Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Stem Cells Dev ; 28(23): 1572-1580, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31637965

ABSTRACT

Fetal bovine serum (FBS) is widely used to culture mesenchymal stem cells (MSCs) in the laboratory; however, FBS has been linked to adverse immune-mediated reactions prompting the search for alternative cell culture medium. Platelet lysate (PL) as an FBS substitute has been shown to promote MSCs growth without compromising their functionality. Fibrinogen contained in PL has been shown to negatively impact the immune modulating properties of MSCs; therefore, we sought to deplete fibrinogen from PL and compare proliferation, viability, and immunomodulatory capacities of MSCs in FBS or PL without fibrinogen. We depleted fibrinogen from equine platelet lysate (ePL) and measured platelet-derived growth factor-beta (PDGF-ß), transforming growth factor-beta (TGF-ß) and tumor necrosis factor-alpha (TNF-α) through ELISA. First, we determined the ability of 10% ePL or fibrinogen-depleted lysate (fdePL) compared with 10% FBS to suppress monocyte activation by measuring TNF-α from culture supernatants. We then evaluated proliferation, viability, and immunomodulatory characteristics of bone marrow-derived MSCs (BM-MSCs) cultured in FBS or ePL with or without fibrinogen. Growth factor concentrations decreased in ePL after fibrinogen depletion. Lipopolysaccharide (LPS)-stimulated monocytes exposed to ePL and fdePL produced less TNF-α than LPS-stimulated monocytes in 10% FBS. BM-MSCs cultured in fdePL exhibited lower proliferation rates, but similar viability compared with BM-MSCs in ePL. BM-MSCs in fdePL did not effectively suppress TNF-α expression from LPS-stimulated monocytes compared with BM-MSCs in FBS. Depleting fibrinogen results in a lysate that suppresses TNF-α expression from LPS-stimulated monocytes, but that does not support proliferation and immune-modulatory capacity of BM-MSCs as effectively as nondepleted lysate.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/drug effects , Fibrinogen/metabolism , Mesenchymal Stem Cells/drug effects , Animals , Blood Platelets/metabolism , Cell Extracts/chemistry , Cell Extracts/pharmacology , Cell Proliferation , Cells, Cultured , Culture Media/chemistry , Culture Media/pharmacology , Horses , Humans , Mesenchymal Stem Cells/cytology , Monocytes/cytology , Monocytes/drug effects
2.
Vet Immunol Immunopathol ; 195: 65-71, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29249319

ABSTRACT

Platelet lysate (PL) has been extensively used for the laboratory expansion of human mesenchymal stem cells (MSC) in order to avoid fetal bovine serum (FBS) which has been associated with immune-mediated host reactions and transmission of bovine-origin microbial contaminants. Before suggesting the routine use of PL for MSC culture, we wanted to further investigate whether PL alone might trigger inflammatory responses when exposed to reactive white blood cells such as monocytes. Our objectives were to evaluate the inflammatory profile of equine monocytes cultured with equine PL (ePL) and to determine if ePL can modulate the expression of inflammatory cytokines in lipopolysaccharide (LPS)-stimulated monocytes. In a first experiment, equine monocytes were isolated and incubated with donor horse serum (DHS), FBS, six individual donors ePL or pooled ePL from all horses. In a second experiment, monocytes were stimulated with E. coli LPS in the presence of 1, 5 or 10% DHS and/or pooled ePL. After 6h of incubation, cell culture supernatants were assayed via ELISA for production of the pro-inflammatory cytokines tumor necrosis factor alpha (TNF-α) and Interleukin 1ß (IL-1ß) as well as for the anti-inflammatory Interleukin 10 (IL-10). Equine monocytes incubated with pooled ePL produced significantly less TNF-α and significantly more IL-10 than monocytes incubated in FBS. A statistically significant difference was not identified for the production of IL-1ß. The second experiment showed that pooled ePL added to LPS-stimulated equine monocytes resulted in a significant reduction in TNF-α and IL-1ß production. IL-10 production was not significantly upregulated by the addition of ePL to LPS-stimulated monocytes. Finally, the addition of ePL to LPS-stimulated monocytes in the presence of various concentrations of DHS resulted to statistically significant decrease of TNF-α and IL-1ß compared to the control groups. This is the first study to demonstrate that ePL suppresses the release of pro-inflammatory cytokines from stimulated equine monocytes. These results encourage further exploration of PL as a homologous media substitute for FBS but also opens the possibility of investigating its use as means to suppress cell-mediated inflammation.


Subject(s)
Horses/immunology , Immunity, Innate/immunology , Monocytes/physiology , Animals , Blood Platelets/metabolism , Cells, Cultured , Culture Media , Female , Immunity, Innate/physiology , Male , Monocytes/immunology
3.
J Tissue Eng Regen Med ; 11(10): 2876-2884, 2017 10.
Article in English | MEDLINE | ID: mdl-27339032

ABSTRACT

Immunogenicity of fetal bovine serum (FBS) poses a problem for its use in the propagation of autologous mesenchymal stromal cells (MSCs) for cell therapy. Human platelet lysate (hPL), an enriched growth factor solution containing mitogenic and angiogenic cues, has potential utility in replacing FBS for human MSC (hMSC) delivery strategies. Despite its potentiation of hMSC number in vitro, little is known concerning its capacity to supplement implanted hMSC-seeded constructs and promote tissue regeneration in vivo. In this study, we tested the effects of incorporating hPL in cell-seeded constructs implanted subcutaneously into immunocompromised rats, investigated in vitro interactions between hPL and rat MSCs (rMSCs) and determined interspecies variability in the PL product [hPL vs rat PL (rPL)] and its effect on cultured MSCs (hPL/hMSCs vs rPL/rMSCs). The overarching aim was to determine the utility of hPL to foster MSC survival in preclinical rodent models. Exposure to hPL-supplemented media resulted in rMSC death, by a process attributable to heat-labile proteins, but not membrane attack complex formation. In the in vitro syngeneic model, the rodent product proved fundamentally distinct from the human product, with rPL having substantially lower growth factor content than hPL. Moreover, contrary to the positive effects of hPL on hMSC expansion, rPL did not reduce rMSC doubling time for the serum concentrations examined. When tested in vivo, hPL did not improve cell survival within hydrogel constructs through 2 weeks postimplantation. In summary, this study highlights the many facets of xenogenicity and interspecies variability that must be considered in the preclinical evaluation of hPL. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Blood Platelets/cytology , Cell Extracts/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Disease Models, Animal , Female , Humans , Hydrogels/pharmacology , Immunocompromised Host , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Rats, Nude , Species Specificity , Tissue Scaffolds/chemistry
4.
Stem Cells ; 34(9): 2429-42, 2016 09.
Article in English | MEDLINE | ID: mdl-27299362

ABSTRACT

We have previously demonstrated that cryopreservation and thawing lead to altered Mesenchymal stromal cells (MSC) functionalities. Here, we further analyzed MSC's fitness post freeze-thaw. We have observed that thawed MSC can suppress T-cell proliferation when separated from them by transwell membrane and the effect is lost in a MSC:T-cell coculture system. Unlike actively growing MSCs, thawed MSCs were lysed upon coculture with activated autologous Peripheral Blood Mononuclear Cells (PBMCs) and the lysing effect was further enhanced with allogeneic PBMCs. The use of DMSO-free cryoprotectants or substitution of Human Serum Albumin (HSA) with human platelet lysate in freezing media and use of autophagy or caspase inhibitors did not prevent thaw defects. We tested the hypothesis that IFNγ prelicensing before cryobanking can enhance MSC fitness post thaw. Post thawing, IFNγ licensed MSCs inhibit T cell proliferation as well as fresh MSCs and this effect can be blocked by 1-methyl Tryptophan, an Indoleamine 2,3-dioxygenase (IDO) inhibitor. In addition, IFNγ prelicensed thawed MSCs inhibit the degranulation of cytotoxic T cells while IFNγ unlicensed thawed MSCs failed to do so. However, IFNγ prelicensed thawed MSCs do not deploy lung tropism in vivo following intravenous injection as well as fresh MSCs suggesting that IFNγ prelicensing does not fully rescue thaw-induced lung homing defect. We identified reversible and irreversible cryoinjury mechanisms that result in susceptibility to host T-cell cytolysis and affect MSC's cell survival and tissue distribution. The susceptibility of MSC to negative effects of cryopreservation and the potential to mitigate the effects with IFNγ prelicensing may inform strategies to enhance the therapeutic efficacy of MSC in clinical use. Stem Cells 2016;34:2429-2442.


Subject(s)
Apoptosis , Cryopreservation , Interferon-gamma/pharmacology , Mesenchymal Stem Cells/cytology , T-Lymphocytes/cytology , Animals , Autophagy/drug effects , Caspases/metabolism , Cell Communication/drug effects , Cell Degranulation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Freezing , Heat-Shock Response/drug effects , Humans , Immunosuppression Therapy , Lung/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Polymerization , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/physiology
5.
Acta Biomater ; 36: 86-98, 2016 05.
Article in English | MEDLINE | ID: mdl-26961805

ABSTRACT

UNLABELLED: Mesenchymal stem cells (MSC) hold promise in promoting vascular regeneration of ischemic tissue in conditions like critical limb ischemia of the leg. However, this approach has been limited in part by poor cell retention and survival after delivery. New biomaterials offer an opportunity to localize cells to the desired tissue after delivery, but also to improve cell survival after delivery. Here we characterize the mechanical and microstructural properties of a novel hydrogel composed of pooled human platelet lysate (PL) and test its ability to promote MSC angiogenic activity using clinically relevant in vitro and in vivo models. This PL hydrogel had comparable storage and loss modulus and behaved as a viscoelastic solid similar to fibrin hydrogels despite having 1/4-1/10th the fibrin content of standard fibrin gels. Additionally, PL hydrogels enabled sustained release of endogenous PDGF-BB for up to 20days and were resistant to protease degradation. PL hydrogel stimulated pro-angiogenic activity by promoting human MSC growth and invasion in a 3D environment, and enhancing endothelial cell sprouting alone and in co-culture with MSCs. When delivered in vivo, the combination of PL and human MSCs improved local tissue perfusion after 8days compared to controls when assessed with laser Doppler perfusion imaging in a murine model of hind limb ischemia. These results support the use of a PL hydrogel as a scaffold for MSC delivery to promote vascular regeneration. STATEMENT OF SIGNIFICANCE: Innovative strategies for improved retention and viability of mesenchymal stem cells (MSCs) are needed for cellular therapies. Human platelet lysate is a potent serum supplement that improves the expansion of MSCs. Here we characterize our novel PL hydrogel's desirable structural and biologic properties for human MSCs and endothelial cells. PL hydrogel can localize cells for retention in the desired tissue, improves cell viability, and augments MSCs' angiogenic activity. As a result of these unique traits, PL hydrogel is ideally suited to serve as a cell delivery vehicle for MSCs injected into ischemic tissues to promote vascular regeneration, as demonstrated here in a murine model of hindlimb ischemia.


Subject(s)
Blood Platelets/chemistry , Endothelial Cells/metabolism , Hydrogels/chemistry , Neovascularization, Physiologic , Animals , Cells, Immobilized/metabolism , Cells, Immobilized/transplantation , Coculture Techniques , Disease Models, Animal , Endothelial Cells/transplantation , Female , Heterografts , Hindlimb/blood supply , Hindlimb/metabolism , Humans , Ischemia/therapy , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Mice , Mice, Inbred NOD , Mice, SCID
6.
Mol Ther ; 23(7): 1248-1261, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25899824

ABSTRACT

Autologous bone marrow-derived mesenchymal stromal cells (MSCs) for adoptive cell therapy of luminal Crohn's disease (CD) are being tested in clinical trials. However, CD is associated with dysregulation of autophagy and its effect on MSC's immunobiology is unknown. Here, we demonstrate no quantitative difference in phenotype, in vitro growth kinetics and molecular signatures to IFNγ between MSCs derived from CD and healthy individuals. CD MSCs were indistinguishable from those derived from healthy controls at inhibiting T-cell proliferation through an indoleamine 2,3-dioxygenase (IDO)-dependent mechanism. Upon IFNγ prelicensing, both MSC populations inhibit T-cell effector functions. Neither a single-nucleotide polymorphism (SNP) rs7820268 in the IDO gene, nor a widely reported CD predisposing SNP ATG16L1rs2241880 modulated the suppressive function of MSCs carrying these haplotypes. IFNγ stimulation or coculture with activated T cells upregulated the expression of autophagy genes and/or vacuoles on MSCs. Pharmacological blockade of autophagy pathway did not reverse the immunosuppressive properties and IFNγ responsiveness of MSCs confirming the absence of a functional link between these two cell biochemical properties. We conclude that autophagy, but not IDO and IFNγ responsiveness, is dispensable for MSC's immunosuppressive properties. MSCs from CD subjects are functionally analogous to those of healthy individuals.


Subject(s)
Crohn Disease/immunology , Immunosuppression Therapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interferon-gamma/genetics , Mesenchymal Stem Cells/immunology , Autophagy/immunology , Bone Marrow Cells , Cell Proliferation/genetics , Coculture Techniques , Crohn Disease/genetics , Crohn Disease/pathology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Mesenchymal Stem Cells/pathology , T-Lymphocytes/immunology
7.
Biol Blood Marrow Transplant ; 21(5): 934-40, 2015 May.
Article in English | MEDLINE | ID: mdl-25659882

ABSTRACT

The success of allogeneic hematopoietic stem cell transplantation (allo-HSCT) is often limited by the development of acute and/or chronic graft-versus-host disease (GVHD). The lack of effective therapies to treat steroid-refractory GVHD patients has bolstered clinical evaluation of mesenchymal stromal cell (MSC) therapy for GVHD. Currently, testing of MSCs for the treatment of GVHD has exclusively used allogeneic MSCs despite emerging evidence that MSCs lose their immunoprivileged status in vivo. We hypothesized that autologous MSCs could be a viable alternative MSC source for treating active GVHD. MSCs were isolated and successfully expanded from the bone marrow of 12 volunteers (ages 2 to 55 years) who had allo-HSCT transplants and subsequently developed GVHD. MSCs from subjects with GVHD demonstrated an initial lag in growth compared with healthy control subjects; however, this lag disappeared with continued ex vivo expansion. Immunophenotype and mesodermal differentiation capacity of MSCs from GVHD patients were indistinguishable from that of healthy control MSCs. In vitro immunomodulatory functional analyses also demonstrated that GVHD MSCs were equivalent to healthy control MSCs with regards to dose dependently suppressing T cell proliferation and up-regulating indoleamine 2,3-dioxygenase expression when primed with IFN-γ. Single tandem repeat chimerism analyses further demonstrated that MSCs expanded from GVHD patients were exclusively recipient derived. Based on these data, we conclude that recipient-derived MSCs from patients with GVHD are analogous to MSCs from healthy volunteers and represent a viable option for clinical testing as an immunomodulatory option for symptomatic GVHD.


Subject(s)
Bone Marrow Cells/immunology , Cell Differentiation/immunology , Hematopoietic Stem Cell Transplantation , Immune Tolerance , Mesenchymal Stem Cells/immunology , Acute Disease , Adolescent , Adult , Allografts , Bone Marrow Cells/pathology , Cells, Cultured , Child , Child, Preschool , Chronic Disease , Female , Humans , Male , Mesenchymal Stem Cells/pathology , Middle Aged
8.
Cytotherapy ; 17(4): 382-91, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25595329

ABSTRACT

Mesenchymal stromal cells deploy immune suppressive properties amenable for use as cell therapy for inflammatory disorders. It is now recognized that mesenchymal stromal cells necessitate priming with an inflammatory milieu, in particular interferon-γ, to exert augmented immunosuppressive effects. It has been recently suggested that the heme-catabolizing enzyme heme oxygenase-1 is an essential component of the mesenchymal stromal cell-driven immune suppressive response. Because mesenchymal stromal cells upregulate indoleamine 2,3-dioxygenase expression on interferon-γ priming and indoleamine 2,3-dioxygenase requires heme as a cofactor for optimal catabolic function, we investigated the potential antagonism of heme oxygenase-1 activity on indoleamine 2, 3-dioxygenase and the impact on mesenchymal stromal cell immune plasticity. We herein sought to evaluate the molecular genetic effect of cytokine priming on human mesenchymal stromal cell heme oxygenase-1 expression and its functional role in differentially primed mesenchymal stromal cells. Contrary to previous reports, messenger RNA and protein analyses demonstrated that mesenchymal stromal cells derived from normal subjects (n = 6) do not express heme oxygenase-1 at steady state or after interferon-γ, tumor necrosis factor-α, and/or transforming growth factor-ß priming. Pharmacological inhibition of heme oxygenase-1 with the use of tin protoporphyrin did not significantly abrogate the ability of mesenchymal stromal cells to suppress T-cell proliferation in vitro. Overall, these results unequivocally demonstrate that under steady state and after cytokine priming, human mesenchymal stromal cells immunoregulate T-cell proliferation independent of heme oxygenase-1.


Subject(s)
Heme Oxygenase-1/metabolism , Immune Tolerance , Lymphocyte Activation/immunology , Mesenchymal Stem Cells/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Cell Proliferation , Cell- and Tissue-Based Therapy , Cells, Cultured , Female , Heme/metabolism , Heme Oxygenase-1/antagonists & inhibitors , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation/therapy , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Male , Mesenchymal Stem Cells/enzymology , Metalloporphyrins/pharmacology , Protoporphyrins/pharmacology , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Young Adult
9.
Stem Cell Reports ; 3(1): 60-72, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-25068122

ABSTRACT

Mesenchymal stromal cells have shown clinical promise; however, variations in treatment responses are an ongoing concern. We previously demonstrated that MSCs are functionally stunned after thawing. Here, we investigated whether this cryopreservation/thawing defect also impacts the postinfusion biodistribution properties of MSCs. Under both static and physiologic flow, compared with live MSCs in active culture, MSCs thawed from cryopreservation bound poorly to fibronectin (40% reduction) and human endothelial cells (80% reduction), respectively. This reduction correlated with a reduced cytoskeletal F-actin content in post-thaw MSCs (60% reduction). In vivo, live human MSCs could be detected in murine lung tissues for up to 24 hr, whereas thawed MSCs were undetectable. Similarly, live MSCs whose actin cytoskeleton was chemically disrupted were undetectable at 24 hr postinfusion. Our data suggest that post-thaw cryopreserved MSCs are distinct from live MSCs. This distinction could significantly affect the utility of MSCs as a cellular therapeutic.


Subject(s)
Actins/metabolism , Cryopreservation , Cytoskeleton/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Cell Survival/physiology , Cells, Cultured , Female , Flow Cytometry , Humans , Male , Mice , Real-Time Polymerase Chain Reaction
10.
J Immunol ; 192(4): 1491-501, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24403533

ABSTRACT

Human bone marrow-derived mesenchymal stromal cells (MSCs) inhibit proliferation of activated T cells, and IFN-γ plays an important role in this process. This IFN-γ-licensed veto property is IDO-dependent. To further decipher the mechanistic underpinnings of MSC veto function on T cells, we investigated the effect of MSCs and IFN-γ-licensed MSCs on T cell effector function as assayed by cytokine secretion of T cells. Although MSCs and IFN-γ-licensed MSCs inhibit T cell proliferation, only IFN-γ-licensed MSCs significantly inhibit Th1 cytokine (IFN-γ, TNF-α, and IL-2) production by T cells. Additionally, IFN-γ-licensed MSCs inhibit T cell degranulation as well as single, double, and triple cytokine-producing T cells. Although IFN-γ-licensed MSCs upregulate their IDO activity, we found that MSC IDO catalytic function is dispensable with regard to MSC-driven inhibition of T cell effector function. Novel flow cytometry based functional screening of MSC-expressed, IFN-γ-licensed inhibitory molecules identified B7H1 and B7DC/PD1 pathways as essential effectors in blocking T cell function. Small interfering RNA-mediated blocking of B7H1 and B7DC reverses the inhibitory potential of IFN-γ-licensed MSCs on T cell effector function. Mechanistic analysis revealed that clustering of MHC and coinhibitory molecules are indispensable for the inhibitory effect of IFN-γ MSCs. Although exogenous IL-2 reverses B7H1-Ig-mediated inhibition of T cell proliferation, it does not affect the veto function of IFN-γ MSCs on both T cell proliferation and effector function. Our results reveal a new immunosuppressive property of IFN-γ-licensed MSCs that inhibits T cell effector function independent of IDO but through the ligands for PD1.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/metabolism , Mesenchymal Stem Cells/metabolism , Th1 Cells/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Bone Marrow Cells , Cell Degranulation/immunology , Cell Proliferation , Cells, Cultured , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Lymphocyte Activation/immunology , Mesenchymal Stem Cells/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , RNA Interference , RNA, Small Interfering , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/biosynthesis
11.
Cytotherapy ; 15(12): 1469-83, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23981539

ABSTRACT

BACKGROUND AIMS: Multipotent mesenchymal stromal cells (MSCs) are clinically useful because of their immunomodulatory and regenerative properties, but MSC therapies are limited by the loss of self-renewal and cell plasticity associated with ex vivo expansion culture and, on transplantation, increased immunogenicity from xenogen exposure during culture. Recently, pooled human platelet lysate (hPL) has been used as a culture supplement to promote MSC growth; however, the effects of hPL on MSCs after fetal bovine serum (FBS) exposure remain unknown. METHODS: MSCs were cultured in medium containing FBS or hPL for up to 16 passages, and cell size, doubling time and immunophenotype were determined. MSC senescence was assessed by means of a fluorometric assay for endogenous ß-galactosidase expression. MSCs cultured with FBS for different numbers of passages were switched to hPL conditions to evaluate the ability of hPL to "rescue" the proliferative capacity of MSCs. RESULTS: hPL culture resulted in more rapid cell proliferation at earlier passages (passage 5 or earlier) than remove FBS; by day 4, hPL (5%) yielded an MSC doubling time of 1.28 days compared with 1.52 days in 16% FBS. MSCs cultured first in FBS and switched to hPL proliferated more and demonstrated less ß-galactosidase production and smaller cell sizes than remove MSCs continuously propagated in FBS. CONCLUSIONS: hPL enables rapid expansion of MSCs without adversely affecting immunophenotype. hPL culture of aged and senescent MSCs demonstrated cellular rejuvenation, reflected by decreased doubling time and smaller cell size. These results suggest that expansion of MSCs in hPL after FBS exposure can enhance cell phenotype and proliferative capacity.


Subject(s)
Cellular Senescence/drug effects , Culture Media/chemistry , Mesenchymal Stem Cells/cytology , Platelet-Rich Plasma/chemistry , Animals , Cattle , Cell Culture Techniques , Cell Proliferation , Flow Cytometry , Humans , Serum/chemistry
12.
Biomaterials ; 34(32): 7840-50, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23891515

ABSTRACT

Human platelet lysate (PL) represents an attractive alternative to fetal bovine serum (FBS) for the ex vivo expansion of human mesenchymal stromal cells (MSCs). However, there is controversy whether MSCs propagated in unfractionated PL retain their immunosuppressive properties. Since fibrinogen can be a major component of PL, we hypothesized that the fibrinogen content in PL negatively affects the suppressor function of MSCs. Pools of outdated plateletpheresis products underwent a double freeze-thaw centrifugation and filtration to produce unfractionated platelet lysates (uPL), followed by a temperature controlled clotting procedure to produce a fibrinogen depleted platelet lysate (fdPL). Fibrinogen depletion affected neither the mitogenic properties of PL or growth factor content, however fdPL was less prone to develop precipitate over time. Functionally, fibrinogen interacted directly with MSCs, dose dependently increased IL-6, IL-8 and MCP-1 protein production, and compromised the ability of MSCs to up-regulate indoleamine dioxygenase (IDO), as well as, mitigate T-cell proliferation. Similarly uPL expanded MSCs showed a reduced capability of inducing IDO and suppressing T-cell proliferation compared to FBS expanded MSCs. Replacing uPL with fdPL largely restored the immune modulating effects of MSCs. Together these data suggest that fibrinogen negatively affects the immunomodulatory functions of MSCs and fdPL can serve as non-xenogenic mitogenic supplement for expansion of clinical grade MSCs for immune modulation.


Subject(s)
Blood Platelets/chemistry , Fibrinogen/metabolism , Immunotherapy , Mesenchymal Stem Cells/drug effects , Cell Culture Techniques , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CCL2/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Mesenchymal Stem Cells/metabolism , T-Lymphocytes/metabolism , Up-Regulation
13.
Cytotherapy ; 14(2): 147-52, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22029655

ABSTRACT

Human mesenchymal stromal cells (MSC) can suppress T-cell activation in vitro in an indoleamine 2,3-dioxygenase (IDO)-dependent manner. However, their clinical effects on immune ailments have been inconsistent, with a recent phase III study showing no benefit in acute graft-versus-host disease (GvHD). We here tested the hypothesis that the banked, cryopreserved MSC often used in clinical trials display biologic properties distinct from that of MSC in the log phase of growth typically examined in pre-clinical studies. In freshly thawed cryopreserved MSC derived from normal human volunteers, we observed that MSC up-regulate heat-shock proteins, are refractory to interferon (IFN)-γ-induced up-regulation of IDO, and are compromised in suppressing CD3/CD28-driven T cell proliferation. Immune suppressor activity, IFN-γ responsiveness and induction of IDO were fully restored following 24 h of MSC tissue culture post-thaw. These results highlight a possible cause for the inefficacy of MSC-based immunotherapy reported in clinical trials using cryopreserved MSC thawed immediately prior to infusion.


Subject(s)
Cryopreservation , Heat-Shock Response , Immunosuppression Therapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/immunology , Mesenchymal Stem Cells/immunology , T-Lymphocytes/immunology , Cell Culture Techniques , Cell Proliferation , Cell Survival , Cells, Cultured , Clinical Trials as Topic , Gene Expression , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interferon-gamma/metabolism , Lymphocyte Activation , Mesenchymal Stem Cells/cytology
14.
J Cardiovasc Dis Res ; 2(1): 3-13, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21716750

ABSTRACT

The fields of regenerative medicine and cellular therapy have been the subject of tremendous hype and hope. In particular, the perceived usage of somatic cells like mesenchymal stromal cells (MSCs) has captured the imagination of many. MSCs are a rare population of cells found in multiple regions within the body that can be readily expanded ex vivo and utilized clinically. Originally, it was hypothesized that transplantation of MSCs to sites of injury would lead to de novo tissue-specific differentiation and thereby replace damaged tissue. Now, it is generally agreed that MSC home to sites of injury and direct positive remodeling via the secretion of paracrine factors. Consequently, their clinical utilization has largely revolved around their abilities to promote neovascularization for ischemic disorders and modulate overly exuberant inflammatory responses for autoimmune and alloimmune conditions. One of the major issues surrounding the development of somatic cell therapies like MSCs is that despite evoking a positive response, long-term engraftment and persistence of these cells is rare. Consequently, very large cell doses need be administered for raising production, delivery, and efficacy issues. In this review, we will outline the field of MSC in the context of ischemia and discuss causes for their lack of persistence. In addition, some of the methodologies be used to enhance their therapeutic potential will be highlighted.

15.
Semin Immunopathol ; 33(6): 535-50, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21533908

ABSTRACT

The fields of regenerative medicine and cellular therapy have been the subject of tremendous hype and hope. In particular, the perceived usage of somatic cells like mesenchymal stromal stem cells (MSCs) has captured the imagination of many. Clinical trials are currently evaluating the therapeutic efficacy of MSCs in disorders ranging from heart disease to pediatric graft-vs-host disease; however, numerous questions still remain regarding mechanism of action, effective dose, and whether these cells can be used in the allogeneic setting. One of the major issues surrounding the development of somatic cell therapies like MSCs is that despite evoking a positive response, long-term engraftment and persistence of these cells is rare. Consequently, very large cell doses need be administered raising production, delivery, and efficacy issues. In this review, we will discuss causes for this lack of persistence and highlight some of the methodologies be used to enhance cell survival post-transplantation.


Subject(s)
Inflammation/immunology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Animals , Cell Death/immunology , Cytotoxicity, Immunologic , Granzymes/metabolism , Humans , Inflammation/metabolism , Mesenchymal Stem Cell Transplantation/adverse effects , Perforin/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
16.
Ann Biomed Eng ; 38(9): 2791-804, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20387119

ABSTRACT

Leukocytes play a pivotal role in the progression of atherosclerosis. A novel three-dimensional in vitro asymmetric stenosis model was used to better investigate the role of local hemodynamics in the adhesion of leukocytes to an established plaque. The adhesion of a human promyelocytic cell line (NB4) on a human abdominal aortic endothelial cell (EC) monolayer was quantified. NB4 cells were circulated over TNF-alpha stimulated and nonstimulated ECs for 1 or 6 h at 1.25 or 6.25 dynes/cm(2) and compared to static conditions. Cytokine stimulation increased significantly EC expression of intercellular adhesion molecule and vascular cell adhesion molecule. Under static conditions, neutrophils adhered overall more than under flow, with decreased adhesion with increasing shear. Adhesion was significantly higher in the recirculation region distal to the stenosis than in the inlet. Preshearing the ECs decreased the expression of cell adhesion molecules in inflamed endothelium and significantly decreased adhesion. However, the ratio of adhesion between the recirculation zone and the inlet increased, hence exhibiting an increased regional difference. This work suggests an important role for neutrophil-EC interactions in the atherosclerotic process, especially in wall shear stress gradient regions. This is important clinically, potentially helping to explain plaque stability.


Subject(s)
Atherosclerosis/physiopathology , Endothelium, Vascular/physiopathology , Hemodynamics , Neutrophils/physiology , Shear Strength/physiology , Aorta, Abdominal/physiopathology , Cell Adhesion/physiology , Cell Adhesion Molecules/analysis , Cell Line, Tumor , Cells, Cultured , Constriction, Pathologic/physiopathology , Humans , Male , Stress, Mechanical , Tumor Necrosis Factor-alpha/physiology , Young Adult
17.
Stem Cells ; 27(2): 467-77, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19338064

ABSTRACT

Mesenchymal stromal cells (MSCs) display robust reparative properties through their ability to limit apoptosis, enhance angiogenesis, and direct positive tissue remodeling. However, low in vivo survival of transplanted cells limits their overall effectiveness and significantly affects their clinical usage. Consequently, identifying strategies to improve cell survival in vivo are a priority. One explanation for their low survival is that MSCs are often transplanted into ischemic tissue, such as infarcted myocardium, where there is poor blood supply and low oxygen tension. Therefore, we examined how MSCs respond to a hypoxic, nutrient-poor stress environment to identify trophic factors that could be manipulated in advance of MSC transplantation. Combining microarray and proteomic screens we identified plasminogen activator inhibitor 1 (PAI-1) as one factor consistently upregulated in our in vitro ischemia-mimicking conditions. Subsequent genetic and chemical manipulation studies define PAI-1 as a negative regulator of MSC survival in vivo. Mechanistically, MSC-derived PAI-1 does not alter MSC survival through a plasmin-dependent mechanism but rather directly impacts on the adhesiveness of MSCs to their surrounding matrices. Thus we can conclude that post-transplantation, PAI-1 negatively impacts MSC survival by promoting anoikis via matrix detachment.


Subject(s)
Mesenchymal Stem Cell Transplantation , Plasminogen Activator Inhibitor 1/physiology , Animals , Blotting, Western , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Movement/genetics , Cell Movement/physiology , Cells, Cultured , Female , Humans , Mice , Mice, Inbred C57BL , Myocardial Ischemia , Oligonucleotide Array Sequence Analysis , Plasminogen Activator Inhibitor 1/metabolism , Proteomics , Reverse Transcriptase Polymerase Chain Reaction
18.
Stem Cells ; 27(3): 489-97, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19074414

ABSTRACT

Mesenchymal stromal cell (MSC) markers are expressed on brain tumor-initiating cells involved in the development of hypoxic glioblastoma. Given that MSCs can survive hypoxia and that the glucose-6-phosphate transporter (G6PT) provides metabolic control that contributes to MSC mobilization and survival, we investigated the effects of low oxygen (1.2% O(2)) exposure on G6PT gene expression. We found that MSCs significantly expressed G6PT and the glucose-6-phosphatase catalytic subunit beta, whereas expression of the glucose-6-phosphatase catalytic subunit alpha and the islet-specific glucose-6-phosphatase catalytic subunit-related protein was low to undetectable. Analysis of the G6PT promoter sequence revealed potential binding sites for hypoxia inducible factor (HIF)-1alpha and for the aryl hydrocarbon receptor (AhR) and its dimerization partner, the AhR nuclear translocator (ARNT), AhR:ARNT. In agreement with this, hypoxia and the hypoxia mimetic cobalt chloride induced the expression of G6PT, vascular endothelial growth factor (VEGF), and HIF-1alpha. Gene silencing of HIF-1alpha prevented G6PT and VEGF induction in hypoxic MSCs whereas generation of cells stably expressing HIF-1alpha resulted in increased endogenous G6PT gene expression. A semisynthetic analog of the polyketide mumbaistatin, a potent G6PT inhibitor, specifically reduced MSC-HIF-1alpha cell survival. Collectively, our data suggest that G6PT may account for the metabolic flexibility that enables MSCs to survive under conditions characterized by hypoxia and could be specifically targeted within developing tumors.


Subject(s)
Anthraquinones/pharmacology , Gene Expression/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mesenchymal Stem Cells/metabolism , Monosaccharide Transport Proteins/physiology , Stromal Cells/drug effects , Stromal Cells/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Hypoxia/drug effects , Cell Movement , Cells, Cultured , Cobalt/pharmacology , Electrophoretic Mobility Shift Assay , Female , Glucose-6-Phosphatase/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoblotting , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Monosaccharide Transport Proteins/antagonists & inhibitors , Monosaccharide Transport Proteins/genetics , Necrosis/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/cytology
19.
Am J Physiol Renal Physiol ; 295(2): F488-96, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18524862

ABSTRACT

We previously demonstrated that erythropoietin (EPO)-secreting mesenchymal stromal cells (MSC) can be used for the long-term correction of renal failure-induced anemia. The present study provides evidence that coimplantation of insulin-like growth factor I (IGF-I)-overexpressing MSC (MSC-IGF) improves MSC-based gene therapy of anemia by providing paracrine support to EPO-secreting MSC (MSC-EPO) within a subcutaneous implant. IGF-I receptor RNA expression in murine MSC was demonstrated by RT-PCR. Functional protein expression was confirmed by immunoblots and MSC responsiveness to IGF-I stimulation in vitro. IGF-I was also shown to improve MSC survival following staurosporin-induced apoptosis in vitro. A cohort of C57Bl/6 mice was rendered anemic by right kidney electrocoagulation and left nephrectomy. MSC-EPO were subsequently admixed in a bovine collagen matrix and implanted, in combination with MSC-IGF or MSC null, by subcutaneous injection in renal failure mice. In mice receiving MSC-EPO coimplanted with MSC-IGF, hematocrit elevation was greater and enhanced compared with control mice; heart function was also improved. MSC-IGF coimplantation, therefore, represents a promising new strategy for enhancing MSC survival within implanted matrices and for improving cell-based gene therapy of renal anemia.


Subject(s)
Anemia/therapy , Genetic Engineering/methods , Genetic Therapy/methods , Insulin-Like Growth Factor I/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Renal Insufficiency/therapy , Anemia/etiology , Anemia/metabolism , Animals , Apoptosis , Cells, Cultured , Disease Models, Animal , Erythropoietin/metabolism , Female , Hematocrit , Insulin-Like Growth Factor I/genetics , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Receptor, IGF Type 1/metabolism , Renal Insufficiency/complications , Renal Insufficiency/metabolism
20.
Cardiovasc Res ; 79(3): 405-15, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18397963

ABSTRACT

AIMS: Mesenchymal stromal cells (MSCs) possess intrinsic features that identify them as useful for treating ischaemic syndromes. Poor in vivo survival/engraftment of MSCs, however, limits their overall effectiveness. In this work, we tested whether genetically engineering MSCs to secrete erythropoietin (Epo) could represent a better therapeutic platform than MSCs in their native form. METHODS AND RESULTS: MSCs from C57Bl/6 mice were retrovirally transduced with either an empty vector or one that causes the production of Epo and were then analysed for the alterations in angiogenic and survival potential. Using a mouse model of myocardial infarction (MI), the regenerative potential of null MSCs and Epo-overexpressing MSCs (Epo+MSCs) was assessed using serial echocardiogram and invasive haemodynamic measurements. Infarct size, capillary density and neutrophil influx were assessed using histologic techniques. Using in vitro assays coupled with an in vivo Matrigel plug assay, we demonstrate that engineering MSCs to express Epo does not alter their immunophenotype or plasticity. However, relative to mock-modified MSCs [wild-type (WT)-MSCs], Epo+MSCs are more resilient to apoptotic stimuli and initiate a more robust host-derived angiogenic response. We also identify and characterize the autocrine loop established on MSCs by having them secrete Epo. Furthermore, in a murine model of MI, animals receiving intracardiac injections of Epo+MSCs exhibited significantly enhanced cardiac function compared with WT-MSCs and saline-injected control animals post-MI, owing to the increased myocardial capillary density and the reduced neutrophilia. CONCLUSION: Epo overexpression enhances the cellular regenerative properties of MSCs by both autocrine and paracrine pathways.


Subject(s)
Erythropoietin/biosynthesis , Genetic Therapy/methods , Myocardial Infarction/therapy , Myocardium/metabolism , Regeneration , Stromal Cells/transplantation , Animals , Apoptosis , Autocrine Communication , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Disease Models, Animal , Erythropoietin/genetics , Female , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/pathology , Neovascularization, Physiologic , Neutrophil Infiltration , Paracrine Communication , Stromal Cells/metabolism , Transduction, Genetic , Ventricular Function, Left , Ventricular Remodeling
SELECTION OF CITATIONS
SEARCH DETAIL
...