Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mol Ther ; 31(12): 3531-3544, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37805713

ABSTRACT

In vivo apoptosis of human mesenchymal stromal cells (MSCs) plays a critical role in delivering immunomodulation. Yet, caspase activity not only mediates the dying process but also death-independent functions that may shape the immunogenicity of apoptotic cells. Therefore, a better characterization of the immunological profile of apoptotic MSCs (ApoMSCs) could shed light on their mechanistic action and therapeutic applications. We analyzed the transcriptomes of MSCs undergoing apoptosis and identified several immunomodulatory factors and chemokines dependent on caspase activation following Fas stimulation. The ApoMSC secretome inhibited human T cell proliferation and activation, and chemoattracted monocytes in vitro. Both immunomodulatory activities were dependent on the cyclooxygenase2 (COX2)/prostaglandin E2 (PGE2) axis. To assess the clinical relevance of ApoMSC signature, we used the peripheral blood mononuclear cells (PBMCs) from a cohort of fistulizing Crohn's disease (CD) patients who had undergone MSC treatment (ADMIRE-CD). Compared with healthy donors, MSCs exposed to patients' PBMCs underwent apoptosis and released PGE2 in a caspase-dependent manner. Both PGE2 and apoptosis were significantly associated with clinical responses to MSCs. Our findings identify a new mechanism whereby caspase activation delivers ApoMSC immunosuppression. Remarkably, such molecular signatures could implicate translational tools for predicting patients' clinical responses to MSC therapy in CD.


Subject(s)
Crohn Disease , Mesenchymal Stem Cells , Humans , Crohn Disease/genetics , Crohn Disease/therapy , Dinoprostone/metabolism , Leukocytes, Mononuclear/metabolism , Secretome , Mesenchymal Stem Cells/metabolism , Immunomodulation , Apoptosis , Caspases
2.
Bone Marrow Transplant ; 57(6): 942-948, 2022 06.
Article in English | MEDLINE | ID: mdl-35396529

ABSTRACT

The improvement of hematopoietic stem and progenitor cell (HSPC) engraftment remains a high-priority goal when limited cell doses are available, such as in cord blood (CB) transplantation and HSC gene therapy. We observed that monocytes are highly effective at improving the engraftment of both CB-CD34+ and lentivirus-transfected CD34+ cells in a xenogeneic model of HSC transplantation. Moreover, monocytes, in particular the CD14+CD16- classical subset, in co-culture systems increase survival and stemness of CB-CD34+ cells. Both soluble factors and direct-cell contact interactions, such as JAG/NOTCH and COX-2/PGE2 pathways, are critically involved in the HSC-monocyte crosstalk. Our results indicate that the infusion of monocytes improves engraftment when cell dose is a limiting factor.


Subject(s)
Fetal Blood , Hematopoietic Stem Cell Transplantation , Antigens, CD34/metabolism , Coculture Techniques , Fetal Blood/metabolism , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells , Humans
3.
Front Immunol ; 11: 1338, 2020.
Article in English | MEDLINE | ID: mdl-32670295

ABSTRACT

The immunosuppressive activity of mesenchymal stromal cells (MSCs) in graft versus host disease (GvHD) is well-documented, but their therapeutic benefit is rather unpredictable. Prospective randomized clinical trials remain the only means to address MSC clinical efficacy. However, the imperfect understanding of MSC biological mechanisms has undermined patients' stratification and the successful design of clinical studies. Furthermore, although MSC efficacy seems to be dependent on patient-associated factors, the role of patients' signature to predict and/or monitor clinical outcomes remains poorly elucidated. The analysis of GvHD patient serum has identified a set of molecules that are associated with high mortality. However, despite their importance in defining GvHD severity, their role in predicting or monitoring response to MSCs has not been confirmed. A new perspective on the use of MSCs for GvHD has been prompted by the recent findings that MSCs are actively induced to undergo apoptosis by recipient cytotoxic cells and that this process is essential to initiate MSC-induced immunosuppression. This discovery has not only reconciled the conundrum between MSC efficacy and their lack of engraftment, but also highlighted the determinant role of the patient in promoting and delivering MSC immunosuppression. In this review we will revisit the extensive use of MSCs for the treatment of GvHD and will elaborate on the need that future clinical trials must depend on mechanistic approaches that facilitate the development of robust and consistent assays to stratify patients and monitor clinical outcomes.


Subject(s)
Graft vs Host Disease , Immunotherapy/methods , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , Animals , Biomarkers/analysis , Humans
5.
Cell Death Dis ; 10(4): 277, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894508

ABSTRACT

Variability among donors, non-standardized methods for isolation, and characterization contribute to mesenchymal stem/stromal cell (MSC) heterogeneity. Induced pluripotent stem cell (iPSCs)-derived MSCs would circumvent many of current issues and enable large-scale production of standardized cellular therapy. To explore differences between native MSCs (nMSCs) and iPSC-derived MSCs (iMSCs), we developed isogeneic lines from Wharton's jelly (WJ) from the umbilical cords of two donors (#12 and #13) under xeno-free conditions. Next, we reprogrammed them into iPSCs (iPSC12 and iPSC13) and subsequently differentiated them back into iMSCs (iMSC12 and iMSC13) using two different protocols, which we named ARG and TEX. We assessed their differentiation capability, transcriptome, immunomodulatory potential, and interferon-γ (IFNG)-induced changes in metabolome. Our data demonstrated that although both differentiation protocols yield iMSCs similar to their parental nMSCs, there are substantial differences. The ARG protocol resulted in iMSCs with a strong immunomodulatory potential and lower plasticity and proliferation rate, whereas the TEX protocol raised iMSCs with a higher proliferation rate, better differentiation potential, though weak immunomodulatory response. Our data suggest that, following a careful selection and screening of donors, nMSCs from umbilical's cord WJ can be easily reprogrammed into iPSCs, providing an unlimited source of material for differentiation into iMSCs. However, the differentiation protocol should be chosen depending on their clinical use.


Subject(s)
Cell Differentiation/physiology , Cellular Reprogramming/physiology , Induced Pluripotent Stem Cells/metabolism , Interferon-gamma/pharmacology , Mesenchymal Stem Cells/metabolism , Metabolome/drug effects , Umbilical Cord/cytology , Cell Plasticity , Cell Proliferation , Cells, Cultured , Cellular Reprogramming Techniques/methods , Female , Humans , Transcriptome/drug effects
6.
Br J Haematol ; 185(1): 89-92, 2019 04.
Article in English | MEDLINE | ID: mdl-30637732

ABSTRACT

Mesenchymal stromal cells (MSCs) have been successfully used for the treatment of steroid-resistant graft-versus-host-disease (GvHD). However, the lack of early predictors of clinical responses impacts on the time at which to add further treatment and consequently the design of informative clinical trials. Here, we present the UK experience of one of the largest cohorts of GvHD patients undergoing MSC infusions so far reported. We show that clinical responses assessed as early as 1 week after MSC infusion predict patients' overall survival. In our cohort, cell dose, patients' age and type of organ involvement are crucial factors associated with clinical responses.


Subject(s)
Graft vs Host Disease/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Adult , Aged , Biomedical Research , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/mortality , Humans , Male , Mesenchymal Stem Cells/cytology , Middle Aged , Prognosis , Treatment Outcome , Young Adult
7.
Biol Blood Marrow Transplant ; 24(11): 2365-2370, 2018 11.
Article in English | MEDLINE | ID: mdl-30031938

ABSTRACT

The immunosuppressive properties of mesenchymal stromal cells (MSC) have been successfully tested to control clinical severe graft-versus host disease and improve survival. However, clinical studies have not yet provided conclusive evidence of their efficacy largely because of lack of patients' stratification criteria. The heterogeneity of MSC preparations is also a major contributing factor, as manufacturing of therapeutic MSC is performed according to different protocols among different centers. Understanding the variability of the manufacturing protocol would allow a better comparison of the results obtained in the clinical setting among different centers. In order to acquire information on MSC manufacturing we sent a questionnaire to the European Society for Blood and Marrow Transplantation centers registered as producing MSC. Data from 17 centers were obtained and analyzed by means of a 2-phase questionnaire specifically focused on product manufacturing. Gathered information included MSC tissue sources, MSC donor matching, medium additives for ex vivo expansion, and data on MSC product specification for clinical release. The majority of centers manufactured MSC from bone marrow (88%), whilst only 2 centers produced MSC from umbilical cord blood or cord tissue. One of the major changes in the manufacturing process has been the replacement of fetal bovine serum with human platelet lysate as medium supplement. 59% of centers used only third-party MSC, whilst only 1 center manufactured exclusively autologous MSC. The large majority of these facilities (71%) administered MSC exclusively from frozen batches. Aside from variations in the culture method, we found large heterogeneity also regarding product specification, particularly in the markers used for phenotypical characterization and their threshold of expression, use of potency assays to test MSC functionality, and karyotyping. The initial data collected from this survey highlight the variability in MSC manufacturing as clinical products and the need for harmonization. Until more informative potency assays become available, a more homogeneous approach to cell production may at least reduce variability in clinical trials and improve interpretation of results.


Subject(s)
Graft vs Host Disease/therapy , Mesenchymal Stem Cells/metabolism , Europe , Graft vs Host Disease/pathology , Humans , Mesenchymal Stem Cells/cytology , Surveys and Questionnaires
8.
Sci Rep ; 7(1): 17595, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29242640

ABSTRACT

We investigated whether maternal metabolic environment affects mesenchymal stromal/stem cells (MSCs) from umbilical cord's Wharton's Jelly (WJ) on a molecular level, and potentially render them unsuitable for clinical use in multiple recipients. In this pilot study on umbilical cords post partum from healthy non-obese (BMI = 19-25; n = 7) and obese (BMI ≥ 30; n = 7) donors undergoing elective Cesarean section, we found that WJ MSC from obese donors showed slower population doubling and a stronger immunosuppressive activity. Genome-wide DNA methylation of triple positive (CD73+CD90+CD105+) WJ MSCs found 67 genes with at least one CpG site where the methylation difference was ≥0.2 in four or more obese donors. Only one gene, PNPLA7, demonstrated significant difference on methylome, transcriptome and protein level. Although the number of analysed donors is limited, our data suggest that the altered metabolic environment related to excessive body weight might bear consequences on the WJ MSCs.


Subject(s)
Mesenchymal Stem Cells/pathology , Mothers , Obesity/pathology , Wharton Jelly/pathology , Adult , CD56 Antigen/metabolism , Case-Control Studies , Cell Differentiation , DNA Methylation , Down-Regulation , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunomodulation , Lipase/genetics , Lysophospholipase , Mesenchymal Stem Cells/metabolism , Obesity/genetics , Obesity/immunology , Obesity/metabolism , Pilot Projects , Pregnancy
9.
Sci Transl Med ; 9(416)2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29141887

ABSTRACT

The immunosuppressive activity of mesenchymal stromal cells (MSCs) is well documented. However, the therapeutic benefit is completely unpredictable, thus raising concerns about MSC efficacy. One of the affecting factors is the unresolved conundrum that, despite being immunosuppressive, MSCs are undetectable after administration. Therefore, understanding the fate of infused MSCs could help predict clinical responses. Using a murine model of graft-versus-host disease (GvHD), we demonstrate that MSCs are actively induced to undergo perforin-dependent apoptosis by recipient cytotoxic cells and that this process is essential to initiate MSC-induced immunosuppression. When examining patients with GvHD who received MSCs, we found a striking parallel, whereby only those with high cytotoxic activity against MSCs responded to MSC infusion, whereas those with low activity did not. The need for recipient cytotoxic cell activity could be replaced by the infusion of apoptotic MSCs generated ex vivo. After infusion, recipient phagocytes engulf apoptotic MSCs and produce indoleamine 2,3-dioxygenase, which is ultimately necessary for effecting immunosuppression. Therefore, we propose the innovative concept that patients should be stratified for MSC treatment according to their ability to kill MSCs or that all patients could be treated with ex vivo apoptotic MSCs.


Subject(s)
Apoptosis/physiology , Mesenchymal Stem Cells/cytology , Animals , Graft vs Host Disease/metabolism , Graft vs Host Disease/physiopathology , Humans , Immunomodulation/genetics , Immunomodulation/physiology , Immunosuppression Therapy/methods , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology
10.
Haematologica ; 102(5): 818-825, 2017 05.
Article in English | MEDLINE | ID: mdl-28183849

ABSTRACT

Bone marrow microenvironment is fundamental for hematopoietic homeostasis. Numerous efforts have been made to reproduce or manipulate its activity to facilitate engraftment after hematopoietic stem cell transplantation but clinical results remain unconvincing. This probably reflects the complexity of the hematopoietic niche. Recent data have demonstrated the fundamental role of stromal and myeloid cells in regulating hematopoietic stem cell self-renewal and mobilization in the bone marrow. In this study we unveil a novel interaction by which bone marrow mesenchymal stromal cells induce the rapid differentiation of CD11b+ myeloid cells from bone marrow progenitors. Such an activity requires the expression of nitric oxide synthase-2. Importantly, the administration of these mesenchymal stromal cell-educated CD11b+ cells accelerates hematopoietic reconstitution in bone marrow transplant recipients. We conclude that the liaison between mesenchymal stromal cells and myeloid cells is fundamental in hematopoietic homeostasis and suggests that it can be harnessed in clinical transplantation.


Subject(s)
Bone Marrow Cells/metabolism , Cell Differentiation , Hematopoiesis , Mesenchymal Stem Cells/metabolism , Myeloid Cells/metabolism , Nitric Oxide Synthase Type II/metabolism , Animals , Bone Marrow Cells/cytology , CD11b Antigen/metabolism , Cell Self Renewal , Hematopoietic Stem Cell Transplantation , Homeostasis , Humans , Mesenchymal Stem Cells/cytology , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/cytology , Nitric Oxide Synthase Type II/genetics
11.
Exp Hematol ; 44(11): 1034-1038, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27449989

ABSTRACT

Because different findings suggest that an immune dysregulation plays a role in the pathogenesis of myelodysplastic syndrome (MDS), we analyzed a large cohort of patients from a homogeneous Sardinian population using ImmunoChip, a genotyping array exploring 147,954 single-nucleotide polymorphisms (SNPs) localized in genomic regions displaying some degree of association with immune-mediated diseases or pathways. The population studied included 133 cases and 3,894 controls, and a total of 153,978 autosomal markers and 971 non-autosomal markers were genotyped. After association analysis, only one variant passed the genome-wide significance threshold: rs71325459 (p = 1.16 × 10-12), which is situated on chromosome 20. The variant is in high linkage disequilibrium with rs35640778, an untested missense variant situated in the RTEL1 gene, an interesting candidate that encodes for an ATP-dependent DNA helicase implicated in telomere-length regulation, DNA repair, and maintenance of genomic stability. The second most associated signal is composed of five variants that fall slightly below the genome-wide significance threshold but point out another interesting gene candidate. These SNPs, with p values between 2.53 × 10-6 and 3.34 × 10-6, are situated in the methylene tetrahydrofolate reductase (MTHFR) gene. The most associated of these variants, rs1537514, presents an increased frequency of the derived C allele in cases, with 11.4% versus 4.4% in controls. MTHFR is the rate-limiting enzyme in the methyl cycle and genetic variations in this gene have been strongly associated with the risk of neoplastic diseases. The current understanding of the MDS biology, which is based on the hypothesis of the sequential development of multiple subclonal molecular lesions, fits very well with the demonstration of a possible role for RTEL1 and MTHFR gene polymorphisms, both of which are related to a variable risk of genomic instability.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Immunomodulation/genetics , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/immunology , Alleles , Computational Biology/methods , Genome-Wide Association Study/methods , Genotype , Humans , Meta-Analysis as Topic , Polymorphism, Single Nucleotide
12.
Mol Ther ; 24(7): 1227-36, 2016 08.
Article in English | MEDLINE | ID: mdl-27133965

ABSTRACT

Mesenchymal stromal cells (MSCs) hold great promise for regenerative medicine. Stable ex vivo gene transfer to MSCs could improve the outcome and scope of MSC therapy, but current vectors require multiple rounds of transduction, involve genotoxic viral promoters and/or the addition of cytotoxic cationic polymers in order to achieve efficient transduction. We describe a self-inactivating foamy virus vector (FVV), incorporating the simian macaque foamy virus envelope and using physiological promoters, which efficiently transduces murine MSCs (mMSCs) in a single-round. High and sustained expression of the transgene, whether GFP or the lysosomal enzyme, arylsulphatase A (ARSA), was achieved. Defining MSC characteristics (surface marker expression and differentiation potential), as well as long-term engraftment and distribution in the murine brain following intracerebroventricular delivery, are unaffected by FVV transduction. Similarly, greater than 95% of human MSCs (hMSCs) were stably transduced using the same vector, facilitating human application. This work describes the best stable gene transfer vector available for mMSCs and hMSCs.


Subject(s)
Gene Transfer Techniques , Genetic Vectors/genetics , Mesenchymal Stem Cells/metabolism , Spumavirus/genetics , Transduction, Genetic , Animals , Cell Line , Gene Expression , Gene Order , Humans , Mesenchymal Stem Cell Transplantation , Mice , Promoter Regions, Genetic , Transgenes
13.
Cytotherapy ; 17(9): 1188-99, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26276002

ABSTRACT

BACKGROUND AIMS: Multipotent mesenchymal stromal cells (MSCs) are distinguished by their ability to differentiate into a number of stromal derivatives of interest for regenerative medicine, but they also have immunoregulatory properties that are being tested in a number of clinical settings. METHODS: We show that brief incubations with rapamycin, everolimus, FK506 or cyclosporine A increase the immunosuppressive potency of MSCs and other cell types. RESULTS: The treated MSCs are up to 5-fold more potent at inhibiting the induced proliferation of T lymphocytes in vitro. We show that this effect probably is due to adsorption of the drug by the MSCs during pre-treatment, with subsequent diffusion into co-cultures at concentrations sufficient to inhibit T-cell proliferation. MSCs contain measurable amounts of rapamycin after a 15-min exposure, and the potentiating effect is blocked by a neutralizing antibody to the drug. With the use of a pre-clinical model of acute graft-versus-host disease, we demonstrate that a low dose of rapamycin-treated but not untreated umbilical cord-derived MSCs significantly inhibit the onset of disease. CONCLUSIONS: The use of treated MSCs may achieve clinical end points not reached with untreated MSCs and allow for infusion of fewer cells to reduce costs and minimize potential side effects.


Subject(s)
Graft vs Host Disease/prevention & control , Immune Tolerance/drug effects , Immunosuppressive Agents/pharmacology , Mesenchymal Stem Cell Transplantation/methods , Sirolimus/pharmacology , Animals , Antibodies, Neutralizing/immunology , Cell Proliferation/drug effects , Coculture Techniques , Cyclosporine/pharmacology , Disease Models, Animal , Everolimus/pharmacology , Female , Graft vs Host Disease/immunology , Humans , Immunosuppression Therapy/methods , Lymphocyte Activation/immunology , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred BALB C , Sirolimus/immunology , T-Lymphocytes/immunology , Tacrolimus/pharmacology , Umbilical Cord/cytology
14.
Leuk Res ; 39(9): 957-63, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26209197

ABSTRACT

Patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) with multilineage dysplasia show several immunological abnormalities. In this clinical setting, by combining flow cytometry and CDR3 spectratyping we monitored the kinetic of the T-cell repertoire during Azacitidine treatment, in order to explore its potential ability to reverse the immune derangement typical of these disorders. We firstly demonstrated by flow cytometry an increase in both CD4+ and CD8+ T-cell frequencies after starting treatment. Moreover, when monitored by spectratyping our patients showed significant changes in their T-cell receptor (TCR) CDR3 profiles, which were much more evident in helper T-cells. In fact, the frequency of BV (beta variable) subfamilies showing a skewed CDR3 profile significantly decreased from baseline to the following evaluations in CD4+ T-cells (81% vs. 70%). This pattern was even more pronounced in patients responding to Azacitidine (90% vs. 61%). Our data show that the overall derangement of the T-cell repertoire detectable in patients with MDS and AML with multilineage dysplasia gradually improves during Azacitidine treatment. These findings therefore suggest that Azacitidine could be potentially able, not only to restore the hematopoietic function, but also to reverse the immune derangement typical of these hematologic disorders.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell Lineage/drug effects , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Aged , Aged, 80 and over , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Lineage/genetics , Cell Lineage/immunology , Female , Flow Cytometry , Gene Expression , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/pathology , Prospective Studies , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Treatment Outcome
15.
Eur J Haematol ; 94(4): 298-309, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25040028

ABSTRACT

Although a number of studies suggest that different immune pathways may play a role in the pathogenesis of non-Hodgkin's lymphomas (NHL), the shape of the T-cell compartment has been only superficially explored in these patients. In our study, we analyzed the peripheral T-cell receptor (TCR) repertoire and the distribution of different T-cell subsets - including regulatory T cells (Treg) - in 30 patients with NHL, by combining flow cytometry and spectratyping. We first demonstrated by flow cytometry an increased frequency of expanded T-cell subpopulations expressing the same TCR beta variable (BV) subfamilies in CD8+ cells from NHL patients when compared with healthy controls, beside a higher frequency of Treg. Moreover, NHL patients were characterized by a higher percentage of BVs showing a skewed CDR3 profile both in CD4+ and CD8+ cells when analyzed by spectratyping. Our data suggest that the T-cell branch of the immune system of patients with B-cell NHL is deeply deranged, as witnessed by the increased degree of activation and skewing of their TCR repertoire along with the higher frequency of Treg.


Subject(s)
Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow/pathology , Clonal Evolution , Complementarity Determining Regions/genetics , Complementarity Determining Regions/metabolism , Female , Flow Cytometry , Humans , Immunophenotyping , Immunotherapy , Lymphocyte Count , Lymphoma, B-Cell/diagnosis , Lymphoma, B-Cell/therapy , Male , Middle Aged , Neoplasm Staging , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocyte Subsets/drug effects
16.
Regen Med ; 9(6): 723-32, 2014.
Article in English | MEDLINE | ID: mdl-25431909

ABSTRACT

AIM: Umbilical cord contains, within Wharton's jelly (WJ), multipotent mesenchymal stromal/stem cells (MSCs) of fetal origin that can be isolated and expanded in vitro with a minimal manipulation and very high efficiency. Our aim was to develop a highly reproducible protocol that has the unique potential to be scaled up and adapted to cGMP requirements for the use in cellular therapy. RESULTS: We found that derivation of WJ MSCs under defined conditions in low oxygen resulted in several folds higher populations of MSCA-1(+) cells (6.0-19.2%) when compared with WJ MSCs derived in the presence of serum (0.1-2.8%) or clinical-grade bone marrow (BM) MSCs cultured under atmospheric O2 (20%). We demonstrate that WJ MSCs derived following our protocol display antiproliferative activity similar to clinical-grade BM MSCs. We also show that these WJ MSCs can be differentiated into adipo-, chondro- and osteo-genic lineages. CONCLUSION: Easy accessibility, abundance and genetic 'naivety' make WJ MSCs logistically a more attractive source for clinical applications than BM MSCs.


Subject(s)
Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Oxygen/metabolism , Umbilical Cord/cytology , Wharton Jelly/cytology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Flow Cytometry , Humans , Serum/chemistry
17.
Acta Haematol ; 128(3): 178-82, 2012.
Article in English | MEDLINE | ID: mdl-22890368

ABSTRACT

Regulatory T cells (T(reg)) have often been ascribed a role in the pathophysiology of several neoplastic diseases considering their potential ability to suppress anti-tumor immunity. This is particularly the case in myelodysplastic syndromes (MDS), which are clonal hematologic disorders characterized by marked immune dysregulation. We analyzed T(reg) frequencies in a cohort of 36 patients with early-stage MDS using a flow-cytometric approach based on the concomitant expression of CD25 and CD127. MDS patients showed a higher frequency of CD4+CD25(high)+CD127(low) T(reg) than healthy controls (1.51 vs. 1.14%), with no specific effect of patient- and disease-related factors. Our data point to impaired anti-tumor immunity in patients with MDS, even in the early stage, which has already been noted in other clonal disorders.


Subject(s)
Myelodysplastic Syndromes/immunology , T-Lymphocytes, Regulatory/immunology , Aged , Aged, 80 and over , Female , Flow Cytometry , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-7 Receptor alpha Subunit/immunology , Male , Middle Aged
18.
Neoplasia ; 14(6): 487-94, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22787430

ABSTRACT

It is widely accepted that a deranged immune system plays a key role in the onset and evolution of classic Kaposi sarcoma (CKS). Nevertheless, the usage of the T-cell receptor (TCR) ß-variable (BV) chain repertoire expressed by peripheral blood lymphocytes in patients with CKS is still unknown. With the aim of providing some further insights into the complex role of the immune system in CKS pathogenesis, we performed an extensive analysis of the TCR BV repertoire in both CD4(+) and CD8(+) T cells in 30 human herpesvirus 8-positive Sardinian patients with CKS and an equal number of age-matched healthy controls. We used a panel of monoclonal antibodies covering approximately 70% of human BV subfamilies and third complementarity determining region (CDR3) spectratyping. Patients with CKS showed an increased frequency of BV expansions in both CD4(+) and CD8(+) lymphocytes, with no prevalent clones. On spectratyping analysis, most of the 720 BV CDR3 profiles obtained from both CD4(+) and CD8(+) T cells in patients with CKS were skewed. In particular, the surprising increase of BV skewing observed in CD4(+) lymphocytes mimics the pattern of progressive TCR BV narrowing described in responses to persistent viral antigen stimulations. Our findings support the hypothesis that CKS evolution is associated with inadequate activation rather than impairment of the immune system.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Sarcoma, Kaposi/immunology , Aged , Aged, 80 and over , Alleles , Antibodies, Viral/blood , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Case-Control Studies , Complementarity Determining Regions/genetics , Female , Herpesvirus 8, Human/immunology , Homozygote , Humans , Lymphocyte Count , Male , Middle Aged , Receptors, Antigen, T-Cell/metabolism , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/metabolism
19.
Immunobiology ; 217(9): 920-5, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22341303

ABSTRACT

Several data suggest that stochastic rearrangements of the TCR could play a pathogenic role in both disease predisposition and protection in type 1 diabetes (T1D). As twin sets offer an enormous potential in evaluating the role of genetic and environmental factors in susceptibility to disease, the main goal of this study was to assess whether the degree of sharing of the expressed TCR repertoire of twin pairs discordant for T1D differs from that of disease concordant pairs. We performed our analysis in 5 pairs of monozygotic twins, 3 of which were concordant and 2 discordant for T1D, by combining flow cytometry and CDR3 spectratyping on both CD4+ and CD8+ T-cells. Our data show that TCR repertoires show increased level of concordance within each twin pair, especially in CD8+ cells, in terms of mean BV expression levels on flow cytometry as well as of CDR3 patterns and frequencies of skewed or oligoclonal BV subfamilies on spectratyping. It is worth noting that the degree of similarity among twins seems to be independent of concordance or discordance for T1D. Our findings seem to suggest that in monozygotic twins with T1D the TCR repertoire is influenced by genetic factors more than by the presence of the autoimmune disorder itself.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Receptors, Antigen, T-Cell/metabolism , Twins, Monozygotic , Adolescent , Adult , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/immunology , Female , Humans , Immunophenotyping , Male , Middle Aged , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...