Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Cytotherapy ; 26(4): 372-382, 2024 04.
Article in English | MEDLINE | ID: mdl-38363250

ABSTRACT

BACKGROUND AIMS: Human mesenchymal stromal cells (hMSCs) and their secreted products show great promise for treatment of musculoskeletal injury and inflammatory or immune diseases. However, the path to clinical utilization is hampered by donor-tissue variation and the inability to manufacture clinically relevant yields of cells or their products in a cost-effective manner. Previously we described a method to produce chemically and mechanically customizable gelatin methacryloyl (GelMA) microcarriers for culture of hMSCs. Herein, we demonstrate scalable GelMA microcarrier-mediated expansion of induced pluripotent stem cell (iPSC)-derived hMSCs (ihMSCs) in 500 mL and 3L vertical wheel bioreactors, offering several advantages over conventional microcarrier and monolayer-based expansion strategies. METHODS: Human mesenchymal stromal cells derived from induced pluripotent cells were cultured on custom-made spherical gelatin methacryloyl microcarriers in single-use vertical wheel bioreactors (PBS Biotech). Cell-laden microcarriers were visualized using confocal microscopy and elastic light scattering methodologies. Cells were assayed for viability and differentiation potential in vitro by standard methods. Osteogenic cell matrix derived from cells was tested in vitro for osteogenic healing using a rodent calvarial defect assay. Immune modulation was assayed with an in vivo peritonitis model using Zymozan A. RESULTS: The optical properties of GelMA microcarriers permit noninvasive visualization of cells with elastic light scattering modalities, and harvest of product is streamlined by microcarrier digestion. At volumes above 500 mL, the process is significantly more cost-effective than monolayer culture. Osteogenic cell matrix derived from ihMSCs expanded on GelMA microcarriers exhibited enhanced in vivo bone regenerative capacity when compared to bone morphogenic protein 2, and the ihMSCs exhibited superior immunosuppressive properties in vivo when compared to monolayer-generated ihMSCs. CONCLUSIONS: These results indicate that the cell expansion strategy described here represents a superior approach for efficient generation, monitoring and harvest of therapeutic MSCs and their products.


Subject(s)
Cell Culture Techniques , Mesenchymal Stem Cells , Humans , Cell Culture Techniques/methods , Bioreactors , Osteogenesis , Bone Regeneration , Cell Proliferation , Cell Differentiation , Cells, Cultured
2.
Stem Cells ; 40(9): 870-882, 2022 09 26.
Article in English | MEDLINE | ID: mdl-35852488

ABSTRACT

Allogeneic mesenchymal stem/stromal cells (MSCs) are frequently used in clinical trials due to their low expression of major histocompatibility complex (MHC) class I and lack of MHC class II. However, the levels of MHC classes I and II in MSCs are increased by inflammatory stimuli, raising concerns over potential adverse effects associated with allogeneic cell therapy. Also, it is unclear how the host immune response to MHC-mismatched MSCs affects the therapeutic efficacy of the cells. Herein, using strategies to manipulate MHC genes in human bone marrow-derived MSCs via the CRISPR-Cas9 system, plasmids, or siRNAs, we found that inhibition of MHC class I-not MHC class II-in MSCs lowered the survival rate of MSCs and their immunosuppressive potency in mice with experimental autoimmune uveoretinitis, specifically by increasing MSC vulnerability to natural killer (NK)-cell-mediated cytotoxicity. A subsequent survey of MSC batches derived from 6 human donors confirmed a significant correlation between MSC survival rate and susceptibility to NK cells with the potency of MSCs to increase MHC class I level upon stimulation. Our overall results demonstrate that MHC class I enables MSCs to evade NK-cell-mediated cytotoxicity and exert immunosuppressive activity.


Subject(s)
Mesenchymal Stem Cells , Animals , HLA Antigens , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/pharmacology , Humans , Killer Cells, Natural , Mice
3.
Int J Mol Sci ; 24(6)2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36982329

ABSTRACT

Extracellular vesicles (EVs) from allogeneic-tissue-derived mesenchymal stem cells (MSCs) are promising to improve Sjögren's syndrome (SS) treatment, but their application is hindered by high variations in and limited expandability of tissue MSCs. We derived standardized and scalable MSCs from iPS cells (iMSCs) and reported that EVs from young but not aging iMSCs (iEVs) inhibited sialadenitis onset in SS mouse models. Here, we aim to determine cellular mechanisms and optimization approaches of SS-inhibitory effects of iEVs. In NOD.B10.H2b mice at the pre-disease stage of SS, we examined the biodistribution and recipient cells of iEVs with imaging, flow cytometry, and qRT-PCR. Intravenously infused iEVs accumulated in the spleen but not salivary glands or cervical lymph nodes and were mainly taken up by macrophages. In the spleen, young but not aging iEVs increased M2 macrophages, decreased Th17 cells, and changed expression of related immunomodulatory molecules. Loading miR-125b inhibitors into aging iEVs significantly improved their effects on repressing sialadenitis onset and regulating immunomodulatory splenocytes. These data indicated that young but not aging iEVs suppress SS onset by regulating immunomodulatory splenocytes, and inhibiting miR-125b in aging iEVs restores such effects, which is promising to maximize production of effective iEVs from highly expanded iMSCs for future clinical application.


Subject(s)
Extracellular Vesicles , Induced Pluripotent Stem Cells , Mesenchymal Stem Cells , MicroRNAs , Sialadenitis , Sjogren's Syndrome , Mice , Animals , Sjogren's Syndrome/therapy , Sjogren's Syndrome/drug therapy , Spleen/metabolism , Induced Pluripotent Stem Cells/metabolism , Tissue Distribution , Mice, Inbred NOD , Sialadenitis/therapy , Sialadenitis/metabolism , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Disease Models, Animal
4.
Mol Ther ; 28(7): 1628-1644, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32380062

ABSTRACT

Accumulating evidence indicates that mesenchymal stem/stromal cell-derived extracellular vesicles (MSC-EVs) exhibit immunomodulatory effects by delivering therapeutic RNAs and proteins; however, the molecular mechanism underlying the EV-mediated immunomodulation is not fully understood. In this study, we found that EVs from early-passage MSCs had better immunomodulatory potency than did EVs from late-passage MSCs in T cell receptor (TCR)- or Toll-like receptor 4 (TLR4)-stimulated splenocytes and in mice with ocular Sjögren's syndrome. Moreover, MSC-EVs were more effective when produced from 3D culture of the cells than from the conventional 2D culture. Comparative molecular profiling using proteomics and microRNA sequencing revealed the enriched factors in MSC-EVs that were functionally effective in immunomodulation. Among them, manipulation of transforming growth factor ß1 (TGF-ß1), pentraxin 3 (PTX3), let-7b-5p, or miR-21-5p levels in MSCs significantly affected the immunosuppressive effects of their EVs. Furthermore, there was a strong correlation between the expression levels of TGF-ß1, PTX3, let-7b-5p, or miR-21-5p in MSC-EVs and their suppressive function. Therefore, our comparative strategy identified TGF-ß1, PTX3, let-7b-5p, or miR-21-5p as key molecules mediating the therapeutic effects of MSC-EVs in autoimmune disease. These findings would help understand the molecular mechanism underlying EV-mediated immunomodulation and provide functional biomarkers of EVs for the development of robust EV-based therapies.


Subject(s)
C-Reactive Protein/genetics , Extracellular Vesicles/transplantation , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Serum Amyloid P-Component/genetics , Sjogren-Larsson Syndrome/therapy , Transforming Growth Factor beta1/genetics , Animals , C-Reactive Protein/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Gene Expression Profiling , Humans , Mesenchymal Stem Cells/metabolism , Mice , Proteomics , Serial Passage , Serum Amyloid P-Component/metabolism , Sjogren-Larsson Syndrome/genetics , Sjogren-Larsson Syndrome/metabolism , Transforming Growth Factor beta1/metabolism
5.
Mol Ther ; 26(1): 162-172, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29301108

ABSTRACT

The cornea is a transparent tissue devoid of blood and lymphatic vessels. However, various inflammatory conditions can cause hemangiogenesis and lymphangiogenesis in the cornea, compromising transparency and visual acuity. Mesenchymal stem/stromal cells (MSCs) have therapeutic potentials in a variety of diseases because of anti-inflammatory properties. Herein, we investigated the effects of MSCs on corneal angiogenesis using a model of suture-induced inflammatory corneal neovascularization. Data demonstrated that an intravenous administration of MSCs suppressed corneal inflammation and neovascularization, inhibiting both hemangiogenesis and lymphangiogenesis. MSCs reduced the levels of vascular endothelial growth factor (VEGF)-C, VEGF-D, Tek, MRC1, and MRC2 in the cornea, which are expressed by pro-angiogenic macrophages. Moreover, the number of CD11b+ monocytes/macrophages in the cornea, spleen, peripheral blood, and draining lymph nodes was decreased by MSCs. Depletion of circulating CD11b+ monocytes by blocking antibodies replicated the effects of MSCs. Importantly, knockdown of tumor necrosis factor alpha (TNF-α)-stimulated gene/protein 6 (TSG-6) in MSCs abrogated the effects of MSCs in inhibiting corneal hemangiogenesis and lymphangiogenesis and monocyte/macrophage infiltration. Together, the results suggest that MSCs inhibit inflammatory neovascularization in the cornea by suppressing pro-angiogenic monocyte/macrophage recruitment in a TSG-6-dependent manner.


Subject(s)
Cell Adhesion Molecules/metabolism , Cornea/metabolism , Keratitis/immunology , Keratitis/metabolism , Lymphangiogenesis , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Biomarkers , Biopsy , Cell Line , Disease Models, Animal , Female , Flow Cytometry , Humans , Keratitis/pathology , Lymph Nodes , Mice , Monocytes/immunology , Monocytes/metabolism , Transcription, Genetic
6.
Mol Ther ; 25(8): 1748-1756, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28647464

ABSTRACT

Much of what we know about immunology suggests that little is to be gained from experiments in which human cells are administered to immunocompetent mice. Multiple reports have demonstrated that this common assumption does not hold for experiments with human mesenchymal stem/stromal cells (hMSCs). The data demonstrate that hMSCs can suppress immune responses to a variety of stimuli in immunocompetent mice by a range of different mechanisms that are similar to those employed by mouse MSCs. Therefore, further experiments with hMSCs in mice will make it possible to generate preclinical data that will improve both the efficacy and safety of the clinical trials with the cells that are now in progress.


Subject(s)
Immune Tolerance , Immunomodulation , Mesenchymal Stem Cells/metabolism , Animals , Disease Models, Animal , Heterografts , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Immunity , Mesenchymal Stem Cells/cytology , Mice , Species Specificity
7.
Proc Natl Acad Sci U S A ; 112(2): 530-5, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25548183

ABSTRACT

Mesenchymal stem or stromal cells (MSCs) have many potential therapeutic applications including therapies for cancers and tissue damages caused by cancers or radical cancer treatments. However, tissue-derived MSCs such as bone marrow MSCs (BM-MSCs) may promote cancer progression and have considerable donor variations and limited expandability. These issues hinder the potential applications of MSCs, especially those in cancer patients. To circumvent these issues, we derived MSCs from transgene-free human induced pluripotent stem cells (iPSCs) efficiently with a modified protocol that eliminated the need of flow cytometric sorting. Our iPSC-derived MSCs were readily expandable, but still underwent senescence after prolonged culture and did not form teratomas. These iPSC-derived MSCs homed to cancers with efficiencies similar to BM-MSCs but were much less prone than BM-MSCs to promote the epithelial-mesenchymal transition, invasion, stemness, and growth of cancer cells. The observations were probably explained by the much lower expression of receptors for interleukin-1 and TGFß, downstream protumor factors, and hyaluronan and its cofactor TSG6, which all contribute to the protumor effects of BM-MSCs. The data suggest that iPSC-derived MSCs prepared with the modified protocol are a safer and better alternative to BM-MSCs for therapeutic applications in cancer patients. The protocol is scalable and can be used to prepare the large number of cells required for "off-the-shelf" therapies and bioengineering applications.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Animals , Cell Line, Tumor , Cell Movement , Coculture Techniques , Epithelial-Mesenchymal Transition , Female , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness
8.
Proc Natl Acad Sci U S A ; 111(47): 16766-71, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25385603

ABSTRACT

Human mesenchymal stem/progenitor cells (hMSCs) from bone marrow and other tissues are currently being administered to large numbers of patients even though there are no biomarkers that accurately predict their efficacy in vivo. Using a mouse model of chemical injury of the cornea, we found that bone-marrow-derived hMSCs isolated from different donors varied widely in their efficacy in modulating sterile inflammation. Importantly, RT-PCR assays of hMSCs for the inflammation-modulating protein TSG-6 expressed by the TNFα-stimulated gene 6 (TSG-6 or TNFAIP6) predicted their efficacy in sterile inflammation models for corneal injury, sterile peritonitis, and bleomycin-induced lung injury. In contrast, the levels of TSG-6 mRNA were negatively correlated with their potential for osteogenic differentiation in vitro and poorly correlated with other criteria for evaluating hMSCs. Also, a survey of a small cohort suggested that hMSCs from female donors compared with male donors more effectively suppressed sterile inflammation, expressed higher levels of TSG-6, and had slightly less osteogenic potential.


Subject(s)
Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Inflammation/pathology , Mesenchymal Stem Cells/cytology , Animals , Cell Adhesion Molecules/genetics , Female , Humans , Inflammation/metabolism , Male , Mice , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction
9.
Cytotherapy ; 17(10): 1332-41, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26227206

ABSTRACT

BACKGROUND AIMS: Previously, we showed that human mesenchymal stromal cells (hMSCs) were activated to express tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) upon TNF-α stimulation, induced cell death in triple-negative breast cancer (TNBC) MDA-MB-231 cells (MDA), and RNA released from apoptotic MDA further increased TRAIL expression in hMSCs. This feed-forward stimulation increased apoptosis in MDA cells. Here, we tested whether TRAIL-expressing hMSCs, in combination with a sub-toxic-dose of a chemotherapy drug doxorubicin, would overcome TRAIL resistance and create synergistic effects on targeting metastatic TNBC. METHODS: To optimize conditions for the combination treatment, we (i) selected an optimal condition to activate hMSCs for TRAIL expression, (ii) selected an optimal dose of doxorubicin treatment, (iii) examined underlying mechanisms in vitro and (iv) tested the efficacy of the optimized conditions in a xenograft mouse model of human breast cancer lung metastasis. RESULTS: The results showed that DNA fragments from apoptotic MDA triggered hMSCs to increase further TRAIL expression in an absent in melanoma 2 (AIM2)-dependent manner, and thus higher TRAIL-expressing hMSCs stimulated with synthetic DNA, poly(deoxyadenylic-deoxythymidylic) acid [poly(dA:dT)], more effectively suppressed tumor progression in vivo. Furthermore, activated hMSCs increased apoptosis in MDA cells when combined with a sub-toxic dose of doxorubicin, which was mediated by up-regulating TRAIL and Fas-related pathways. When we combined the optimized conditions, pre-activated hMSCs with poly (dA:dT) synergistically reduced tumor burden even with minimal doxorubicin treatment in a xenograft mouse model of human breast cancer lung metastasis. CONCLUSIONS: These results suggest that the treatment of hMSCs with a sub-toxic dose of doxorubicin can overcome TRAIL resistance and be a potential novel therapy for TNBC metastasis treatment.


Subject(s)
Apoptosis , Doxorubicin/therapeutic use , Mesenchymal Stem Cell Transplantation , TNF-Related Apoptosis-Inducing Ligand/metabolism , Triple Negative Breast Neoplasms/therapy , Animals , Cell Line, Tumor , DEAD-box RNA Helicases/genetics , DNA Fragmentation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Humans , Interferon-Induced Helicase, IFIH1 , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mesenchymal Stem Cells/metabolism , Mice , Poly dA-dT/pharmacology , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays
10.
Transplantation ; 108(6): 1368-1375, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38409732

ABSTRACT

BACKGROUND: Corneal transplantation is the most common transplant procedure worldwide. Despite immune and angiogenic privilege of the cornea, 50% to 70% of corneal transplants fail in high-risk recipients, primarily because of immune rejection. Therefore, it is crucial to identify predictive biomarkers of rejection to improve transplant survival. METHODS: In search for predictive biomarkers, we performed proteomics analysis of serum extracellular vesicles (EVs) in a fully major histocompatibility complex-mismatched (C57BL/6-to-BALB/c) murine corneal transplantation model, wherein 50% of transplants undergo rejection by day 28 following transplantation. RESULTS: Our time course study revealed a decrease in the number of serum EVs on day 1, followed by a gradual increase by day 7. A comparative analysis of proteomics profiles of EVs from transplant recipients with rejection (rejectors) and without rejection (nonrejectors) found a distinct enrichment of histocompatibility 2, Q region locus 2, which is a part of major histocompatibility complex-class I of donor C57BL/6 mice, in day 7 EVs of rejectors, compared with nonrejectors, syngeneic controls, or naïve mice. In contrast, serum amyloid A2, a protein induced in response to injury, was increased in day 7 EVs of nonrejectors. CONCLUSIONS: Our findings offer noninvasive EV-based potential biomarkers for predicting corneal allograft rejection or tolerance.


Subject(s)
Biomarkers , Corneal Transplantation , Extracellular Vesicles , Graft Rejection , Mice, Inbred BALB C , Mice, Inbred C57BL , Proteomics , Graft Rejection/blood , Graft Rejection/immunology , Graft Rejection/diagnosis , Animals , Extracellular Vesicles/metabolism , Biomarkers/blood , Proteomics/methods , Mice , Graft Survival , Disease Models, Animal , Predictive Value of Tests , Male
11.
Nat Commun ; 15(1): 3283, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38637507

ABSTRACT

While poly(ethylene glycol) (PEG) hydrogels are generally regarded as biologically inert blank slates, concerns over PEG immunogenicity are growing, and the implications for tissue engineering are unknown. Here, we investigate these implications by immunizing mice against PEG to stimulate anti-PEG antibody production and evaluating bone defect regeneration after treatment with bone morphogenetic protein-2-loaded PEG hydrogels. Quantitative analysis reveals that PEG sensitization increases bone formation compared to naive controls, whereas histological analysis shows that PEG sensitization induces an abnormally porous bone morphology at the defect site, particularly in males. Furthermore, immune cell recruitment is higher in PEG-sensitized mice administered the PEG-based treatment than their naive counterparts. Interestingly, naive controls that were administered a PEG-based treatment also develop anti-PEG antibodies. Sex differences in bone formation and immune cell recruitment are also apparent. Overall, these findings indicate that anti-PEG immune responses can impact tissue engineering efficacy and highlight the need for further investigation.


Subject(s)
Biocompatible Materials , Tissue Engineering , Female , Male , Mice , Animals , Biocompatible Materials/pharmacology , Osteogenesis , Bone Regeneration , Polyethylene Glycols/pharmacology , Hydrogels/pharmacology
12.
bioRxiv ; 2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38645051

ABSTRACT

Dysfunction of the retinal pigment epithelium (RPE) is a common shared pathology in major degenerative retinal diseases despite variations in the primary etiologies of each disease. Due to their demanding and indispensable functional roles throughout the lifetime, RPE cells are vulnerable to genetic predisposition, external stress, and aging processes. Building upon recent advancements in stem cell technology for differentiating healthy RPE cells and recognizing the significant roles of small extracellular vesicles (sEV) in cellular paracrine and autocrine actions, we investigated the hypothesis that the RPE-secreted sEV alone can restore essential RPE functions and rescue photoreceptors in RPE dysfunction-driven retinal degeneration. Our findings support the rationale for developing intravitreal treatment of sEV. We demonstrate that intravitreally delivered sEV effectively penetrate the full thickness of the retina. Xenogenic intraocular administration of human-derived EVs did not induce acute immune reactions in rodents. sEV derived from human embryonic stem cell (hESC)-derived fully differentiated RPE cells, but not sEV-depleted conditioned cell culture media (CCM minus sEV), rescued photoreceptors and their function in a Royal College of Surgeons (RCS) rat model. This model is characterized by photoreceptor death and retinal degeneration resulting from a mutation in the MerTK gene in RPE cells. From the bulk RNA sequencing study, we identified 447 differently expressed genes in the retina after hESC-RPE-sEV treatment compared with the untreated control. Furthermore, 394 out of 447 genes (88%) showed a reversal in expression toward the healthy state in Long-Evans (LE) rats after treatment compared to the diseased state. Particularly, detrimental alterations in gene expression in RCS rats, including essential RPE functions such as phototransduction, vitamin A metabolism, and lipid metabolism were partially reversed. Defective photoreceptor outer segment engulfment due to intrinsic MerTK mutation was partially ameliorated. These findings suggest that RPE-secreted sEV may play a functional role similar to that of RPE cells. Our study justifies further exploration to fully unlock future therapeutic interventions with sEV in a broad array of degenerative retinal diseases.

13.
Blood ; 118(2): 330-8, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21551236

ABSTRACT

Human mesenchymal stem/progenitor cells (hMSCs) repair tissues and modulate immune systems but the mechanisms are not fully understood. We demonstrated that hMSCs are activated by inflammatory signals to secrete the anti-inflammatory protein, TNF-α-stimulated gene 6 protein (TSG-6) and thereby create a negative feedback loop that reduces inflammation in zymosan-induced peritonitis. The results demonstrate for the first time that TSG-6 interacts through the CD44 receptor on resident macrophages to decrease zymosan/TLR2-mediated nuclear translocation of the NF-κB. The negative feedback loop created by MSCs through TSG-6 attenuates the inflammatory cascade that is initiated by resident macrophages and then amplified by mesothelial cells and probably other cells of the peritoneum. Because inflammation underlies many pathologic processes, including immune responses, the results may explain the beneficial effects of MSCs and TSG-6 in several disease models.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/pharmacology , Macrophages, Peritoneal/drug effects , Mesenchymal Stem Cells/metabolism , NF-kappa B/metabolism , Peritonitis/prevention & control , Toll-Like Receptor 2/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/therapeutic use , Cells, Cultured , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Humans , Macrophages, Peritoneal/metabolism , Male , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peritonitis/chemically induced , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Zymosan
14.
Mol Ther ; 20(11): 2143-52, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22929658

ABSTRACT

Mesenchymal stem/progenitor cells (MSCs) were reported to enhance the survival of cellular and organ transplants. However, their mode of action was not established. We here used a mouse model of corneal allotransplantation and demonstrated that peri-transplant intravenous (i.v.) infusion of human MSCs (hMSCs) decreased the early surgically induced inflammation and reduced the activation of antigen-presenting cells (APCs) in the cornea and draining lymph nodes (DLNs). Subsequently, immune rejection was decreased, and allograft survival was prolonged. Quantitative assays for human GAPDH revealed that <10 hMSCs out of 1 × 10(6) injected cells were recovered in the cornea 10 hours to 28 days after i.v. infusion. Most of hMSCs were trapped in lungs where they were activated to increase expression of the gene for a multifunctional anti-inflammatory protein tumor necrosis factor-α stimulated gene/protein 6 (TSG-6). i.v. hMSCs with a knockdown of TSG-6 did not suppress the early inflammation and failed to prolong the allograft survival. Also, i.v. infusion of recombinant TSG-6 reproduced the effects of hMSCs. Results suggest that hMSCs improve the survival of corneal allografts without engraftment and primarily by secreting TSG-6 that acts by aborting early inflammatory responses. The same mechanism may explain previous reports that MSCs decrease rejection of other organ transplants.


Subject(s)
Corneal Transplantation/methods , Graft Rejection/prevention & control , Mesenchymal Stem Cell Transplantation , Administration, Intravenous , Animals , Anti-Inflammatory Agents/administration & dosage , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/physiology , Cell Adhesion Molecules/administration & dosage , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Cornea/immunology , Cornea/metabolism , Cornea/pathology , Female , Gene Knockdown Techniques , Graft Rejection/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Inflammation Mediators/metabolism , Lung/immunology , Lung/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Macrophages/immunology , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Transcriptome , Transplantation, Homologous
15.
Mol Ther ; 20(4): 788-97, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22294148

ABSTRACT

Oxidative stress and photoreceptor apoptosis are prominent features of many forms of retinal degeneration (RD) for which there are currently no effective therapies. We previously observed that mesenchymal stem/stromal cells reduce apoptosis by being activated to secrete stanniocalcin-1 (STC-1), a multifunctional protein that reduces oxidative stress by upregulating mitochondrial uncoupling protein-2 (UCP-2). Therefore, we tested the hypothesis that intravitreal injection of STC-1 can rescue photoreceptors. We first tested STC-1 in the rhodopsin transgenic rat characterized by rapid photoreceptor loss. Intravitreal STC-1 decreased the loss of photoreceptor nuclei and transcripts and resulted in measurable retinal function when none is otherwise present in this rapid degeneration. We then tested STC-1 in the Royal College of Surgeons (RCS) rat characterized by a slower photoreceptor degeneration. Intravitreal STC-1 reduced the number of pyknotic nuclei in photoreceptors, delayed the loss of photoreceptor transcripts, and improved function of rod photoreceptors. Additionally, STC-1 upregulated UCP-2 and decreased levels of two protein adducts generated by reactive oxygen species (ROS). Microarrays from the two models demonstrated that STC-1 upregulated expression of a similar profile of genes for retinal development and function. The results suggested that intravitreal STC-1 is a promising therapy for various forms of RD including retinitis pigmentosa and atrophic age-related macular degeneration (AMD).


Subject(s)
Glycoproteins/pharmacology , Retinal Degeneration/drug therapy , Animals , Electroretinography , Enzyme-Linked Immunosorbent Assay , Humans , Ion Channels/genetics , Ion Channels/metabolism , Macular Degeneration/drug therapy , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Rats , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Retinal Degeneration/metabolism , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/drug therapy , Uncoupling Protein 2
16.
Proc Natl Acad Sci U S A ; 107(31): 13724-9, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20643923

ABSTRACT

Previous reports suggested that culture as 3D aggregates or as spheroids can increase the therapeutic potential of the adult stem/progenitor cells referred to as mesenchymal stem cells or multipotent mesenchymal stromal cells (MSCs). Here we used a hanging drop protocol to prepare human MSCs (hMSCs) as spheroids that maximally expressed TNFalpha stimulated gene/protein 6 (TSG-6), the antiinflammatory protein that was expressed at high levels by hMSCs trapped in the lung after i.v. infusion and that largely explained the beneficial effects of hMSCs in mice with myocardial infarcts. The properties of spheroid hMSCs were found to depend critically on the culture conditions. Under optimal conditions for expression of TSG-6, the hMSCs also expressed high levels of stanniocalcin-1, a protein with both antiinflammatory and antiapoptotic properties. In addition, they expressed high levels of three anticancer proteins: IL-24, TNFalpha-related apoptosis inducing ligand, and CD82. The spheroid hMSCs were more effective than hMSCs from adherent monolayer cultures in suppressing inflammatory responses in a coculture system with LPS-activated macrophages and in a mouse model for peritonitis. In addition, the spheroid hMSCs were about one-fourth the volume of hMSCs from adherent cultures. Apparently as a result, larger numbers of the cells trafficked through the lung after i.v. infusion and were recovered in spleen, liver, kidney, and heart. The data suggest that spheroid hMSCs may be more effective than hMSCs from adherent cultures in therapies for diseases characterized by sterile tissue injury and unresolved inflammation and for some cancers that are sensitive to antiinflammatory agents.


Subject(s)
Mesenchymal Stem Cells/cytology , Animals , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Aggregation , Cell Survival , Cells, Cultured , Glycoproteins/metabolism , Humans , Kangai-1 Protein/immunology , Ligands , Macrophages/immunology , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Oligonucleotide Array Sequence Analysis , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology
17.
Proc Natl Acad Sci U S A ; 107(39): 16875-80, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20837529

ABSTRACT

Previous reports demonstrated that adult stem/progenitor cells from bone marrow (multipotent mesenchymal stem cells; MSCs) can repair injured tissues with little evidence of engraftment or differentiation. In exploring this phenomenon, our group has recently discovered that the therapeutic benefits of MSCs are in part explained by the cells being activated by signals from injured tissues to express an anti-inflammatory protein TNF-α-stimulated gene/protein 6 (TSG-6). Therefore, we elected to test the hypothesis that TSG-6 would have therapeutic effects in inflammatory but noninfectious diseases of the corneal surface. We produced a chemical and mechanical injury of the cornea in rats by brief application of 100% ethanol followed by mechanical debridement of corneal and limbal epithelium. Recombinant human TSG-6 or PBS solution was then injected into the anterior chamber of the eye. TSG-6 markedly decreased corneal opacity, neovascularization, and neutrophil infiltration. The levels of proinflammatory cytokines, chemokines, and matrix metalloproteinases were also decreased. The data indicated that TSG-6, a therapeutic protein produced by MSCs in response to injury signals, can protect the corneal surface from the excessive inflammatory response following injury.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cell Adhesion Molecules/therapeutic use , Cornea/drug effects , Corneal Neovascularization/drug therapy , Keratitis/drug therapy , Animals , Anterior Chamber/drug effects , Anterior Chamber/pathology , Cornea/pathology , Corneal Injuries , Corneal Neovascularization/pathology , Keratitis/chemically induced , Keratitis/pathology , Male , Rats , Rats, Inbred Lew
18.
Sci Adv ; 9(45): eadi2387, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37948519

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) have been evaluated in >1500 clinical trials, but outcomes remain suboptimal because of knowledge gaps in quality attributes that confer potency. We show that TWIST1 directly represses TSG6 expression that TWIST1 and TSG6 are inversely correlated across bone marrow-derived MSC (BM-MSC) donor cohorts and predict interdonor differences in their proangiogenic, anti-inflammatory, and immune suppressive activity in vitro and in sterile inflammation and autoimmune type 1 diabetes preclinical models. Transcript profiling of TWIST1HiTSG6Low versus TWISTLowTSG6Hi BM-MSCs revealed previously unidentified roles for TWIST1/TSG6 in regulating cellular oxidative stress and TGF-ß2 in modulating TSG6 expression and anti-inflammatory activity. TWIST1 and TSG6 levels also correlate to donor stature and predict differences in iPSC-derived MSC quality attributes. These results validate TWIST1 and TSG6 as biomarkers that predict interdonor differences in potency across laboratories and assay platforms, thereby providing a means to manufacture MSC products tailored to specific diseases.


Subject(s)
Mesenchymal Stem Cells , Humans , Anti-Inflammatory Agents/pharmacology , Biomarkers/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation , Immunologic Factors/metabolism , Mesenchymal Stem Cells/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
19.
Stem Cells ; 29(10): 1572-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21837654

ABSTRACT

Previous reports demonstrated that the deleterious effects of chemical injury to the cornea were ameliorated by local or systemic administration of adult stem/progenitor cells from bone marrow referred to as mesenchymal stem or stromal cells (MSCs). However, the mechanisms for the beneficial effects of MSCs on the injured cornea were not clarified. Herein, we demonstrated that human MSCs (hMSCs) were effective in reducing corneal opacity and inflammation without engraftment after either intraperitoneal (i.p.) or intravenous (i.v.) administration following chemical injury to the rat cornea. A quantitative assay for human mRNA for glyceraldehyde 3-phosphate dehydrogenase (GAPDH) demonstrated that less than 10 hMSCs were present in the corneas of rats 1-day and 3 days after i.v. or i.p. administration of 1 × 10(7) hMSCs. In vitro experiments using a transwell coculture system demonstrated that chemical injury to corneal epithelial cells activated hMSCs to secrete the multipotent anti-inflammatory protein TNF-α stimulated gene/protein 6 (TSG-6). In vivo, the effects of i.v. injection of hMSCs were largely abrogated by knockdown of TSG-6. Also, the effects of hMSCs were essentially duplicated by either i.v. or topical administration of TSG-6. Therefore, the results demonstrated that systemically administered hMSCs reduce inflammatory damage to the cornea without engraftment and primarily by secretion of the anti-inflammatory protein TSG-6 in response to injury signals from the cornea.


Subject(s)
Cell Adhesion Molecules/metabolism , Cornea/immunology , Corneal Opacity/therapy , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Mesenchymal Stem Cells/immunology , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Coculture Techniques , Corneal Injuries , Enzyme-Linked Immunosorbent Assay , Epithelium, Corneal/immunology , Epithelium, Corneal/injuries , Gene Knockdown Techniques , Humans , Injections, Intraperitoneal , Injections, Intravenous , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred BALB C , Models, Animal , RNA, Small Interfering , Rats , Rats, Inbred Lew , Transfection
20.
J Cell Biochem ; 112(11): 3073-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21748781

ABSTRACT

Systemic administration of MSCs resulted in remarkable functional improvements in injured tissues without either long-term engraftment or differentiation in many clinical and experimental situations. Emerging evidence suggest that most of the beneficial effects of MSCs could be explained by secretion of soluble factors that have multiple effects including modulation of inflammatory and immune reactions, protection from cell death, and stimulation of endogenous progenitor cells. In this review, we focus on the therapeutic factors that account for the beneficial effects of MSCs in animal models of human diseases.


Subject(s)
Mesenchymal Stem Cells/metabolism , Wound Healing , Apoptosis , Humans , Inflammation/prevention & control , Regeneration
SELECTION OF CITATIONS
SEARCH DETAIL