progenitors, namely the mesenchymal stem cells (MSCs), are primarily affected in MDS by evaluating the reserves, the functional properties, as well as the cytogenetic characteristics, in comparison to BM hematopoietic cells, in patients with de novo MDS (n = 13). The number, differentiation potential toward adipocytes/chondrocytes/osteoblasts and immunosuppressive function in terms of inhibition of mitogen-induced T-cell proliferation did not differ significantly between patient and normal (n = 20) MSCs. Patient MSCs did not show any aberrations in the production of proinflammatory or growth-promoting cytokines and did not harbor the cytogenetic abnormalities present in hematopoietic cells. Occasional patient and normal MSC cultures, however, developed irrelevant chromosomal alterations (trisomies 5 and 7) with uncertain pathophysiologic significance. Compared to controls, patient MSCs displayed impaired proliferative and clonogenic potential through passages that might represent a nonspecific abnormality associated with the chronic inflammatory process present in patients' BM. These data suggest that BM MSCs from MDS patients do not belong to the abnormal clone and do not represent the main cellular source contributing to the inflammatory marrow microenvironment.


Subject(s)
Bone Marrow Cells/physiology , Chromosome Aberrations , Hematopoietic Stem Cells/physiology , Mesenchymal Stem Cells/physiology , Myelodysplastic Syndromes/physiopathology , Aged , Aged, 80 and over , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Immunophenotyping , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Middle Aged , Myelodysplastic Syndromes/pathology , T-Lymphocytes/physiology
11.
Curr Stem Cell Res Ther ; 4(1): 61-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19149631

ABSTRACT

Mesenchymal stromal cells (MSC) isolated from a variety of adult tissues including the bone marrow (BM), have the capacity to differentiate into different cell types such as bone and cartilage and have therefore attracted scientific interest as potential therapeutic tools for tissue repair. MSC display also immunosuppressive and anti-inflammatory properties and their putative therapeutic role in a variety of inflammatory autoimmune diseases is currently under investigation. Joint destruction, caused by persistent inflammation, renders rheumatoid arthritis (RA) a possible clinical target for cartilage and bone repair using BM MSCs for their tissue repair and immunoregulatory effects. A number of studies, based mainly on experimental animal models, have recently provided interesting data on the potential of BM-MSCs to suppress local inflammation and tissue damage in RA whereas tissue engineering and cell-scaffold technology represents an emerging field of research. This review deals with the biological repair/regeneration of joint tissues in RA via MSC-based therapies. In view of the current interest in the autologous usage of BM MSC in RA, all available data on the biological properties of patient MSCs including the immunoregulatory characteristics, differentiation capacity towards osteocytes/chondrocytes, clonogenic/proliferative potential and molecular/protein profile and the possible influence of the RA milieu will be also summarized.


Subject(s)
Arthritis, Rheumatoid/therapy , Mesenchymal Stem Cells/metabolism , Osteocytes/pathology , Stem Cell Transplantation , Stromal Cells/metabolism , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/therapy , Arthritis, Rheumatoid/pathology , Bone Regeneration/physiology , Cell Differentiation , Humans , Immunosuppression Therapy , Inflammation , Mesenchymal Stem Cells/pathology , Mice , Osteocytes/metabolism , Stromal Cells/pathology , Tissue Engineering
12.
Tissue Eng Part C Methods ; 14(4): 333-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18800875

ABSTRACT

Bone marrow (BM) mesenchymal stem cells (MSCs) represent an interesting field of research for their in vitro properties and the in vivo therapeutic applications. In the present study, we compared the clonogenic and differentiation capacity of MSCs present in three BM-derived populations-namely, the CD105(+)/CD45(-) cells, the glycophorin A (GlycoA)(-)/CD45(-) cells, and the BM mononuclear cells (BMMCs)-by growing/expanding clones from single colony-forming unit fibroblasts (CFU-F). We also quantified the Oct-4 and Nanog mRNA in the CD105(+)/CD45(-) and GlycoA(-)/CD45(-) cells to define the fraction containing more immature MSCs. We found that basic-fibroblast growth factor (bFGF) favors the long-term survival and growth of the more immature MSCs but has no significant effect on MSC clonogenic potential. CFU-F number and clone recovery were higher in CD105(+)/CD45(-) compared to GlycoA(-)/CD45(-) (p < 0.0001 and p = 0.0364, respectively) cells or BMMCs (p < 0.0001 and p = 0.0007, respectively). The relative mRNA expression of Oct-4 and Nanog was significantly increased in CD105(+)/CD45(-) compared to GlycoA(-)/CD45(-) cells (p < 0.0001 and p < 0.0001, respectively). No significant difference was found in the immunophenotypic characteristics and differentiation potential of clones derived from all three cellular sources. These data suggest that the CD105(+)/CD45(-) BM cell fraction is enriched in immature MSCs and, accordingly, represents an appropriate source for MSC culture initiation.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Antigens, CD/biosynthesis , Cell Differentiation , Cells, Cultured , Endoglin , Gene Expression Regulation , Homeodomain Proteins/biosynthesis , Humans , Leukocyte Common Antigens/biosynthesis , Nanog Homeobox Protein , Octamer Transcription Factor-3/biosynthesis , Phenotype , RNA, Messenger/metabolism , Receptors, Cell Surface/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...