Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Cell Death Dis ; 9(6): 695, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29891848

ABSTRACT

Fas-L is a TNF family member known to trigger cell death. It has recently become evident that Fas-L can transduce also non-apoptotic signals. Mesenchymal stem cells (MSCs) are multipotent cells that are derived from various adult tissues. Although MSCs from different tissues display common properties they also display tissue-specific characteristics. Previous works have demonstrated massive apoptosis following Fas-L treatment of bone marrow-derived MSCs both in vitro and following their administration in vivo. We therefore set to examine Fas-L-induced responses in adipose-derived stem cells (ASCs). Human ASCs were isolated from lipoaspirates and their reactivity to Fas-L treatment was examined. ASCs responded to Fas-L by simultaneous apoptosis and proliferation, which yielded a net doubling of cell quantities and a phenotypic shift, including reduced expression of CD105 and increased expression of CD73, in association with increased bone differentiation potential. Treatment of freshly isolated ASCs led to an increase in large colony forming unit fibroblasts, likely produced by early stem cell progenitor cells. Fas-L-induced apoptosis and proliferation signaling were found to be independent as caspase inhibition attenuated Fas-L-induced apoptosis without impacting proliferation, whereas inhibition of PI3K and MEK, but not of JNK, attenuated Fas-L-dependent proliferation, but not apoptosis. Thus, Fas-L signaling in ASCs leads to their expansion and phenotypic shift toward a more potent stem cell state. We speculate that these reactions ensure the survival of ASC progenitor cells encountering Fas-L-enriched environments during tissue damage and inflammation and may also enhance ASC survival following their administration in vivo.


Subject(s)
Adipose Tissue/cytology , Fas Ligand Protein/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Antigens, CD/metabolism , Apoptosis/drug effects , Bone and Bones/cytology , Caspase Inhibitors/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Colony-Forming Units Assay , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism
2.
Oncotarget ; 7(41): 67061-67070, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27563813

ABSTRACT

INTRODUCTION: Multiple myeloma is still incurable in most cases. Polyclonal anti T lymphocyte globulins (ATG) have been reported to kill human myeloma cells in vitro and in mouse models. METHODS: Anti-human-myeloma globulins (AMG) were produced by immunizing rabbits with human myeloma cell lines RPMI-8226 (AMG-8226) or KMS-12-BM (AMG-12-BM). Cytotoxicity of the polyclonal antibodies was analyzed in vitro and in a xenograft NOD-SCID mouse model. RESULTS: Both AMG had stronger cytotoxicity against myeloma cells compared to ATG. In primary T cells, AMG-8226 showed greater complement-dependent cytotoxicity (CDC) than ATG, whereas complement-independent cytotoxicity did not differ. Effects on non-hematopoietic cell lines were also similar. Competitive blocking assays revealed fourfold more antibodies against CD38 in AMG-8226 compared to ATG. Low concentrations of AMG-8226 and ATG increased ADCC. At higher concentrations, ATG inhibited ADCC more potently than AMG-8226. Combinations of ATG and AMG-8226 with melphalan or bortezomib showed additive to synergistic cytotoxicity on myeloma cells. The cytotoxic effects of AMG and ATG were confirmed in the xenograft NOD-SCID mouse model. CONCLUSION: Our data show more potent antimyeloma effects of AMG compared to ATG. These results lay the ground for the development of polyclonal antibodies for the treatment of multiple myeloma.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/drug effects , Antilymphocyte Serum/pharmacology , Antineoplastic Agents/pharmacology , Multiple Myeloma , Animals , Cell Line, Tumor , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Xenograft Model Antitumor Assays
3.
Stem Cell Rev Rep ; 11(6): 826-40, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26250539

ABSTRACT

Mesenchymal stem cells (MSCs) serve as supporting and regulatory cells, by providing tissues with multiple factors and are also known for their immunosuppressive capabilities. Our laboratory had previously shown that MSCs expressed toll-like receptor (TLR) 2 and are activated by its ligand Pam3Cys. TLR2 is an important component of the innate immune system, as it recognizes bacterial lipopeptides, thus priming a pro-inflammatory immune response. This study showed that Pam3Cys attached extensively to cells of both wild-type and TLR2 deficient cultured MSCs, thus, independently of TLR2. The TLR2 independent binding occurred through the adsorption of the palmitoyl moieties of Pam3Cys. It was further showed that Pam3Cys was transferred from cultured MSCs to immune cells. Moreover, Pam3Cys provided to the immune cells induced a pro-inflammatory response in vitro and in vivo. Overall, it is demonstrated herein that a TLR2 ligand bound to MSCs also through a TLR2 independent mechanism. Furthermore, the ligand incorporated by MSCs is subsequently released to stimulate an immune response both in vitro and in vivo. It is thus suggested that during bacterial infection, stromal cells may retain a reservoir of the TLR2 ligands, in a long-term manner, and release them slowly to maintain an immune response.


Subject(s)
Lipoproteins/metabolism , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Toll-Like Receptor 2/agonists , Animals , Cell Proliferation , Cells, Cultured , Female , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology
4.
Biochim Biophys Acta ; 1849(4): 371-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25038585

ABSTRACT

Over a decade of intensive investigation of the possible plasticity of mammalian cells has eventually substantiated that mammalian species are endowed with a remarkable capacity to change mature cell fates. We review below the evidence for the occurrence of processes such as dedifferentiation and transdifferentiation within mammalian tissues in vivo, and in cells removed from their protective microenvironment and seeded in culture under conditions poorly resembling their physiological state in situ. Overall, these studies point to one major conclusion: stressful conditions, whether due to in vivo tissue damage or otherwise to isolation of cells from their in vivo restrictive niches, lead to extreme fate changes. Some examples of dedifferentiation are discussed in detail showing that rare cells within the population tend to turn back into less mature ones due to severe cell damage. It is proposed that cell stress, mechanistically sensed by isolation from neighboring cells, leads to dedifferentiation, in an attempt to build a new stem cell reservoir for subsequent regeneration of the damaged tissue. This article is part of a Special Issue entitled: Stress as a fundamental theme in cell plasticity.


Subject(s)
Cell Dedifferentiation , Cell Physiological Phenomena , Stress, Physiological/physiology , Animals , Cellular Reprogramming , Humans , Mammals
5.
Stem Cell Res Ther ; 5(6): 139, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25519840

ABSTRACT

INTRODUCTION: Mesenchymal stem cells (MSCs) are multipotent and have been derived from various tissues. Although MSCs share many basic features, they often display subtle tissue specific differences. We previously demonstrated that bone marrow (BM) MSCs frequently become polyploid in culture. This tendency was mediated by a reduction in the expression of H19 long non-coding RNA during the transition from a diploid to a polyploid state. METHODS: MSCs were derived from both BM and adipose tissue of mice and expanded under normoxic and hypoxic culture conditions. Cells were stained by propidium iodide and their ploidy was evaluated by FACS. Gene expression of independent MSC preparations was compared by quantitative real time PCR and protein expression levels by Western blot analysis. p53 silencing in MSCs was performed by a specific small hairpin RNA (shRNA). RESULTS: We set to examine whether genomic instability is common to MSCs originating from different tissues. It is demonstrated that adipose derived MSCs (ASCs) tend to remain diploid during culture while a vast majority of BM MSCs become polyploid. The diploid phenotype of ASCs is correlated with reduced H19 expression compared to BM MSCs. Under hypoxic conditions (3% oxygen) both ASCs and BM MSCs demonstrate increased RNA expression of H19 and Vascular endothelial growth factor A. Importantly, ASC gene expression is significantly less variable than BM MSCs under both oxygen conditions, indicating to their superior homogeneity. Gene expression analysis revealed that p53 target genes, often induced by DNA damage, are up-regulated in ASCs under basal conditions. However, p53 activation following treatment with DNA damaging agents was strongly elevated in BM MSCs compared to ASCs. We found that p53 is involved in maintaining the stable diploid state of ASCs as p53 shRNA induced ploidy changes in ASCs but not in BM MSCs. CONCLUSIONS: The increased genomic stability of murine ASCs together with their lower H19 expression and relative homogeneity suggest a tissue specific higher stability of ASCs compared to BM MSCs, possibly due to higher activity of p53. The tissue specific differences between MSCs from a different tissue source may have important consequences on the use of various MSCs both in vitro and in vivo.


Subject(s)
Adipose Tissue/cytology , Genomic Instability , Mesenchymal Stem Cells/metabolism , RNA, Long Noncoding/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cells, Cultured , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Organ Specificity , Ploidies , Tumor Suppressor Protein p53/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
6.
Stem Cells ; 32(8): 2008-20, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24715711

ABSTRACT

Mesenchymal stromal cell populations include a fraction, termed mesenchymal stem cells, exhibiting multipotency. Other cells within this population possess a lesser differentiation range. This was assumed to be due to a mesenchymal cellular cascade topped by a multipotent cell, which gives rise to progeny with diminishing differentiation potentials. Here, we show that mesenchymal cells, a priori exhibiting a limited differentiation potential, may gain new capacities and become multipotent following single-cell isolation. These fate changes were accompanied by upregulation of differentiation promoting genes, many of which also became H4K20me1 methylated. Early events in the process included TGFß and Wnt modulation, and downregulation of hypoxia signaling. Indeed, hypoxic conditions inhibited the observed cell changes. Overall, cell isolation from neighboring partners caused major molecular changes and particularly, a newly established epigenetic state, ultimately leading to the acquisition of new differentiation potentials and an altered cell fate.


Subject(s)
Cell Differentiation/physiology , Cell Separation , Mesenchymal Stem Cells/cytology , Animals , Bone Marrow Cells/cytology , Cell Culture Techniques , Cell Lineage , Chromatin Immunoprecipitation , Clone Cells/cytology , Flow Cytometry , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
7.
Stem Cell Rev Rep ; 10(3): 376-88, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24664302

ABSTRACT

The outstanding heterogeneity of stem cell populations is a major obstacle on the way to their clinical application. It is therefore paramount to identify the molecular mechanisms that underlay this heterogeneity. Individually derived bone marrow mesenchymal stromal cells (MSCs) preparations, studied here, diverged markedly in various properties, despite of being all tripotent in their differentiation potential. Microarray analysis showed that MSC diversity is evident also in highly variable gene expression patterns. Differentially expressed genes were significantly enriched in toll-like receptors (TLRs) and differentiation pathways. Marked differences were observed in LPS binding protein (LBP) and transforming growth factor (TGF)ß1 expression. These differences correlated with MSC functionality. Therefore, the possible contribution of these molecules to MSC diversity was examined. In the TLR signaling pathway, LBP levels predicted the ability of specific MSCs to secrete interleukin (IL)-6 in response to LPS. A relatively higher expression of TGFß1 endowed MSCs with a capacity to respond to IL-1ß by reduced osteogenic differentiation. This study thus demonstrates major diversity within MSC isolates, which appears early on following derivation and persists following long-term culture. MSC heterogeneity results from highly variable transcriptome. Differential expression of LBP and TGFß1, along with other genes, in different MSC preparations, produces the variable responses to external stimuli.


Subject(s)
Acute-Phase Proteins/metabolism , Carrier Proteins/metabolism , Interleukin-1beta/physiology , Membrane Glycoproteins/metabolism , Mesenchymal Stem Cells/metabolism , Toll-Like Receptors/agonists , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Differentiation , Cells, Cultured , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Mesenchymal Stem Cells/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Toll-Like Receptors/metabolism , Transcriptome
8.
Stem Cells Dev ; 23(6): 676-86, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24200242

ABSTRACT

Umbilical cord blood (UCB) is a good source of hematopoietic progenitors with increasing implementation in the clinical transplant setting. This study evaluates the molecular mechanisms of progenitor resistance to apoptosis triggered by Fas cross-linking. CD34(+) and lineage-negative progenitors survive short-term ex vivo incubation and are not induced into apoptosis by Fas cross-linking. Furthermore, brief exposure of UCB cells to Fas-ligand for 24-48 h does not impair quantitative severe combine immune deficiency (SCID) reconstitution activity and appears to foster myelomonocyte reconstitution. The transcriptome of Fas receptor-positive CD34(+) cells that survived an apoptotic challenge showed significant transcriptional upregulation of caspase-8, mucosa-associated lymphoid tissue lymphoma translocation gene-1 (MALT1), HtrA2, and GSK3ß in addition to higher levels of c-FLICE inhibitory protein (FLIP), Bcl-2, and cytosolic inhibitor of apoptosis protein (cIAP) in all Fas-positive cells. Most prominent is the transcriptional upregulation of several key components the NFκB1 pathway including the membrane receptors TGF-ß, interleukin-1 (IL-1), and TCR, the associated factor TNF receptor-associated factor-6 (TRAF6), and the converting enzymes TGF-ß-activated kinase-1 (TAK1), double-stranded RNA-activated protein kinase (PKR), and α-catalytic subunit of IκB kinase (IKKα), that promote activation and nuclear translocation of this transcription factor. These data indicate that hematopoietic progenitors are not insensitive to apoptosis but are actively shielded from the extrinsic and intrinsic apoptotic pathways. This may occur through inherent transcriptional upregulation of the entire NFκB pathway in the presence of competent apoptotic signaling.


Subject(s)
Apoptosis , Fas Ligand Protein/metabolism , Hematopoietic Stem Cells/cytology , NF-kappa B/metabolism , Signal Transduction/drug effects , Caspases/metabolism , Cells, Cultured , Hematopoietic Stem Cells/metabolism , Humans , NF-kappa B/drug effects , Transcription, Genetic , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , fas Receptor/metabolism
9.
J Am Heart Assoc ; 2(5): e000253, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-24080908

ABSTRACT

BACKGROUND: Human mesenchymal stromal cells (hMSCs) from adipose cardiac tissue have attracted considerable interest in regard to cell-based therapies. We aimed to test the hypothesis that hMSCs from the heart and epicardial fat would be better cells for infarct repair. METHODS AND RESULTS: We isolated and grew hMSCs from patients with ischemic heart disease from 4 locations: epicardial fat, pericardial fat, subcutaneous fat, and the right atrium. Significantly, hMSCs from the right atrium and epicardial fat secreted the highest amounts of trophic and inflammatory cytokines, while hMSCs from pericardial and subcutaneous fat secreted the lowest. Relative expression of inflammation- and fibrosis-related genes was considerably higher in hMSCs from the right atrium and epicardial fat than in subcutaneous fat hMSCs. To determine the functional effects of hMSCs, we allocated rats to hMSC transplantation 7 days after myocardial infarction. Atrial hMSCs induced greatest infarct vascularization as well as highest inflammation score 27 days after transplantation. Surprisingly, cardiac dysfunction was worst after transplantation of hMSCs from atrium and epicardial fat and minimal after transplantation of hMSCs from subcutaneous fat. These findings were confirmed by using hMSC transplantation in immunocompromised mice after myocardial infarction. Notably, there was a correlation between tumor necrosis factor-α secretion from hMSCs and posttransplantation left ventricular remodeling and dysfunction. CONCLUSIONS: Because of their proinflammatory properties, hMSCs from the right atrium and epicardial fat of cardiac patients could impair heart function after myocardial infarction. Our findings might be relevant to autologous mesenchymal stromal cell therapy and development and progression of ischemic heart disease.


Subject(s)
Mesenchymal Stem Cells , Myocardium/cytology , Pericardium/cytology , Animals , Cells, Cultured , Heart , Humans , Mesenchymal Stem Cell Transplantation , Mice , Myocardial Infarction/surgery , Myocardial Ischemia/etiology
10.
Cancer Res ; 72(24): 6403-13, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23047867

ABSTRACT

Mesenchymal stromal cells (MSC) are used extensively in clinical trials; however, the possibility that MSCs have a potential for malignant transformation was raised. We examined the genomic stability versus the tumor-forming capacity of multiple mouse MSCs. Murine MSCs have been shown to be less stable and more prone to malignant transformation than their human counterparts. A large series of independently isolated MSC populations exhibited low tumorigenic potential under syngeneic conditions, which increased in immunocompromised animals. Unexpectedly, higher ploidy correlated with reduced tumor-forming capacity. Furthermore, in both cultured MSCs and primary hepatocytes, polyploidization was associated with a dramatic decrease in the expression of the long noncoding RNA H19. Direct knockdown of H19 expression in diploid cells resulted in acquisition of polyploid cell traits. Moreover, artificial tetraploidization of diploid cancer cells led to a reduction of H19 levels, as well as to an attenuation of the tumorigenic potential. Polyploidy might therefore serve as a protective mechanism aimed at reducing malignant transformation through the involvement of the H19 regulatory long noncoding RNA.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Silencing/physiology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Polyploidy , RNA, Long Noncoding/genetics , Animals , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Genomic Instability , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasms/genetics , RNA, Long Noncoding/antagonists & inhibitors
11.
Stem Cell Rev Rep ; 8(2): 343-54, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21881833

ABSTRACT

In human multiple myeloma (MM), the tumor cells exhibit strict dependence on bone marrow (BM) stromal elements. It has been suggested that, in turn, MM cells modify multipotent stromal cells (MSCs), diverting them to support the myeloma. We investigated MM-derived MSCs by comparing their toll-like receptor (TLR) responses to those of MSCs derived from healthy controls. We now report that MM-derived MSCs manifested intact proliferation responses and IL-6 secretion and their adipose and osteogenic differentiation responses to TLR ligands were also similar to those of healthy controls, ranging from augmentation to inhibition. However, MM-derived MSCs were found to be defective in IL-8 secretion and ERK1/2 phosphorylation following TLR-2 activation. Moreover, MM-derived MSCs failed to respond to EGF by elevation of ERK1/2 phosphorylation. The persistence of these changes in extensively cultured MM-derived MSCs, suggests that these cells are stably, if not irreversibly modified.


Subject(s)
Epidermal Growth Factor/pharmacology , Mesenchymal Stem Cells/pathology , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Toll-Like Receptors/metabolism , Adult , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Interleukin-8/metabolism , Kinetics , Ligands , Lipoproteins/pharmacology , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Middle Aged , Phosphorylation/drug effects
12.
Mol Immunol ; 49(1-2): 239-52, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21943707

ABSTRACT

The existence of incomplete T cell receptor (TCR) mRNA forms, including germline transcripts and products of unfruitful TCR rearrangements, has long been known. However, it is unclear whether these molecules are functional. We have previously shown that T cells also contain truncated TCRß peptides that lack the N-terminal part and contain C-terminus sequences. These partial forms of TCRß, target the mitochondrion and induce apoptosis, exhibiting a novel mode of TCR mediated cell death. Here we aimed at analyzing the minimal TCR sequences that direct the peptide to the mitochondrion. It is shown that truncated TCRß, targets mitochondria and induces mitochondrial perinuclear clustering, in both monkey COS-7 and human 293 cells. These phenomena are mediated by the C-terminus of the molecule. Whereas the positively charged amino acids flanking the transmembrane domain (TMD) of TCRß are beneficial for this process, they are not essential. Indeed, the isolated TMD of TCRß serves as a sufficient mitochondrial targeting sequence. These results indicate that any given partial form of TCRß, that contains the TMD, is bound to be sequestered by the mitochondrion. This may assure that incomplete TCR forms would not interfere with correct TCR complex formation.


Subject(s)
Apoptosis/physiology , Mitochondria/metabolism , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Amino Acid Sequence , Animals , Blotting, Western , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Protein Structure, Tertiary , Transfection
13.
Stem Cell Rev Rep ; 7(3): 560-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21437576

ABSTRACT

Cultured mesenchymal stromal cell (MSC) populations are best characterized by the capacity of some cells within this population to differentiate into mesodermal derivatives such as osteoblasts, chondrocytes and adipocytes. However, this progenitor property is not shared by all cells within the MSC population. Furthermore, MSCs exhibit variability in their phenotypes, including proliferation capacity, expression of cell surface markers and ability to secrete cytokines. These facts raise three major questions: (1) Does the in vitro observed variability reflect the existence of MSC subsets in vivo? (2) What is the molecular basis of the in vitro observed heterogeneity? and (3) What is the biological significance of this variability? This review considers the possibility that the variable nature of MSC populations contributes to the capacity of adult mammalian tissues to adapt to varying microenvironmental demands.


Subject(s)
Mesenchymal Stem Cells/cytology , Stromal Cells/cytology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Culture Techniques , Cell Differentiation/physiology , Cells, Cultured , Humans , Mesenchymal Stem Cells/physiology , Stromal Cells/physiology
14.
Med Sci (Paris) ; 27(3): 303-7, 2011 Mar.
Article in French | MEDLINE | ID: mdl-21447304

ABSTRACT

Stem cells are a non-coherent group of cells that have little in common. Despite the fact that these diverse cell types are regarded as belonging to the same category, they do not share molecular markers. The definition of stemness is therefore descriptive, relating to potentials of the cells rather than to the actual properties that they harbor. This situation is confusing and causes unnecessary debates in this field of research. It is therefore of paramount importance to find a new, molecular definition of stemness, that would consist of the cellular machineries which constitute the stem cell state.


Subject(s)
Stem Cells/physiology , Animals , Cell Dedifferentiation , Cell Differentiation , Cell Transdifferentiation , Molecular Biology
15.
Stem Cell Rev Rep ; 7(3): 488-93, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21279479

ABSTRACT

Differentiation cascades are arranged hierarchically; stem cells positioned at the top of the hierarchy generate committed progenitors that, in turn, proliferate and further differentiate stepwise into mature progeny. This rigid, irreversible structure ensures the phenotypic stability of adult tissues. However, such rigidity may be problematic under conditions of tissue damage when reconstitution is required. Although it may seem unlikely that the restrictions on changes in cell phenotypes would be lifted to enable tissue reconstitution, it is nevertheless possible that mammalian tissues are endowed with sufficient flexibility to enable their adaptation to extreme conditions.


Subject(s)
Cell Dedifferentiation/physiology , Mammals , Stress, Physiological , Animals , Cell Differentiation , Homeostasis , Humans , Neoplasms/etiology , Stem Cells/physiology
16.
Cell Stem Cell ; 8(1): 10-1, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21211778

ABSTRACT

Mesenchymal stromal cells (MSCs) are capable of differentiating into bone-forming osteoblasts. A recent Nature Medicine study (Medici et al., 2010) shows that the mislocalized bone in the human disease fibrodisplasia ossificans progressiva (FOP) originates from vascular endothelium that gives rise to MSCs.

17.
Cancer Microenviron ; 3(1): 15-28, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-21209772

ABSTRACT

Multiple myeloma cells are reminiscent of hemopoietic stem cells in their strict dependence upon the bone marrow microenvironment. However, from all other points of view, multiple myeloma cells differ markedly from stem cells. The cells possess a mature phenotype and secrete antibodies, and have thus made the whole journey to maturity, while maintaining a tumor phenotype. Not much credence was given to the possibility that the bulk of plasma-like multiple myeloma tumor cells is generated from tumor-initiating cells. Although interleukin-6 is a major contributor to the formation of the tumor's microenvironment in multiple myeloma, it is not a major factor within hemopoietic stem cell niches. The bone marrow niche for myeloma cells includes the activity of inflammatory cytokines released through osteoclastogenesis. These permit maintenance of myeloma cells within the bone marrow. In contrast, osteoclastogenesis constitutes a signal that drives hemopoietic stem cells away from their bone marrow niches. The properties of the bone marrow microenvironment, which supports myeloma cell maintenance and proliferation, is therefore markedly different from the characteristics of the hemopoietic stem cell niche. Thus, multiple myeloma presents an example of a hemopoietic tumor microenvironment that does not resemble the corresponding stem cell renewal niche.

18.
Exp Cell Res ; 315(11): 1904-13, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19328780

ABSTRACT

Multiple myeloma is characterized by the malignant growth of immunoglobulin producing plasma cells, predominantly in the bone marrow. The effects of primary human mesenchymal stromal cells on the differentiation phenotype of multiple myeloma cells were studied by co-culture experiments. The incubation of multiple myeloma cells with bone marrow-derived mesenchymal stromal cells resulted in significant reduction of the expression of the predominant plasma cell differentiation markers CD38 and CD138, and cell surface immunoglobulin light chain. While the down-regulation of CD138 by stromal cells was completely dependent on their adhesive interactions with the multiple myeloma cells, interleukin-6 induced specific down-regulation of CD38. Mesenchymal stromal cells or their conditioned media inhibited the growth of multiple myeloma cell line, thereby reducing the overall amounts of secreted light chains. Analysis of primary multiple myeloma bone marrow samples reveled that the expression of CD38 on multiple myeloma cells was not affected by adhesive interactions. The ex vivo propagation of primary multiple myeloma cells resulted in significant increase in their differentiation markers. Overall, the data indicate that the bone marrow-derived mesenchymal stromal cells revert multiple myeloma cells to less differentiated phenotype by the combined activities of adhesive interactions and interleukin-6.


Subject(s)
Interleukin-6/physiology , Mesoderm/cytology , Mesoderm/physiology , Multiple Myeloma/pathology , Multiple Myeloma/physiopathology , Stromal Cells/cytology , Stromal Cells/physiology , ADP-ribosyl Cyclase 1/metabolism , Antibodies, Monoclonal/pharmacology , Biomarkers, Tumor/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Adhesion , Cell Differentiation/drug effects , Cell Line , Cell Line, Tumor , Coculture Techniques , Down-Regulation , Humans , Immunoglobulin kappa-Chains/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/pharmacology , Membrane Glycoproteins/metabolism , Mesoderm/drug effects , Neutralization Tests , Phenotype , Plasma Cells/cytology , Plasma Cells/physiology , Recombinant Proteins/pharmacology , Stromal Cells/drug effects , Syndecan-1/metabolism
19.
Blood ; 113(15): 3530-41, 2009 Apr 09.
Article in English | MEDLINE | ID: mdl-18931338

ABSTRACT

The default pathway of cell-surface T-cell receptor (TCR) complex formation, and the subsequent transport to the membrane, is thought to entail endoplasmic reticulum (ER) localization followed by proteasome degradation of the unassembled chains. We show herein an alternative pathway: short, incomplete peptide versions of TCRbeta naturally occur in the thymus. Such peptides, which have minimally lost the leader sequence or have been massively truncated, leaving only the very C terminus intact, are sorted preferentially to the mitochondrion. As a consequence of the mitochondrial localization, apoptotic cell death is induced. Structure function analysis showed that both the specific localization and induction of apoptosis depend on the transmembrane domain (TMD) and associated residues at the COOH-terminus of TCR. Truncated forms of TCR, such as the short peptides that we detected in the thymus, may be products of protein degradation within thymocytes. Alternatively, they may occur through the translation of truncated mRNAs resulting from unfruitful rearrangement or from germline transcription. It is proposed that mitochondria serve as a subcellular sequestration site for incomplete TCR molecules.


Subject(s)
Apoptosis/immunology , Mitochondria/metabolism , Protein Transport/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , COS Cells , Cell Survival/immunology , Chlorocebus aethiops , Gene Rearrangement, T-Lymphocyte , Green Fluorescent Proteins/genetics , Immunoglobulin Variable Region/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Protein Structure, Tertiary , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/genetics , Signal Transduction/immunology , Thymus Gland/cytology
20.
PLoS One ; 3(11): e3707, 2008.
Article in English | MEDLINE | ID: mdl-19002260

ABSTRACT

BACKGROUND: The tumor suppressor p53 is an important regulator that controls various cellular networks, including cell differentiation. Interestingly, some studies suggest that p53 facilitates cell differentiation, whereas others claim that it suppresses differentiation. Therefore, it is critical to evaluate whether this inconsistency represents an authentic differential p53 activity manifested in the various differentiation programs. METHODOLOGY/PRINCIPAL FINDINGS: To clarify this important issue, we conducted a comparative study of several mesenchymal differentiation programs. The effects of p53 knockdown or enhanced activity were analyzed in mouse and human mesenchymal cells, representing various stages of several differentiation programs. We found that p53 down-regulated the expression of master differentiation-inducing transcription factors, thereby inhibiting osteogenic, adipogenic and smooth muscle differentiation of multiple mesenchymal cell types. In contrast, p53 is essential for skeletal muscle differentiation and osteogenic re-programming of skeletal muscle committed cells. CONCLUSIONS: These comparative studies suggest that, depending on the specific cell type and the specific differentiation program, p53 may exert a positive or a negative effect, and thus can be referred as a "guardian of differentiation" at large.


Subject(s)
Cell Differentiation/genetics , Mesenchymal Stem Cells/cytology , Tumor Suppressor Protein p53/physiology , Adipogenesis , Animals , Cell Lineage/genetics , Cells, Cultured , Down-Regulation , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Trans-Activators/genetics , Trans-Activators/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...