Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
NPJ Regen Med ; 5: 3, 2020.
Article in English | MEDLINE | ID: mdl-32133156

ABSTRACT

Therapeutic approaches requiring the intravenous injection of autologous or allogeneic mesenchymal stromal cells (MSCs) are currently being evaluated for treatment of a range of diseases, including orthopaedic injuries. An alternative approach would be to mobilise endogenous MSCs into the blood, thereby reducing costs and obviating regulatory and technical hurdles associated with development of cell therapies. However, pharmacological tools for MSC mobilisation are currently lacking. Here we show that ß3 adrenergic agonists (ß3AR) in combination with a CXCR4 antagonist, AMD3100/Plerixafor, can mobilise MSCs into the blood in mice and rats. Mechanistically we show that reversal of the CXCL12 gradient across the bone marrow endothelium and local generation of endocannabinoids may both play a role in this process. Using a spine fusion model we provide evidence that this pharmacological strategy for MSC mobilisation enhances bone formation.

2.
Blood Adv ; 1(22): 1934-1943, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29296840

ABSTRACT

Pharmacological mobilization of hematopoietic progenitor cells (HPCs) is used clinically to harvest HPCs for bone marrow transplants. It is now widely accepted that the CXCR4:CXCL12 chemokine axis plays a critical role in the retention of HPCs in the bone marrow, and CXCR4 antagonists have been developed for their mobilization. The first of this class of drugs to be US Food and Drug Administration-approved was the bicyclam AMD3100. In addition to mobilizing HPCs and leukocytes in naïve mice, AMD3100 has been shown to mobilize mesenchymal progenitor cells (MPCs) in vascular endothelial growth factor (VEGF-A) pretreated mice. AMD3100 binds to the transmembrane region of CXCR4 and is thought to mobilize HPCs by reversing the gradient of CXCL12 across the bone marrow endothelium. Consistent with this hypothesis, our data show that selective neutralization of CXCL12, with chalcone 4-phosphate (C4P), inhibited AMD3100-stimulated mobilization of HPCs and leukocytes in naïve mice and MPCs in VEGF-A pretreated mice. In contrast it is shown here that the CXCR4 antagonist KRH3955 that binds to the extracellular loop of CXCR4 does not reverse the CXCL12 chemokine gradient. However, this drug efficiently mobilizes HPCs, a response that is not inhibited by C4P. In contrast, KRH3955 does not mobilize MPCs in VEGF-A pretreated mice. These data suggest that CXCR4 antagonists that bind to distinct regions of the receptor mobilize progenitor cells by distinct molecular mechanisms.

3.
Cytotherapy ; 16(2): 191-202, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24438900

ABSTRACT

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs) suppress T-cell proliferation, especially after activation with inflammatory cytokines. We compared the dynamic action of unprimed and interferon (IFN)-γ plus tumor necrosis factor (TNF)-α-pretreated human bone marrow-derived MSCs on resting or activated T cells. METHODS: MSCs were co-cultured with allogeneic peripheral blood mononuclear cells (PBMCs) at high MSC-to-PBMC ratios in the absence or presence of concomitant CD3/CD28-induced T-cell activation. The kinetic effects of MSCs on cytokine production and T-cell proliferation, cell cycle and apoptosis were assessed. RESULTS: Unprimed MSCs increased the early production of IFN-γ and interleukin (IL)-2 by CD3/CD28-activated PBMCs before suppressing T-cell proliferation. In non-activated PBMC co-cultures, low levels of IL-2 and IL-10 synthesis were observed with MSCs in addition to low levels of CD69 expression by T cells and no T-cell proliferation. MSCs also decreased apoptosis in resting and activated T cells and inhibited the transition of these cells into the sub-G0/G1 and the S phases. With inhibition of indoleamine 2,3 dioxygenase, MSCs increased CD3/CD28-induced T-cell proliferation. After priming with IFN-γ plus TNF-α, MSCs were less potent at increasing cytokine production by CD3/CD28-activated PBMCs and more effective at inhibiting T-cell proliferation but had preserved anti-apoptotic functions. CONCLUSIONS: Unprimed MSCs induce a transient increase in IFN-γ and IL-2 synthesis by activated T cells. Pre-treatment of MSCs with IFN-γ plus TNF-α may increase their effectiveness and safety in vivo.


Subject(s)
Bone Marrow Cells/cytology , Mesenchymal Stem Cells/immunology , T-Lymphocytes/immunology , Adult , Aged , Antigens, CD/metabolism , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Female , Humans , Immunosuppression Therapy , Inflammation Mediators/metabolism , Lymphocyte Activation , Male , Middle Aged
4.
Cytotherapy ; 14(10): 1164-70, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22574720

ABSTRACT

BACKGROUND AIMS: Apoptosis of radiosensitive cells in the bone marrow and gut is a serious, at times life-threatening, complication arising from radiation exposure. METHODS: We investigated whether adoptive transfer of allogeneic bone marrow-derived mesenchymal stromal cells (MSC) could exert cytoprotective and life-sparing effects in a mouse model of sublethal total body irradiation (TBI). RESULTS: We demonstrated that a single intraperitoneal injection of C57Bl/6 MSC given to major histocompatibility complex (MHC)-mismatched Balb/c mice within 24 h of sublethal TBI significantly reduced mortality in a dose-dependent manner. Histologic analysis and Ki67 immunostaining of jejunum sections collected 3 and 6 days post-TBI indicated that MSC protected the gastrointestinal epithelium from TBI-induced damage and significantly accelerated recovery of the gut by stimulating proliferation of the crypt cell pool. Using interleukin-6(-/-) (IL-6) MSC, we demonstrated that IL-6 expressed by MSC played a role in gastrointestinal epithelium regeneration. CONCLUSIONS: Our results suggest that allogeneic MHC-mismatched MSC may be exploited to reduce gastrointestinal complications and mortality arising from ionizing radiation exposure.


Subject(s)
Adoptive Transfer , Interleukin-6/metabolism , Intestinal Mucosa/physiopathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Regeneration/radiation effects , Whole-Body Irradiation , Animals , Bone Marrow Cells/cytology , Intestinal Mucosa/radiation effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous
5.
J Cell Physiol ; 227(11): 3535-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22378308

ABSTRACT

The discovery of the immunosuppressive properties of mesenchymal stromal cells (MSCs) has given new hope to patients suffering from autoimmune diseases as their lack of immunogenicity in addition to their immunosuppressive and regenerative properties makes them an ideal biological agent for the treatment of various disorders ranging from autoimmune diseases to tissue injury. The translational promises of a safer and more effective therapy has however suffered a setback with the recent release of the results from Phase III randomized clinical trial using MSCs for treatment of steroid refractory acute graft-versus-host disease (GvHD), which failed to meet its primary efficacy endpoint. In this review, we will address the current knowledge of the immunosuppressive mechanisms of MSCs from in vitro studies to animal models and then look upon the results obtained from human clinical trials in order to provide failure analysis of negative studies. We will conclude by proposing future directions which could help address this issue and allow rational development of MSCs as an effective and useful cell-based immunotherapeutic.


Subject(s)
Autoimmune Diseases/therapy , Immunosuppression Therapy/trends , Mesenchymal Stem Cells , Regeneration/genetics , Animals , Cell- and Tissue-Based Therapy/methods , Graft vs Host Disease/therapy , Humans , Immunosuppression Therapy/methods , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Randomized Controlled Trials as Topic , Regeneration/physiology , Translational Research, Biomedical
6.
Mol Ther ; 20(1): 187-95, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21934657

ABSTRACT

Clinical trials testing the use of either autologous or allogeneic human bone marrow-derived mesenchymal stromal cells (MSC) as a cell-based pharmaceutical for suppression of autoimmune and alloimmune ailments are underway. Reported results from completed trials vary in effectiveness within and between studies without any clear mechanistic explanation. We propose that these discrepancies may arise from intrinsic variability in the immunosuppressive potential of each MSC donor source. Here, we demonstrate that tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ)-activated MSC derived from normal adult volunteers suppress T cell proliferation in vitro in a variegated manner, an observation linked to IFN-mediated indoleamine 2,3-dioxygenase (IDO) upregulation. We also demonstrate that MSC IDO activity is implicated in the differentiation of monocytes into interleukin (IL)-10-secreting M2 immunosuppressive macrophages (CD14(+)/CD206(+)). Those monocyte-derived M2 are in turn implicated in the suppression of T cell proliferation in an IL-10-independent manner, thus amplifying the immunosuppressive effect generated by MSC. In summary, the immune plasticity of IFN-γ and TNF-α licensed veto function of MSC vary among donors and defines a central role to inducible IDO activity and its bystander effect on lymphomyeloid immune effectors.


Subject(s)
Cell Differentiation , Cytokines/pharmacology , Immune Tolerance , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Macrophages/cytology , Mesenchymal Stem Cells/enzymology , Aged , Bystander Effect , Cells, Cultured , Female , Humans , Immune Tolerance/drug effects , Immune Tolerance/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Male , Mesenchymal Stem Cells/drug effects , Middle Aged , Models, Biological , Monocytes/cytology , Monocytes/immunology , T-Lymphocytes/immunology , Tissue Donors
7.
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
8.
Blood ; 117(25): 6801-12, 2011 Jun 23.
Article in English | MEDLINE | ID: mdl-21527526

ABSTRACT

Bone-derived mesenchymal stem cells (MSCs) are important cells for use in cell therapy, tissue engineering, and regenerative medicine, but also to study bone development, homeostasis, and repair. However, little is known about their developmental ontology and in vivo identity. Because fibroblast growth factors (FGFs) play key roles in bone development and their receptors are developmentally regulated in bones, we hypothesized that MSCs should express FGF receptors (FGFRs), reflecting their developmental origin and potential. We show here that FGFR1/2 are expressed by rare mesenchymal progenitors in putative MSC niches in vivo, including the perichondrium, periosteum, and trabecular marrow. FGFR1⁺ cells often appeared as pericytes. These cells display a characteristic MSC phenotype in vitro when expanded with FGF-2, which appears to maintain MSC stemness by inhibiting cellular senescence through a PI3K/AKT-MDM2 pathway and by promoting proliferation. FGFRs may therefore be involved in MSC self-renewal. In summary, FGFR1/2 are developmentally regulated markers of MSCs in vivo and in vitro and are important in maintaining MSC stemness.


Subject(s)
Cellular Senescence , Gene Expression Regulation, Developmental , Mesenchymal Stem Cells/cytology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Animals , Bone and Bones/cytology , Bone and Bones/embryology , Bone and Bones/metabolism , Cell Proliferation , Cells, Cultured , Female , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism
9.
Biomaterials ; 32(1): 295-305, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20864158

ABSTRACT

Gene therapy for hemophilia B and other hereditary plasma protein deficiencies showed great promise in pre-clinical and early clinical trials. However, safety concerns about in vivo delivery of viral vectors and poor post-transplant survival of ex vivo modified cells remain key hurdles for clinical translation of gene therapy. We here describe a 3D scaffold system based on porous hydroxyapatite-PLGA composites coated with biomineralized collagen 1. When combined with autologous gene-engineered factor IX (hFIX) positive mesenchymal stem cells (MSCs) and implanted in hemophilic mice, these scaffolds supported long-term engraftment and systemic protein delivery by MSCs in vivo. Optimization of the scaffolds at the macro-, micro- and nanoscales provided efficient cell delivery capacity, MSC self-renewal and osteogenesis respectively, concurrent with sustained delivery of hFIX. In conclusion, the use of gene-enhanced MSC-seeded scaffolds may be of practical use for treatment of hemophilia B and other plasma protein deficiencies.


Subject(s)
Genetic Therapy/methods , Hemophilia B/therapy , Mesenchymal Stem Cells/metabolism , Tissue Scaffolds/chemistry , Animals , Calcium Phosphates/pharmacology , Cell Lineage/drug effects , Cell Proliferation/drug effects , Ceramics/pharmacology , Factor IX/genetics , Factor IX/therapeutic use , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/ultrastructure , Mice , Nanoparticles/ultrastructure , Particle Size , Porosity/drug effects
10.
Cancer Res ; 70(20): 7742-7, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20924101

ABSTRACT

It is unknown whether mesenchymal stromal cells (MSC) can regulate immune responses targeting tumor autoantigens of low immunogenicity. We tested here whether immunization with MSC could break immune tolerance towards the ErbB-2/HER-2/neu tumor antigen and the effects of priming with IFN-γ and tumor necrosis factor-α (TNF-α) on this process. BALB/c- and C57BL/6-derived MSC were lentivirally transduced to express a kinase-inactive rat neu mutant (MSC/Neu). Immunization of BALB/c mice with nontreated or IFN-γ-primed allogeneic or syngeneic MSC/Neu induced similar levels of anti-neu antibody titers; however, only syngeneic MSC/Neu induced protective neu-specific CD8(+) T cell responses. Compared to immunization with nontreated or IFN-γ-primed syngeneic MSC/Neu, the number of circulating neu-specific CD8(+) T cells and titers of anti-neu antibodies were observed to be decreased after immunizations with IFN-γ- plus TNF-α-primed MSC/Neu. In addition, syngeneic MSC/Neu seemed more efficient than IFN-γ-primed MSC/Neu at inducing a protective therapeutic antitumor immune response resulting in the regression of transplanted neu-expressing mammary tumor cells. In vitro antigen-presenting cell assays performed with paraformaldehyde-fixed or live MSC showed that priming with IFN-γ plus TNF-α, compared to priming with IFN-γ alone, increased antigen presentation as well as the production of immunosuppressive factors. These data suggest that whereas MSC could effectively serve as antigen-presenting cells to induce immune responses aimed at tumor autoantigens, these functions are critically regulated by IFN-γ and TNF-α.


Subject(s)
Breast Neoplasms/immunology , Interferon-gamma/therapeutic use , Mammary Neoplasms, Experimental/immunology , Mesenchymal Stem Cells/immunology , Receptor, ErbB-2/biosynthesis , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Breast Neoplasms/therapy , Cancer Vaccines/therapeutic use , Female , Humans , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Promoter Regions, Genetic , Rats , Stromal Cells/immunology , Stromal Cells/pathology
11.
Ann Neurol ; 68(4): 540-5, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20661924

ABSTRACT

Human mesenchymal stem cells (hMSCs) are being considered for clinical trials of multiple sclerosis (MS). We examined the effects of adult bone marrow-derived hMSCs on responses of primary human Th1, Th17, and Th1/17 double-expressing T-cell subsets, all implicated in MS. As expected, soluble products from hMSCs inhibited Th1 responses; however, Th17 responses were increased. Secretion of interleukin (IL)-10, considered anti-inflammatory, was decreased. Pretreating hMSCs with the proinflammatory cytokine IL-1ß accentuated these effects, and caused decreases in the Th1/17 subset. These findings underscore the importance of further preclinical work and immune-monitoring to define hMSC effects on disease-relevant immune responses under variable conditions.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Mesenchymal Stem Cells/immunology , Th1 Cells/cytology , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cytokines/immunology , Cytokines/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry/methods , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Mesenchymal Stem Cells/chemistry , T-Lymphocytes, Helper-Inducer/classification , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology
12.
Blood ; 114(13): 2632-8, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19654411

ABSTRACT

Recent studies involving bone marrow mesenchymal stromal cells (MSCs) demonstrated that interferon (IFN)-gamma stimulation induces major histocompatibility complex (MHC) class II-mediated antigen presentation in MSCs both in vitro and in vivo. Concordantly, we investigated the ability of MSCs to present extracellular antigen through their MHC class I molecules, a process known as cross-presentation. Using an in vitro antigen presentation assay, we demonstrated that murine MSCs can cross-present soluble ovalbumin (OVA) to naive CD8(+) T cells from OT-I mice. Cross-presentation by MSC was proteasome dependent and partly dependent on transporter associated with antigen-processing molecules. Pretreatment of MSC with IFN-gamma increased cross-presentation by up-regulating antigen processing and presentation. However, although the transcription of the transporter associated with antigen processing-1 molecules and the immunoproteasome subunit LMP2 induced by IFN-gamma was inhibited by transforming growth factor-beta, the overall cross-presentation capacity of MSCs remained unchanged after transforming growth factor-beta treatment. These observations were validated in vivo by performing an immune reconstitution assay in beta(2)-microglobulin(-/-) mice and show that OVA cross-presentation by MSCs induces the proliferation of naive OVA-specific CD8(+) T cells. In conclusion, we demonstrate that MSCs can cross-present exogenous antigen and induce an effective CD8(+) T-cell immune response, a property that could be exploited as a therapeutic cell-based immune biopharmaceutic for the treatment of cancer or infectious diseases.


Subject(s)
Antigen-Presenting Cells/immunology , Antigens/immunology , Cross-Priming/immunology , Mesenchymal Stem Cells/immunology , Stromal Cells/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/physiology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Female , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Solubility , Stromal Cells/metabolism , Stromal Cells/physiology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology , beta 2-Microglobulin/genetics
13.
J Immunol ; 182(12): 7963-73, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494321

ABSTRACT

Bone marrow-derived mesenchymal stromal cells (MSC) possess an immune plasticity manifested by either an immunosuppressive or, when activated with IFN-gamma, an APC phenotype. Herein, TLR expression by MSC and their immune regulatory role were investigated. We observed that human MSC and macrophages expressed TLR3 and TLR4 at comparable levels and TLR-mediated activation of MSC resulted in the production of inflammatory mediators such as IL-1beta, IL-6, IL-8/CXCL8, and CCL5. IFN-alpha or IFN-gamma priming up-regulated production of these inflammatory mediators and expression of IFNB, inducible NO synthase (iNOS), and TRAIL upon TLR activation in MSC and macrophages, but failed to induce IL-12 and TNF-alpha production in MSC. Nonetheless, TLR activation in MSC resulted in the formation of an inflammatory site attracting innate immune cells, as evaluated by human neutrophil chemotaxis assays and by the analysis of immune effectors retrieved from Matrigel-embedded MSC injected into mice after in vitro preactivation with cytokines and/or TLR ligands. Hence, TLR-activated MSC are capable of recruiting immune inflammatory cells. In addition, IFN priming combined with TLR activation may increase immune responses induced by Ag-presenting MSC through presentation of Ag in an inflammatory context, a mechanism that could be applied in a cell-based vaccine.


Subject(s)
Cytokines/immunology , Cytokines/metabolism , Stromal Cells/immunology , Stromal Cells/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Adult , Animals , Cells, Cultured , Female , Gene Expression Regulation , Humans , Inflammation/immunology , Inflammation/metabolism , Interferon Regulatory Factor-1/metabolism , Ligands , Ligases/metabolism , Male , Mice , Phenotype , Proto-Oncogene Proteins c-rel/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptor 4/genetics
14.
Exp Hematol ; 37(3): 416-20, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19135773

ABSTRACT

Myeloproliferative disorders (MPDs) are often associated with the presence of the JAK2-V617F mutation in hematopoietic cells. It is currently not known if this mutation is carried as well by bone marrow mesenchymal stromal cells (MSCs) in these patients. To test this hypothesis, we recruited seven patients with JAK2-V617F(+) MPD, isolated marrow MSCs and characterized their phenotype and mesenchymal differentiation capacity, and probed for JAK2-V617F genomic DNA mutation. We found that MSCs of most patients could be culture-expanded and had a phenotype and differentiation capacity similar to that of MSCs derived from normal subjects. Using real-time polymerase chain reaction and melting curve analysis with probes specific for the JAK2-V617F DNA mutation, we did not find the mutation in any of the MSC samples studied. These results demonstrate that, in the setting of MPD, MSC do not originate from the mutated hematopoietic progenitor clone.


Subject(s)
Bone Marrow Cells/pathology , Janus Kinase 2/genetics , Mesenchymal Stem Cells/pathology , Mutation, Missense , Myeloproliferative Disorders/genetics , Cell Lineage , Humans , Myeloproliferative Disorders/pathology , Polymerase Chain Reaction , Tumor Cells, Cultured
15.
Mol Ther ; 17(2): 369-72, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19088705

ABSTRACT

Autologous bone marrow mesenchymal stromal cells (MSCs) have been successfully used for the delivery of erythropoietin (EPO) in murine models of anemia and myocardial infarction. For clinical applications where a transient effect would be adequate, such as myocardial infarction, the use of EPO-engineered universal donor allogeneic MSCs would be a substantial convenience. We thus investigated whether MSCs from C57BL/6 mice would permit robust transient EPO delivery in normal BALB/c allorecipients. Implantation of MSCs overexpressing murine EPO led to increases in hematocrit in syngeneic and allogeneic mice, but the latter eventually developed severe anemia due to acquired neutralizing anti-EPO antibodies. As MSCs constitutively produce the CCL2 chemokine which may behave as an adjuvant to the anti-EPO immune response, experiments were performed using EPO-engineered MSCs derived from CCL2(-/-) mice and similar results were obtained. In conclusion, MHC-mismatched MSCs can break the tolerance to autoantigens and lead to the development of pathogenic autoantibodies.


Subject(s)
Anemia/etiology , Antibodies/immunology , Erythropoietin/immunology , Erythropoietin/metabolism , Mesenchymal Stem Cell Transplantation/methods , Stromal Cells/cytology , Animals , Cell Line , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Stromal Cells/physiology , Transplantation, Homologous
16.
Cancer Res ; 68(12): 4810-8, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18559528

ABSTRACT

Bone marrow-derived mesenchymal stromal cells (MSCs), beneficial for regenerative medicine applications due to their wide differentiation capabilities, also hold promise as cellular vehicles for the delivery of therapeutic plasma-soluble gene products due to their ease of handling, expansion, and genetic engineering. We hypothesized that MSCs, gene enhanced to express interleukin-12 (IL-12) and then embedded in a matrix, may act as an anticancer neo-organoid when delivered s.c. in autologous/syngeneic hosts. We performed such experiments in mice and noted that primary murine MSCs retrovirally engineered to secrete murine IL-12 can significantly interfere with growth of 4T1 breast cancer cells in vivo, with a more substantial anticancer action achieved when these cells are embedded in a matrix. Plasma of mice that received the IL-12 MSC-containing neo-organoids showed increased levels of IL-12 and IFN-gamma. Histopathologic analysis revealed less tumor cells in implants of 4T1 cells with IL-12 MSCs, and the presence of necrotic tumor islets and necrotic capillaries, suggesting antiangiogenesis. We also showed that the anticancer effect exerted by the IL-12 MSCs is immune mediated because it is absent in immunodeficient mice, is not due to systemic IL-12 delivery, and also occurs in a B16 melanoma model. This study therefore establishes the feasibility of using gene-enhanced MSCs in a cell-based neo-organoid approach for cancer treatment.


Subject(s)
Bone Marrow Cells/metabolism , Immunotherapy , Interleukin-12/therapeutic use , Mammary Neoplasms, Experimental/therapy , Mesoderm/cytology , Organoids , Stromal Cells/immunology , Animals , Cells, Cultured , Female , Interleukin-12/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/therapy , Mesoderm/immunology , Mesoderm/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neovascularization, Physiologic , Stromal Cells/metabolism , Stromal Cells/pathology , Survival Rate
17.
J Immunol ; 179(3): 1549-58, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17641021

ABSTRACT

Mesenchymal stromal cells (MSC) possess immunosuppressive properties, yet when treated with IFN-gamma they acquire APC functions. To gain insight into MSC immune plasticity, we explored signaling pathways induced by IFN-gamma required for MHC class II (MHC II)-dependent Ag presentation. IFN-gamma-induced MHC II expression in mouse MSC was enhanced by high cell density or serum deprivation and suppressed by TGF-beta. This process was regulated by the activity of the type IV CIITA promoter independently of STAT1 activation and the induction of the IFN regulatory factor 1-dependent B7H1/PD-L1 encoding gene. The absence of direct correlation with the cell cycle suggested that cellular connectivity modulates IFN-gamma responsiveness for MHC II expression in mouse MSC. TGF-beta signaling in mouse MSC involved ALK5 and ALK1 TGF-beta RI, leading to the phosphorylation of Smad2/Smad3 and Smad1/Smad5/Smad8. An opposite effect was observed in human MSC where IFN-gamma-induced MHC II expression occurred at the highest levels in low-density cultures; however, TGF-beta reduced IFN-gamma-induced MHC II expression and its signaling was similar as in mouse MSC. This suggests that the IFN-gamma-induced APC features of MSC can be modulated by TGF-beta, serum factors, and cell density in vitro, although not in the same way in mouse and human MSC, via their convergent effects on CIITA expression.


Subject(s)
Antigen Presentation/immunology , Cell Communication/immunology , HLA-D Antigens/biosynthesis , Histocompatibility Antigens Class II/biosynthesis , Interferon-gamma/physiology , Mesenchymal Stem Cells/immunology , Transforming Growth Factor beta/physiology , Adult , Animals , Antigen Presentation/genetics , Cell Communication/genetics , Cell Count , Cell Line , Down-Regulation/genetics , Down-Regulation/immunology , Female , HLA-D Antigens/genetics , HLA-D Antigens/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Hybridomas , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Stromal Cells/cytology , Stromal Cells/immunology , Stromal Cells/metabolism , Trans-Activators/biosynthesis , Trans-Activators/genetics , Trans-Activators/metabolism
18.
Mol Cancer Ther ; 6(2): 441-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17308045

ABSTRACT

Proteolytic processing of human plasminogen generates potent antiangiogenic peptides such as angiostatin. The plasminogen kringle 5 (K5) domain, which is distinct from angiostatin, possesses potent antiangiogenic properties on its own, which can be exploited in cancer therapy. It has been recently observed that antiangiogenic agents promote leukocyte-vessel wall interaction as part of their antitumor effect. Although we have previously shown that K5 suppresses cancer growth in tumor xenograft models, its modulation of inflammation in experimental mice with intact immune systems is unknown. To determine whether K5 possesses immune proinflammatory properties, we investigated the effects of K5 in an immune competent model of breast cancer and observed that tumor rejection is substantially reduced in nonobese diabetic/severe combined immunodeficient and BALB/c nude when compared with wild-type BALB/c mice, suggesting an important role for T-lymphoid cells in the antitumor effect of K5. Tumor explant analysis shows that K5 enhances tumor recruitment of CD3(+) lymphoid cells, in particular, the NKT phenotype. We also observed a significant decrease in tumor-associated microvessel length and density consistent with antiangiogenic activity. Histologic analysis of K5 tumors also revealed a robust neutrophilic infiltrate, which may be explained by the neutrophil chemotactic activity of K5 as well as its ability to promote CD64 up-regulation within the CD11b(+) adhesive neutrophil population. In sum, our findings confirm that the K5 protein acts as a potent angiostatic agent and possesses a novel proinflammatory role via its ability to recruit tumor-associated neutrophils and NKT lymphocytes, leading to a potent antitumor response.


Subject(s)
Angiogenesis Inhibitors/metabolism , Mammary Neoplasms, Experimental/therapy , Peptide Fragments/physiology , Plasminogen/physiology , Signal Transduction , Adenocarcinoma/blood supply , Adenocarcinoma/immunology , Adenocarcinoma/therapy , Animals , CD3 Complex/metabolism , Collagen/metabolism , Disease Progression , Drug Combinations , Genes, MHC Class I/physiology , Humans , Immunity, Cellular , Inflammation/metabolism , Laminin/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/prevention & control , Neutrophils/metabolism , Proteoglycans/metabolism , Retroviridae , Survival Rate , Xenograft Model Antitumor Assays
19.
Blood ; 109(5): 2234-42, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17082320

ABSTRACT

We hypothesized that a granulocyte macrophage colony-stimulating factor (GMCSF) and interleukin 15 (IL-15) fusokine (GIFT15) would possess greater immune-stimulatory properties than their combined use. Unexpectedly, tumor cells engineered to secrete GIFT15 protein led to suppression of natural killer (NK) and NKT-cell recruitment in vivo, suggesting an unanticipated immune-suppressive effect. We found GIFT15 to have pleiotropic effects on an array of immune-competent cells. Among these, macrophages treated with GIFT15 secrete de novo the tissue inhibitor of metalloproteinase-2 (TIMP-2); activated matrix metalloproteinase-2 (MMP-2); transforming growth factor-beta (TGF-beta); as well as vascular endothelial growth factor (VEGF). We show that the GIFT15 fusokine has increased affinity for the alpha chain component of the IL-15R, leading to aberrant signaling through the beta chain manifested by the hyperphosphorylation of STAT3 both in macrophages and splenocytes. Suppression of common gamma chain-mediated STAT5 phosphorylation and blockade of the IL-15-dependent IFN-gamma response in mouse splenocytes were also observed. We tested GIFT15 as an immunosuppressor and demonstrated that it allowed engraftment of allogeneic B16F0 and human xenograft U87GM glioma cells in immunocompetent mice. Thus, GIFT15 defines a new class of fusokine that mediates proangiogenic and immunosuppressive effects via aberrant signaling by the IL-15R in lymphomyeloid cells.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Janus Kinases/metabolism , Receptors, Interleukin-15/metabolism , Recombinant Fusion Proteins/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/immunology , Amino Acid Sequence , Animals , Cell Line , Cytokines , Female , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interferon-gamma/metabolism , Interleukin-15/immunology , Interleukin-15/metabolism , Killer Cells, Natural/metabolism , Lymphocyte Activation , Macrophages/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Structure, Quaternary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction/drug effects , Spleen/cytology , Spleen/metabolism , T-Lymphocytes, Cytotoxic/metabolism
20.
J Am Soc Nephrol ; 17(6): 1576-84, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16672321

ABSTRACT

The goal of this research was to develop a strategy to couple stem cell and gene therapy for in vivo delivery of erythropoietin (Epo) for treatment of anemia of ESRD. It was shown previously that autologous bone marrow stromal cells (MSCs) can be genetically engineered to secrete pharmacologic amounts of Epo in normal mice. Therefore, whether anemia in mice with mild to moderate chronic renal failure (CRF) can be improved with Epo gene-modified MSCs (Epo+MSCs) within a subcutaneous implant was examined. A cohort of C57BL/6 mice were rendered anemic by right kidney electrocoagulation and left nephrectomy. In these CRF mice, the hematocrit (Hct) dropped from a prenephrectomy baseline of approximately 55% to 40% after induction of renal failure. MSCs from C57BL/6 donor mice were genetically engineered to secrete murine Epo at a rate of 3 to 4 units of Epo/10(6) cells per 24 h, embedded in a collagen-based matrix, and implanted subcutaneously in anemic CRF mice. It was observed that Hct increased after administration of Epo+MSCs, according to cell dose. Implants of 3 million Epo+MSCs per mouse had no effect on Hct, whereas 10 million led to a supraphysiologic effect. The Hct of CRF mice that received 4.5 or 7.5 million Epo+MSCs rose to a peak 54+/-4.0 or 63+/-5.5%, respectively, at 3 wk after implantation and remained above 48 or 54% for >19 wk. Moreover, mice that had CRF and received Epo+MSCs showed significantly greater swimming exercise capacity. In conclusion, these results demonstrate that subcutaneous implantation of Epo-secreting genetically engineered MSCs can correct anemia that occurs in a murine model of CRF.


Subject(s)
Anemia/therapy , Bone Marrow Cells/cytology , Erythropoietin/administration & dosage , Genetic Engineering/methods , Renal Insufficiency/therapy , Stromal Cells/cytology , Anemia/etiology , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Renal Insufficiency/complications , Renal Insufficiency/pathology , Retroviridae/genetics , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...