Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Exp Cell Res ; 403(1): 112586, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33839146

ABSTRACT

Muscle stem cells (MuSC) are considered as a reliable source of therapeutic cells to restore diseased muscles. However in most cases, injected MuSC-derived myoblasts are rapidly destroyed by the host immune response, which impairs the beneficial effect. By contrast, human mesenchymal stromal cells (MSC), have been reported to exhibit potent immune regulatory functions. Thus, we investigated, in vitro, the multipotent differentiation- and immunosuppressive capacities of human myoblasts and compared these features with those of human MSC. Myoblasts shared numerous cell surface markers with MSC, including CD73, CD90, CD105 and CD146. Both cell type were negative for HLA-DR and CD45, CD34 and CD31. CD56, a myogenic marker, was expressed by myoblasts exclusively. Myoblasts displayed multipotent potential capabilities with differentiation in chondrocytes, adipocytes and osteoblasts in vitro. Myoblasts also inhibited allogenic T cell proliferation in vitro in a dose dependent manner, very similarly to MSC. This effect was partly mediated via the activation of indolamine 2,3 dioxygenase enzyme (IDO) after IFNγ exposure. Altogether, these data demonstrate that human myoblasts can differentiate in various mesenchymal linages and exhibit powerful immunosuppressive properties in vitro. Such features may open new therapeutic strategies for MuSC-derived myoblasts.


Subject(s)
Adipocytes/metabolism , Cell Proliferation/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mesenchymal Stem Cells/cytology , Adult , Bone Marrow Cells/cytology , Cell Differentiation/physiology , Chondrocytes/metabolism , Humans , Immunomodulation/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Middle Aged , Myoblasts/metabolism , Osteoblasts/metabolism
2.
Cells ; 9(6)2020 06 21.
Article in English | MEDLINE | ID: mdl-32575851

ABSTRACT

Mesenchymal stromal cells (MSCs) were obtained from human bone marrow and amplified in cultures supplemented with human platelet lysate. Once semi-confluent, cells were seeded in solid collagen scaffolds that were rapidly colonized by the cells generating a 3D cell scaffold. Here, they acquired a myofibroblast phenotype and when exposed to appropriate chemical stimulus, developed tension and cell shortening, similar to those of striated and smooth muscle cells. Myofibroblasts contained a molecular motor-the non-muscle myosin type IIA (NMMIIA) whose crossbridge (CB) kinetics are dramatically slow compared with striated and smooth muscle myosins. Huxley's equations were used to determine the molecular mechanical properties of NMMIIA. Thank to the great number of NMMIIA molecules, we determined the statistical mechanics (SM) of MSCs, using the grand canonical ensemble which made it possible to calculate various thermodynamic entities such as the chemical affinity, statistical entropy, internal energy, thermodynamic flow, thermodynamic force, and entropy production rate. The linear relationship observed between the thermodynamic force and the thermodynamic flow allowed to establish that MSC-laden in collagen scaffolds were in a near-equilibrium stationary state (affinity ≪ RT), MSCs were also seeded in solid collagen scaffolds functionalized with the tripeptide Arg-Gly-Asp (RGD). This induced major changes in NMMIIA SM particularly by increasing the rate of entropy production. In conclusion, collagen scaffolds laden with MSCs can be viewed as a non-muscle contractile bioengineered tissue operating in a near-equilibrium linear regime, whose SM could be substantially modified by the RGD peptide.


Subject(s)
Collagen/metabolism , Mesenchymal Stem Cells/metabolism , Nonmuscle Myosin Type IIA/metabolism , Tissue Scaffolds/chemistry , Cell Differentiation , Humans , Oligopeptides , Thermodynamics
3.
J Orthop Res ; 38(6): 1398-1406, 2020 06.
Article in English | MEDLINE | ID: mdl-31883135

ABSTRACT

The replacement of a native hip joint by a metal-on-metal prosthesis may induce deleterious inflammatory side effects that are associated with the release of wear particles and metal ions. These events are referred to the adverse reaction to metal debris (ARMD) and the adverse local tissue reaction (ALTR). While wear particles seem involved in ARMD, the role of metal ions in ALTR and their impact on myoblasts, located in the prosthesis vicinity, has not been fully identified. To clarify this issue we investigated, using an in vitro culture system, the effect of cobalt and/or chromium ions (Co2+ and/or Cr3+ ) on human myoblast proliferation, cellular differentiation, and inflammatory marker expression. Freshly isolated human myoblasts were cultured in media supplemented with graded concentrations of Co2+ and/or Cr3+ . Co2+ induced a concentration-dependent decrease of both myoblast viability and myogenic differentiation while Cr3+ did not. Co2+ or Co2+ /Cr3+ also induced the upregulation of ICAM-1, whereas HLA-DR expression was unaffected. Moreover, allogenic monocytes induced the synergistic increase of Co2+ -induced ICAM-1 expression. We also found that Co2+ stabilized HIF-1α and increased TLR4, tumor necrosis factor-alpha (TNF-α), and interleukin 1ß (IL-1ß) expression in a dose and time-dependent manner in human myoblasts. This study showed that Co2+ , but not Cr3+ , was toxic toward myoblasts and induced, in the surviving cells, expression of inflammatory markers such as ICAM-1, TLR4, TNF-α, and IL-1ß. This suggests that Co2+ , most efficiently in the presence of monocytes, may be a key inducer of ALTR, which may, if severe and long-lasting, eventually result in prosthesis loosening.


Subject(s)
Chromium/adverse effects , Cobalt/adverse effects , Myoblasts/drug effects , Adolescent , Adult , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Intercellular Adhesion Molecule-1/genetics , Interleukin-1beta/genetics , Muscle Development/drug effects , Myoblasts/physiology , Toll-Like Receptor 4/genetics , Young Adult
4.
PLoS One ; 14(10): e0222683, 2019.
Article in English | MEDLINE | ID: mdl-31574082

ABSTRACT

Mesenchymal stem cells (MSCs) were obtained from human bone marrow and amplified in cultures supplemented with human platelet lysate in order to generate myofibroblasts. When MSCs were seeded in solid collagen scaffolds, they differentiated into myofibroblasts that were observed to strongly bind to the substrate, forming a 3D cell scaffold network that developed tension and shortening after KCl stimulation. Moreover, MSC-laden scaffolds recapitulated the Frank-Starling mechanism so that active tension increased in response to increases in the initial length of the contractile system. This constituted a bioengineering tissue that exhibited the contractile properties observed in both striated and smooth muscles. By using the A. F. Huxley formalism, we determined the myosin crossbridge (CB) kinetics of attachment (f1) and detachment (g1 and g2), maximum myosin ATPase activity, molar myosin concentration, unitary CB force and maximum CB efficiency. CB kinetics were dramatically slow, characterizing the non-muscle myosin type IIA (NMMIIA) present in myofibroblasts. When MSCs were seeded in solid collagen scaffolds functionalized with Arg-Gly-Asp (RGD), contractility increased and CB kinetics were modified, whereas the unitary NMMIIA-CB force and maximum CB efficiency did not change. In conclusion, we provided a non-muscle bioengineering tissue whose molecular mechanical characteristics of NMMIIA were very close to those of a non-muscle contractile tissue such as the human placenta.


Subject(s)
Muscle, Smooth/metabolism , Myosin Heavy Chains/chemistry , Oligopeptides/metabolism , Peptides/metabolism , Blood Platelets/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Collagen/chemistry , Collagen/metabolism , Humans , Kinetics , Mesenchymal Stem Cells/metabolism , Muscle Contraction/genetics , Myofibroblasts/metabolism , Myosin Heavy Chains/genetics , Myosins/chemistry , Myosins/metabolism , Oligopeptides/chemistry , Peptides/chemistry , Potassium Chloride/pharmacology
5.
Stem Cells Int ; 2018: 6134787, 2018.
Article in English | MEDLINE | ID: mdl-29853916

ABSTRACT

Mesenchymal stromal stem cells (MSC) that reside in the bone marrow (BM) can be amplified in vitro. In 2-dimension (D) cultures, MSC exhibit a morphology similar to fibroblasts, are able to inhibit T lymphocyte and natural killer cell proliferation, and can be differentiated into adipocytes, chondrocytes, or osteoblasts if exposed to specific media. Here we show that medullar MSC cultured in 2D formed an adherent stroma of cells expressing well-organized microfilaments containing α-smooth muscle actin and nonmuscle myosin heavy chain IIA. MSC could be grown in 3D in collagen membranes generating a structure which, upon exposition to 50 mM KCl or to an alternating electric current, developed a contractile strength that averaged 34 and 45 µN/mm2, respectively. Such mechanical tension was similar in intensity and in duration to that of human placenta and was annihilated by isosorbide dinitrate or 2,3-butanedione monoxime. Membranes devoid of MSC did not exhibit a significant contractility. Moreover, MSC nested in collagen membranes were able to control T lymphocyte proliferation, and differentiated into adipocytes, chondrocytes, or osteoblasts. Our observations show that BM-derived MSC cultured in collagen membranes spontaneously differentiate into contractile myofibroblasts exhibiting unexpected properties in terms of cell differentiation potential and of immunomodulatory function.

6.
Sci Rep ; 7(1): 3462, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28615691

ABSTRACT

Satellite cells, localized within muscles in vivo, are Pax7+ muscle stem cells supporting skeletal muscle growth and regeneration. Unfortunately, their amplification in vitro, required for their therapeutic use, is associated with reduced regenerative potential. In the present study, we investigated if human myogenic reserve cells (MRC) obtained in vitro, represented a reliable cell source for muscle repair. For this purpose, primary human myoblasts were freshly isolated and expanded. After 2 days of differentiation, 62 ± 2.9% of the nuclei were localized in myotubes and 38 ± 2.9% in the mononucleated non-fusing MRC. Eighty percent of freshly isolated human MRC expressed a phenotype similar to human quiescent satellite cells (CD56+/Pax7+/MyoD-/Ki67- cells). Fourteen days and 21 days after cell transplantation in immunodeficient mice, live human cells were significantly more numerous and the percentage of Pax7+/human lamin A/C+ cells was 2 fold higher in muscles of animals injected with MRC compared to those injected with human myoblasts, despite that percentage of spectrin+ and lamin A/C+ human fibers in both groups MRC were similar. Taken together, these data provide evidence that MRC generated in vitro represent a promising source of cells for improving regeneration of injured skeletal muscles.


Subject(s)
Muscle Development , Regeneration , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/metabolism , Adult , Animals , Biomarkers , Cell Differentiation , Cell Survival , Cells, Cultured , Humans , Immunocompromised Host , Mice , Mice, Transgenic , Models, Animal , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/transplantation , Stem Cell Transplantation/methods , Young Adult
7.
Transfus Med Hemother ; 40(1): 14-20, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23637645

ABSTRACT

BACKGROUND: Over the last 2 decades, cord blood (CB) has become an important source of blood stem cells. Clinical experience has shown that CB is a viable source for blood stem cells in the field of unrelated hematopoietic blood stem cell transplantation. METHODS: Studies of CB units (CBUs) stored and ordered from the US (National Marrow Donor Program (NMDP) and Swiss (Swiss Blood Stem Cells (SBSQ)) CB registries were conducted to assess whether these CBUs met the needs of transplantation patients, as evidenced by units being selected for transplantation. These data were compared to international banking and selection data (Bone Marrow Donors Worldwide (BMDW), World Marrow Donor Association (WMDA)). Further analysis was conducted on whether current CB banking practices were economically viable given the units being selected from the registries for transplant. It should be mentioned that our analysis focused on usage, deliberately omitting any information about clinical outcomes of CB transplantation. RESULTS: A disproportionate number of units with high total nucleated cell (TNC) counts are selected, compared to the distribution of units by TNC available. Therefore, the decision to use a low threshold for banking purposes cannot be supported by economic analysis and may limit the economic viability of future public CB banking. CONCLUSIONS: We suggest significantly raising the TNC level used to determine a bankable unit. A level of 125 × 10(7) TNCs, maybe even 150 × 10(7) TNCs, might be a viable banking threshold. This would improve the return on inventory investments while meeting transplantation needs based on current selection criteria.

8.
PLoS One ; 8(1): e53719, 2013.
Article in English | MEDLINE | ID: mdl-23341981

ABSTRACT

OBJECTIVE: Impaired liver regeneration is associated with a poor outcome in patients with decompensated alcoholic liver disease (ALD). We assessed whether autologous bone marrow mononuclear cell transplantation (BMMCT) improved liver function in decompensated ALD. DESIGN: 58 patients (mean age 54 yrs; mean MELD score 19, all with cirrhosis, 81% with alcoholic steatohepatitis at baseline liver biopsy) were randomized early after hospital admission to standard medical therapy (SMT) alone (n = 30), including steroids in patients with a Maddrey's score ≥32, or combined with G-CSF injections and autologous BMMCT into the hepatic artery (n = 28). Bone marrow cells were harvested, isolated and reinfused the same day. The primary endpoint was a ≥3 points decrease in the MELD score at 3 months, corresponding to a clinically relevant improvement in liver function. Liver biopsy was repeated at week 4 to assess changes in Ki67+/CK7+ hepatic progenitor cells (HPC) compartment. RESULTS: Both study groups were comparable at baseline. After 3 months, 2 and 4 patients died in the BMMCT and SMT groups, respectively. Adverse events were equally distributed between groups. Moderate alcohol relapse occurred in 31% of patients. The MELD score improved in parallel in both groups during follow-up with 18 patients (64%) from the BMMCT group and 18 patients (53%) from the SMT group reaching the primary endpoint (p = 0.43 (OR 1.6, CI 0.49-5.4) in an intention to treat analysis. Comparing liver biopsy at 4 weeks to baseline, steatosis improved (p<0.001), and proliferating HPC tended to decrease in both groups (-35 and -33%, respectively). CONCLUSION: Autologous BMMCT, compared to SMT is a safe procedure but did not result in an expanded HPC compartment or improved liver function. These data suggest either insufficient regenerative stimulation after BMMCT or resistance to liver regenerative drive in patients with decompensated alcoholic cirrhosis. TRIAL REGISTRATION: Controlled-Trials.com ISRCTN83972743.


Subject(s)
Bone Marrow Transplantation , Liver Diseases, Alcoholic/surgery , Adolescent , Adult , Aged , Biomarkers/blood , Bone Marrow Transplantation/adverse effects , Cell Proliferation , Cytokines/blood , Female , Hepatic Artery/surgery , Hepatocytes/pathology , Humans , Liver/pathology , Liver/physiopathology , Liver/surgery , Liver Diseases, Alcoholic/blood , Liver Diseases, Alcoholic/pathology , Liver Diseases, Alcoholic/physiopathology , Male , Middle Aged , Recovery of Function , Regeneration , Stem Cell Transplantation/adverse effects , Time Factors , Transplantation, Autologous/adverse effects , Young Adult
9.
J Leukoc Biol ; 93(5): 681-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23325926

ABSTRACT

Mammalian IDO is a heme-containing enzyme whose main activity in mammals is to degrade the essential amino acid tryp into l-kynurenine. Although the link between its enzymatic activity and the immune response is not straightforward, several lines of evidence suggest that this enzyme is involved in fighting infections and paradoxically, also in the establishment of the immune tolerance associated with fetus implantation and with the development of oncogenic processes. IDO is associated with the successful development of the fetus. It participates early in pregnancy to the efficient invasion of the uterine mucosa by the nascent trophoblast and remains active throughout the whole process, as illustrated by the decrease in systemic tryp from the second trimester of gestation and the return to normal values after delivery. The short-term activation of IDO in response to invading pathogens and emerging tumors participates in the elimination of these threats, whereas the sustained activation of IDO often results in a state of immune tolerance that may favor chronic infections and the uncontrolled proliferation of malignant cells. However, despite these potential deleterious effects of IDO, the enzyme is instrumental in maintaining the peripheral tolerance that is required to avoid autoimmune diseases. Below, we review the implication of IDO activation upon the physiological development of the fetus and the pathological development of tumors and discuss whether such an enzyme could be used as a therapeutic tool to decrease the rate of allograft rejections via its potent immunomodulatory properties.


Subject(s)
Immune Tolerance , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Neoplasms/immunology , Pregnancy/immunology , Transplantation, Homologous/immunology , Animals , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry
10.
Cell Transplant ; 22(7): 1213-26, 2013.
Article in English | MEDLINE | ID: mdl-23031415

ABSTRACT

Myoblast transplantation represents a promising therapeutic strategy in the treatment of several genetic muscular disorders including Duchenne muscular dystrophy. Nevertheless, such an approach is impaired by the rapid death, limited migration, and rejection of transplanted myoblasts by the host. Low molecular weight dextran sulfate (DXS), a sulfated polysaccharide, has been reported to act as a cytoprotectant for various cell types. Therefore, we investigated whether DXS could act as a "myoblastprotectant" either in vitro or in vivo after transplantation in immunodeficient mice. In vitro, DXS bound human myoblasts in a dose-dependent manner and significantly inhibited staurosporine-mediated apoptosis and necrosis. DXS pretreatment also protected human myoblasts from natural killer cell-mediated cytotoxicity. When human myoblasts engineered to express the renilla luciferase transgene were transplanted in immunodeficient mice, bioluminescence imaging analysis revealed that the proportion of surviving myoblasts 1 and 3 days after transplantation was two times higher when cells were preincubated with DXS compared to control (77.9 ± 10.1% vs. 39.4 ± 4.9%, p = 0.0009 and 38.1 ± 8.5% vs. 15.1 ± 3.4%, p = 0.01, respectively). Taken together, we provide evidence that DXS acts as a myoblast protectant in vitro and is able in vivo to prevent the early death of transplanted myoblasts.


Subject(s)
Apoptosis/drug effects , Dextran Sulfate/pharmacology , Myoblasts/transplantation , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Child, Preschool , Dextran Sulfate/chemistry , Female , Graft Survival/immunology , Humans , Infant , Killer Cells, Natural/immunology , Luminescent Measurements , Mice , Mice, Inbred NOD , Molecular Weight , Myoblasts/cytology , Necrosis , Staurosporine/toxicity , Transplantation, Heterologous
11.
Cytotherapy ; 14(2): 215-22, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22136296

ABSTRACT

BACKGROUND AIMS: Umbilical cord blood (UCB) is a source of hematopoietic stem cells that initially was used exclusively for the hematopoietic reconstitution of pediatric patients. It is now suggested for use for adults as well, a fact that increases the pressure to obtain units with high cellularity. Therefore, the optimization of UCB processing is a priority. METHODS: The present study focused on parameters influencing total nucleated cell (TNC), mononucleated cell (MNC) and CD34+ cell (CD34C) recovery after routine volume reduction of 1553 UCB units using hydroxyethyl starch-induced sedimentation with an automated device, under routine laboratory conditions. RESULTS: We show that the unit volume rather than the TNC count significantly affects TNC, MNC and CD34C processing efficiency (PEf), and this in a non-linear fashion: when units were sampled according to the collection volume, including pre-loaded anticoagulant (gross volume), PEf increased up to a unit volume of 110-150 mL and decreased thereafter. Thus units with initial gross volumes < 90 mL and > 170 mL similarly exhibited a poor PEf. CONCLUSIONS: These data identify unit gross volume as a major parameter influencing PEf and suggest that fractionation of large units should be contemplated only when the resulting volume of split units is > 90 mL.


Subject(s)
Cell Culture Techniques/methods , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Leukocytes, Mononuclear/cytology , Antigens, CD34/immunology , Blood Sedimentation , Humans , Hydroxyethyl Starch Derivatives/chemistry , T-Lymphocytes/cytology , T-Lymphocytes/immunology
12.
Pediatr Transplant ; 16(2): 131-6, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22151234

ABSTRACT

TA-TMA is a pathology that occurs after allogenic HSC transplantation with an incidence of 4-13%, and represents one of the most severe vascular damage related with this therapy. We report here the case of a nine-yr-old girl suffering from a severe refractory aplastic anemia who received an unrelated, 9/10 HLA-matched HSC. Soon after transplantation, the patient developed a graft-versus-host disease (GvHD), a TA-TMA, and renal insufficiency. These pathologies remained refractory to the various treatments undertaken and required several hospitalizations in the intensive care unit. On day 106 post-HSC transfusion, after several episodes of intensive care, the patient was infused with mismatched, third-party MSCs. Schizocyte levels rapidly decreased after MSC infusion, and two wk later, most biological parameters returned to normal. Erythrocyte and thrombocyte transfusions were discontinued, and the patient remained stable for 10 wk. Thereafter, TA-TMA symptoms, viral reactivation, pleural and cardiac effusions reappeared and lead to the death of the patient. Our observations suggest that allogenic MSC infusion may decrease the symptoms of TA-TMA, but further investigation is required to determine how and when MSC should be infused to develop a long-lasting protective effect.


Subject(s)
Anemia, Aplastic/surgery , Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cell Transplantation , Postoperative Complications/surgery , Thrombotic Microangiopathies/surgery , Child , Fatal Outcome , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/surgery , Humans , Renal Insufficiency/etiology , Renal Insufficiency/surgery , Thrombotic Microangiopathies/etiology
13.
Cell Transplant ; 20(5): 681-91, 2011.
Article in English | MEDLINE | ID: mdl-21054933

ABSTRACT

Mesenchymal stromal cells (MSCs) are potent immunomodulators that have successfully been used to circumvent various types of inflammations, including steroid-resistant graft-versus-host disease. Although initially believed to be restricted to multipotent MSCs, this immunoregulatory function is shared with differentiated cells from the mesenchymal lineage such as skin fibroblasts (SFs). Mesenchymal cell-induced immunoregulation is so potent that it may allow the reactivation of dormant malignancies, a fact that would preclude using such cells as therapeutic agents. Because NK cells are pivotal effectors controlling tumor cell containment we investigated the effect of allogenic MSCs and SFs on NK cell function in vitro. When NK cells were incubated with IL-15 and MSCs or SFs for 6 days, their proliferation and cytotoxic activity were significantly decreased compared to NK cells cultured with IL-15 alone or with human venous endothelial cells. Cytotoxic activity inhibition reached 86% when assayed on MHC-I(+) allogenic primary hematopoietic blasts, and was associated with a significant decrease in cytolytic granule exocytosis and in perforin release. Stromal cell-mediated inhibition was effective only if cell-cell proximity was long lasting: when NK cells were activated with IL-15 in the absence of MSCs and assayed for cytotoxicity in their presence no inhibition occurred. MSC inhibition was ultimately mediated by a soluble factor generated upon incubation with NK cells activated by IL-15 or IL-2. The indoleamine 2,3 dioxygenase was activated in MSCs and SFs because L-kynurenine was detected in inhibitory supernatants, but its blockade did not restore NK cell functions. The profound inhibition of cytotoxic activity directed against allogenic hematopoietic blasts exerted by MSCs and SFs on NK cells may be a concern. Should this occur in vivo it may induce the inability of NK cells to control residual or dormant malignant diseases after infusion of therapeutic MSCs.


Subject(s)
Bone Marrow Cells/cytology , Killer Cells, Natural/cytology , Mesenchymal Stem Cells/cytology , Cell Lineage , Cell Proliferation , Cells, Cultured , Endothelial Cells/cytology , Exocytosis , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-15/pharmacology , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Killer Cells, Natural/physiology , Kynurenine/metabolism , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Perforin/metabolism
14.
PLoS One ; 4(11): e7904, 2009 Nov 19.
Article in English | MEDLINE | ID: mdl-19936258

ABSTRACT

BACKGROUND: A characteristic SYT-SSX fusion gene resulting from the chromosomal translocation t(X;18)(p11;q11) is detectable in almost all synovial sarcomas, a malignant soft tissue tumor widely believed to originate from as yet unidentified pluripotent stem cells. The resulting fusion protein has no DNA binding motifs but possesses protein-protein interaction domains that are believed to mediate association with chromatin remodeling complexes. Despite recent advances in the identification of molecules that interact with SYT-SSX and with the corresponding wild type SYT and SSX proteins, the mechanisms whereby the SYT-SSX might contribute to neoplastic transformation remain unclear. Epigenetic deregulation has been suggested to be one possible mechanism. METHODOLOGY/PRINCIPAL FINDINGS: We addressed the effect of SYT/SSX expression on the transcriptome of four independent isolates of primary human bone marrow mesenchymal stem cells (hMSC). We observed transcriptional changes similar to the gene expression signature of synovial sarcoma, principally involving genes whose regulation is linked to epigenetic factors, including imprinted genes, genes with transcription start sites within a CpG island and chromatin related genes. Single population analysis revealed hMSC isolate-specific transcriptional changes involving genes that are important for biological functions of stem cells as well as genes that are considered to be molecular markers of synovial sarcoma including IGF2, EPHRINS, and BCL2. Methylation status analysis of sequences at the H19/IGF2 imprinted locus indicated that distinct epigenetic features characterize hMSC populations and condition the transcriptional effects of SYT-SSX expression. CONCLUSIONS/SIGNIFICANCE: Our observations suggest that epigenetic features may define the cellular microenvironment in which SYT-SSX displays its functional effects.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Mesenchymal Stem Cells/cytology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Adolescent , Alleles , Child , Chromatin/metabolism , CpG Islands , DNA/genetics , Gene Expression Profiling , Humans , Sarcoma, Synovial/metabolism , Transcription, Genetic , Translocation, Genetic
15.
PLoS One ; 3(7): e2634, 2008 Jul 09.
Article in English | MEDLINE | ID: mdl-18648544

ABSTRACT

BACKGROUND: The EWS-FLI-1 fusion protein is associated with 85-90% of Ewing's sarcoma family tumors (ESFT), the remaining 10-15% of cases expressing chimeric genes encoding EWS or FUS fused to one of several ets transcription factor family members, including ERG-1, FEV, ETV1 and ETV6. ESFT are dependent on insulin-like growth factor-1 (IGF-1) for growth and survival and recent evidence suggests that mesenchymal progenitor/stem cells constitute a candidate ESFT origin. METHODOLOGY/PRINCIPAL FINDINGS: To address the functional relatedness between ESFT-associated fusion proteins, we compared mouse progenitor cell (MPC) permissiveness for EWS-FLI-1, EWS-ERG and FUS-ERG expression and assessed the corresponding expression profile changes. Whereas all MPC isolates tested could stably express EWS-FLI-1, only some sustained stable EWS-ERG expression and none could express FUS-ERG for more than 3-5 days. Only 14% and 4% of the total number of genes that were respectively induced and repressed in MPCs by the three fusion proteins were shared. However, all three fusion proteins, but neither FLI-1 nor ERG-1 alone, activated the IGF1 promoter and induced IGF1 expression. CONCLUSION/SIGNIFICANCE: Whereas expression of different ESFT-associated fusion proteins may require distinct cellular microenvironments and induce transcriptome changes of limited similarity, IGF1 induction may provide one common mechanism for their implication in ESFT pathogenesis.


Subject(s)
Gene Expression Regulation , Insulin-Like Growth Factor I/metabolism , Mesenchymal Stem Cells/cytology , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/chemistry , RNA-Binding Protein FUS/metabolism , Transcription Factors/metabolism , Animals , Gene Expression Profiling , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS
16.
Cancer Res ; 68(7): 2176-85, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18381423

ABSTRACT

Ewing's sarcoma family tumors (ESFT) express the EWS-FLI-1 fusion gene generated by the chromosomal translocation t(11;22)(q24;q12). Expression of the EWS-FLI-1 fusion protein in a permissive cellular environment is believed to play a key role in ESFT pathogenesis. However, EWS-FLI-1 induces growth arrest or apoptosis in differentiated primary cells, and the identity of permissive primary human cells that can support its expression and function has until now remained elusive. Here we show that expression of EWS-FLI-1 in human mesenchymal stem cells (hMSC) is not only stably maintained without inhibiting proliferation but also induces a gene expression profile bearing striking similarity to that of ESFT, including genes that are among the highest ESFT discriminators. Expression of EWS-FLI-1 in hMSCs may recapitulate the initial steps of Ewing's sarcoma development, allowing identification of genes that play an important role early in its pathogenesis. Among relevant candidate transcripts induced by EWS-FLI-1 in hMSCs, we found the polycomb group gene EZH2, which we show to play a critical role in Ewing's sarcoma growth. These observations are consistent with our recent findings using mouse mesenchymal progenitor cells and provide compelling evidence that hMSCs are candidate cells of origin of ESFT.


Subject(s)
Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Oncogene Proteins, Fusion/biosynthesis , Proto-Oncogene Protein c-fli-1/biosynthesis , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Differentiation/physiology , Enhancer of Zeste Homolog 2 Protein , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Histone-Lysine N-Methyltransferase , Humans , Immunocompromised Host , Mice , Oncogene Proteins, Fusion/genetics , Phenotype , Polycomb Repressive Complex 2 , Proteins/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA-Binding Protein EWS , Sarcoma, Ewing/genetics , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/pathology
17.
J Cell Physiol ; 214(3): 588-94, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17786951

ABSTRACT

The regulatory effect of human multipotent mesenchymal stromal cells (MSC) on allogenic T lymphocytes is extremely powerful and of important clinical relevance, but the mechanisms underlying this process are not fully elucidated. We report here that T lymphocytes activated with a sub-mitogenic stimulus such as phytohemaglutinin alone (PHA), or with mitogenic stimuli such as PHA + interleukin-2 (P-IL2), or immobilized anti-CD3 + anti-CD28 mAb (a3-28), tightly bound allogenic MSC and transmigrated within 4 h under them, where they remained for approximately 60 h. Allogenic MSC induced T cell proliferation in cultures containing sub-mitogenic PHA concentrations, and inhibited the mitogenic effect of P-IL2 or a3-28. Anti-gamma-IFN mAb or L-tryptophan complementation partially restored proliferation in P-IL2 and a3-28 cultures, whereby gamma-IFN-synthesizing CD3+ cells were detectable. MSC-lymphocyte contact hindrance using transwells abrogated proliferation in PHA cultures, restored it integrally in P-IL2 cultures, and partially in a3-28 cultures. These data suggest that MSC-induced T lymphocyte regulation results from the combination of various processes. Allogenic cell-cell contact, as demonstrated by the PHA co-cultures is per se stimulatory, whereas gamma-IFN synthesized by activated T lymphocytes, which activates indolamine 2,3-dioxygenase in MSC, and L-tryptophan depletion, which is induced by this enzyme, are inhibitory. Transmigration is nevertheless pivotal for the establishment of the inhibition by these mediators because it targets lymphocytes under the stroma in small extracellular spaces surrounded by MSC, where L-tryptophan is efficiently destroyed, leading to T lymphocyte proliferation arrest. In conclusion lymphocyte transmigration under allogenic MSC potentiates the inhibitory effect of soluble mediators generated by these cells.


Subject(s)
Cell Movement , Lymphocyte Activation/immunology , Mesoderm/cytology , Multipotent Stem Cells/cytology , Stromal Cells/cytology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Blood Platelets , CD3 Complex/metabolism , Cell Adhesion , Cell Differentiation , Cell Proliferation , Humans , Interferon-gamma/metabolism , Phenotype
18.
Exp Hematol ; 34(11): 1471-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17046566

ABSTRACT

OBJECTIVE: We investigated whether the expression of CD11b on precursors derived in vitro from CD34+ hematopoietic stem cells was related to their ability to generate CD11b- and CD11b+ Langerhans dendritic cells (LC). METHODS: Human CD34+ cells purified from cord blood were cultured with FLT3 ligand, thrombopoietin, and stem cell factor (FTS) for 2 weeks, analyzed, and sorted by FACS. Sorted fractions were cultured as above, or differentiated into LC with GM-CSF, IL-4, and TGF-beta1 (G4-TGF) for 6 days. The capacity of LC to internalize langerin and dextran was assessed. RESULTS: Ex vivo, human CD34+ cells were CD11b- and mostly CLA+. After 2 weeks of culture with FTS, CD34- CLA- CD11b- and CD34- CLA- CD11b+ cells emerged. CD11b- cells were the most ancestral because they were the only ones to proliferate with FTS, and constantly generated CD11b+ cells. Both CD11b- and CD11b+ sorted cells generated E-cadherin+ langerin+ LC after incubation with G4-TGF. The former fraction contained 46% +/- 15% of E-cadherin+ and 10% +/- 5% of langerin+ cells, whereas in the latter fraction these values reached respectively 66% +/- 23% and 30% +/- 16% (mean +/- SD, n = 7, p < 0.056). Looking at functional properties, CD11b- and CD11b+ LC were similar in terms of langerin and dextran endocytosis. By contrast, only CD11b+ LC internalized fluorescent LPS. CONCLUSION: Human CD34+ CD11b- cells differentiate in FTS culture into a CD34- CD11b- precursor that in turn generates CD34- CD11b+ cells. These cells are enriched in LC precursors compared to CD34- CD11b- cells. Both CD11b- and CD11b+ LC are generated in vitro, and each fraction may assume different functions in inflammatory situations.


Subject(s)
Antigens, CD34/immunology , Antigens, CD/biosynthesis , CD11b Antigen/immunology , Fetal Blood/cytology , Hematopoietic Stem Cells/immunology , Langerhans Cells/immunology , Lectins, C-Type/biosynthesis , Mannose-Binding Lectins/biosynthesis , Antigens, CD34/biosynthesis , CD11b Antigen/biosynthesis , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-4/pharmacology , Langerhans Cells/cytology , Langerhans Cells/drug effects , Lipopolysaccharides/pharmacology , Membrane Proteins/pharmacology , Stem Cell Factor/pharmacology , Thrombopoietin/pharmacology , Transforming Growth Factor beta1/pharmacology
19.
Swiss Med Wkly ; 136(21-22): 333-7, 2006 May 27.
Article in English | MEDLINE | ID: mdl-16826633

ABSTRACT

Postnatal stem cells are present in many adult tissues, and are thought to ensure homoeostasis by replacing functionally declining cells by newly differentiated ones. Postnatal stem cells used as such or after in vitro manipulation hold out strong hopes for reconstructive therapies. For instance, the grafting of native haematopoietic stem cells (HSC) restores haematopoiesis in genetically deficient individuals or in lethally conditioned leukaemic patients, and systemic injection of in vitro amplified mesenchymal stem cells (MSC) induces recovery of bone growth in patients with osteogenesis imperfecta. Moreover, cells differentiated in vitro from postnatal stem cells exhibiting a specific function can also be used for cell therapy. Myeloid dendritic cells (DC) derived from cultures of HSC may induce tumour-specific cytotoxic T lymphocytes to eradicate the tumour via antigen recognition. In addition, long-lived MSC has been engineered to secrete specific proteins coded by a transgene and used as a source of therapeutic molecules in vivo. All these approaches require large quantities of cells that cannot be obtained (with the exception of HSC) directly from the donor. In vitro procedures allowing the production of therapeutic cells from postnatal stem cells are needed and are at present under development. Below we discuss the rationale and methods currently available for generation of therapeutic cells derived from haematopoietic and mesenchymal stem cells.


Subject(s)
Cell Transplantation/trends , Hematopoietic Stem Cells , Mesenchymal Stem Cells , Animals , Antigen-Presenting Cells/cytology , Cell Differentiation , Cells, Cultured , Child , Dendritic Cells/immunology , Forecasting , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Humans , Leukemia/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mice , Osteogenesis Imperfecta/therapy , Papio
20.
J Leukoc Biol ; 78(4): 836-44, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16199730

ABSTRACT

Postnatal stem cells regulate the homeostasis of the majority of our tissues. They continuously generate new progenitors and mature, functional cells to replace old cells, which cannot assume the tissue function anymore and are eliminated. Blood, skin, gut mucosa, muscle, cartilage, nerves, cornea, retina, liver, and many other structures are regulated by stem cells. As a result of their ability to produce large numbers of functionally mature cells, postnatal stem cells represent a promising tool for regenerative therapy. Indeed, unmanipulated stem cells or their progeny amplified in vitro are already used in some clinical applications to restore the function of injured or genetically deficient tissues. However, despite our cumulating understanding concerning postnatal stem cells, many aspects of their functionality remain unclear. For instance, in most tissues, we cannot reliably define the phenotype of the postnatal stem cells sustaining its survival. We do not know to which extent the environment surrounding the stem cell-the niche-which is a key actor insuring stem cell self-maintenance, is also implicated in the maintenance of stem cell lineage specificity. Moreover, we have to clarify whether postnatal stem cells are capable of undertaking "transdifferentiation", that is, the conversion of one cell type into another under physiological conditions. Answering these questions should help us to draw a more accurate picture of postnatal stem cell biology and should lead to the design of safe, effective therapies.


Subject(s)
Cell Lineage/physiology , Stem Cells/cytology , Stem Cells/physiology , Cell Differentiation/physiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Models, Immunological
SELECTION OF CITATIONS
SEARCH DETAIL
...