Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 230
Filter
1.
J Immunol ; 213(3): 257-267, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38856632

ABSTRACT

Psoriasis is a common inflammatory skin disorder with no cure. Mesenchymal stem cells (MSCs) have immunomodulatory properties for psoriasis, but the therapeutic efficacies varied, and the molecular mechanisms were unknown. In this study, we improved the efficacy by enhancing the immunomodulatory effects of umbilical cord-derived MSCs (UC-MSCs). UC-MSCs stimulated by TNF-α and IFN-γ exhibited a better therapeutic effect in a mouse model of psoriasis. Single-cell RNA sequencing revealed that the stimulated UC-MSCs overrepresented a subpopulation expressing high tryptophanyl-tRNA synthetase 1 (WARS1). WARS1-overexpressed UC-MSCs treat psoriasis-like skin inflammation more efficiently than control UC-MSCs by restraining the proinflammatory macrophages. Mechanistically, WARS1 maintained a RhoA-Akt axis and governed the immunomodulatory properties of UC-MSCs. Together, we identify WARS1 as a master regulator of UC-MSCs with enhanced immunomodulatory capacities, which paves the way for the directed modification of UC-MSCs for escalated therapeutic efficacy.


Subject(s)
Immunomodulation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Mesenchymal Stem Cells/immunology , Animals , Mice , Humans , Mesenchymal Stem Cell Transplantation/methods , Tryptophan-tRNA Ligase/genetics , Psoriasis/immunology , Psoriasis/therapy , Disease Models, Animal , Single-Cell Analysis , Sequence Analysis, RNA , Umbilical Cord/cytology , Umbilical Cord/immunology , Mice, Inbred C57BL , Cells, Cultured
2.
Med Sci Monit ; 28: e934660, 2022 Feb 14.
Article in English | MEDLINE | ID: mdl-35153292

ABSTRACT

BACKGROUND Fundamental and clinical interest in mesenchymal stem cells (MSCs) has risen dramatically over the past 3 decades. The immunomodulatory and differentiation abilities are the main mechanisms in vitro and in vivo. However, increasing evidence casts doubt on the stemness and immunogenicity of MSCs. MATERIAL AND METHODS We conducted a high-throughput 10x RNA sequencing and Smart-seq2 scRNA-seq analysis to reveal gene expression of Wharton jelly MSCs (WJ-MSCs) at a single-cell level. Multipotent differentiation, subpopulations, marker genes, human leucocyte antigen (HLA) gene expression, and cell cluster trajectory analysis were evaluated. RESULTS The WJ-MSCs had considerable heterogeneity between cells in terms of gene expression. They highly, partially, and hardly expressed genes related to mesodermal differentiation, endodermal differentiation, and ectodermal differentiation, respectively. Some cells seem to be bipotent or unipotent stem cells. Further, Monocle and cell cluster trajectory analysis demonstrated that 1 of the 3 divided clusters performed as stem cells, accounting for 12.6% of the population. The marker genes for a stem cell cluster were CRIM1, GLS, PLOD2, NEXN, ACTR2, FN1, MBNL1, LMOD1, COL3A1, NCL, SEC62, EPRS, COL5A2, COL8A1, and VCAN. In addition, the MSCs also highly, partially, and hardly expressed HLA-I antigen genes, HLA-II genes, and the HLA-G gene, respectively, indicating that MSCs probably have immunogenicity. A Kyoto Encyclopedia of Genes and Genomes pathway analysis of the 3 clusters demonstrated that they were mainly connected with viral infectious diseases, cancer, and endocrine and metabolic disorders. The most expressed transcription factors were zf-C2H2, HMG/HMGY, and Homeobox. CONCLUSIONS We found that only a subpopulation of WJ-MSCs are real stem cells and WJ-MSCs probably do not have immune privilege.


Subject(s)
Immune Privilege , Mesenchymal Stem Cells/cytology , RNA/genetics , Umbilical Cord/cytology , Wharton Jelly/cytology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Mesenchymal Stem Cells/immunology , Sequence Analysis, RNA , Transcription Factors , Umbilical Cord/immunology , Wharton Jelly/immunology , Wharton Jelly/metabolism
3.
Acta Haematol ; 145(1): 63-71, 2022.
Article in English | MEDLINE | ID: mdl-34284381

ABSTRACT

INTRODUCTION: Autoimmune hemolytic anemia is a potentially lethal disease characterized by autoimmune hemolysis. Although human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) have been reported as a promising therapy, there is limited evidence regarding warm autoimmune hemolytic anemia (wAIHA) patients. This study aimed to investigate the potential therapeutic effects of hUC-MSCs via immune regulation in wAIHA patients. METHODS: Peripheral blood mononuclear cells (PBMCs) from 10 wAIHA patients and 8 healthy controls were isolated from peripheral blood and cultured for 3 days with or without the presence of hUC-MSCs; PBMCs were co-cultured with hUC-MSCs using Transwell assays. The supernatant cytokine levels were measured after culture through AimPlex Multiple Immunoassays for Flow, including IL-2, IL-4, IL-10, IFN-γ, TNF-α, and IL-17A. The percentages of regulatory T cells, regulatory B cells, and Th1/Th2 in PBMCs were also assessed before and after culturing. RESULTS: In the wAIHA group, hUC-MSCs could upregulate the Treg and Breg proportions after culturing for 3 days, and the Treg and Breg percentages increased after co-culturing with hUC-MSCs in the wAIHA group compared with PBMC cultured alone for 3 days (8.29 ± 8.59 vs. 6.82 ± 1.32, 3.82 ± 1.87 vs. 1.75 ± 1.20, respectively). Compared with the PBMC wAIHA group, the levels of TNF-α (2.13 ± 2.07 vs. 16.20 ± 21.13 pg/mL, p = 0.019) and IL-10 (10.51 ± 18.42 vs. 37.78 ± 44.20 pg/mL, p = 0.012) were significantly elevated in the PBMC + hUC-MSCs wAIHA group. CONCLUSION: The hUC-MSCs contributed to the increasing proportion of regulatory cell populations in PBMCs of wAIHA patients, thereby potentially regulating autoimmune response; thus, hUC-MSCs may be a promising approach for wAIHA treatment.


Subject(s)
Anemia, Hemolytic, Autoimmune/immunology , Cytokines/immunology , Immunomodulation , Lymphocytes/immunology , Mesenchymal Stem Cells/immunology , Umbilical Cord/immunology , Adult , Aged , Aged, 80 and over , Anemia, Hemolytic, Autoimmune/pathology , Coculture Techniques , Female , Humans , Lymphocytes/pathology , Male , Mesenchymal Stem Cells/pathology , Middle Aged , Umbilical Cord/pathology
4.
Biomolecules ; 11(8)2021 08 02.
Article in English | MEDLINE | ID: mdl-34439807

ABSTRACT

Regenerative medicine is a dynamically developing field of human and veterinary medicine. The animal model was most commonly used for mesenchymal stem cells (MSCs) treatment in experimental and preclinical studies with a satisfactory therapeutic effect. Year by year, the need for alternative treatments in veterinary medicine is increasing, and other applications for promising MSCs and their biological derivatives are constantly being sought. There is also an increase in demand for other methods of treating disease states, of which the classical treatment methods did not bring the desired results. Cell therapy can be a realistic option for treating human and animal diseases in the near future and therefore additional research is needed to optimize cell origins, numbers, or application methods in order to standardize the treatment process and assess its effects. The aim of the following work was to summarize available knowledge about stem cells in veterinary medicine and their possible application in the treatment of chosen musculoskeletal disorders in dogs and horses.


Subject(s)
Cell- and Tissue-Based Therapy/veterinary , Mesenchymal Stem Cell Transplantation/veterinary , Mesenchymal Stem Cells/immunology , Musculoskeletal Diseases/therapy , Musculoskeletal Diseases/veterinary , Veterinary Medicine/methods , Adipose Tissue/cytology , Adipose Tissue/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell- and Tissue-Based Therapy/methods , Dogs , Female , Horses , Humans , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Musculoskeletal Diseases/immunology , Musculoskeletal Diseases/pathology , Placenta/cytology , Placenta/immunology , Pregnancy , Regenerative Medicine/methods , Transplantation, Autologous , Transplantation, Homologous , Umbilical Cord/cytology , Umbilical Cord/immunology
5.
Neuroimmunomodulation ; 28(1): 1-21, 2021.
Article in English | MEDLINE | ID: mdl-33910207

ABSTRACT

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) has devastating effects on the population worldwide. Given this scenario, the extent of the impact of the disease on more vulnerable individuals, such as pregnant women, is of great concern. Although pregnancy may be a risk factor in respiratory virus infections, there are no considerable differences regarding COVID-19 severity observed between pregnant and nonpregnant women. In these circumstances, an emergent concern is the possibility of neurodevelopmental and neuropsychiatric harm for the offspring of infected mothers. Currently, there is no stronger evidence indicating vertical transmission of SARS-CoV-2; however, the exacerbated inflammatory response observed in the disease could lead to several impairments in the offspring's brain. Furthermore, in the face of historical knowledge on possible long-term consequences for the progeny's brain after infection by viruses, we must consider that this might be another deleterious facet of COVID-19. In light of neuroimmune interactions at the maternal-fetal interface, we review here the possible harmful outcomes to the offspring brains of mothers infected by SARS-CoV-2.


Subject(s)
COVID-19/immunology , Neurodevelopmental Disorders/physiopathology , Neuroimmunomodulation/immunology , Pregnancy Complications, Infectious/immunology , Prenatal Exposure Delayed Effects/physiopathology , COVID-19/metabolism , COVID-19/physiopathology , Cytokine Release Syndrome/immunology , Decidua/immunology , Female , Humans , Immune Tolerance/immunology , Infectious Disease Transmission, Vertical , Neuroimmunomodulation/physiology , Placenta/immunology , Pregnancy , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/physiopathology , SARS-CoV-2 , Umbilical Cord/immunology
6.
Front Immunol ; 11: 576261, 2020.
Article in English | MEDLINE | ID: mdl-33329542

ABSTRACT

T cell-mediated immune tolerance is a state of unresponsiveness of T cells towards specific self or non-self antigens. This is particularly essential during prenatal/neonatal period when T cells are exposed to dramatically changing environment and required to avoid rejection of maternal antigens, limit autoimmune responses, tolerate inert environmental and food antigens and antigens from non-harmful commensal microorganisms, promote maturation of mucosal barrier function, yet mount an appropriate response to pathogenic microorganisms. The cell-intrinsic and cell extrinsic mechanisms promote the generation of prenatal/neonatal T cells with distinct features to meet the complex and dynamic need of tolerance during this period. Reduced exposure or impaired tolerance in early life may have significant impact on allergic or autoimmune diseases in adult life. The uniqueness of conventional and regulatory T cells in human umbilical cord blood (UCB) may also provide certain advantages in UCB transplantation for hematological disorders.


Subject(s)
Immune Tolerance , T-Lymphocytes/immunology , Thymus Gland/immunology , Age Factors , Animals , Cord Blood Stem Cell Transplantation/adverse effects , Humans , Infant , Infant, Newborn , Phenotype , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Self Tolerance , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Transplantation, Homologous , Umbilical Cord/immunology
7.
Sci Rep ; 10(1): 18978, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33149204

ABSTRACT

Differentiation of mesenchymal stem cells (MSCs) derived from two different sources of fetal tissues such as umbilical cord blood (UCB) and tissue (UCT) into skeletal muscle have remained underexplored. Here, we present a comparative analysis of UCB and UCT MSCs, in terms of surface markers, proliferation and senescence marker expression. We find that CD45-CD34- MSCs obtained from UCT and UCB of term births display differences in the combinatorial expression of key MSC markers CD105 and CD90. Importantly, UCT MSCs display greater yield, higher purity, shorter culture time, and lower rates of senescence in culture compared to UCB MSCs. Using a robust myogenic differentiation protocol, we show that UCT MSCs differentiate more robustly into muscle than UCB MSCs by transcriptomic sequencing and specific myogenic markers. Functional assays reveal that CD90, and not CD105 expression promotes myogenic differentiation in MSCs and could explain the enhanced myogenic potential of UCT MSCs. These results suggest that in comparison to large volumes of UCB that are routinely used to obtain MSCs and with limited success, UCT is a more reliable, robust, and convenient source of MSCs to derive cells of the myogenic lineage for both therapeutic purposes and increasing our understanding of developmental processes.


Subject(s)
Fetal Blood/cytology , Gene Expression Profiling/methods , Mesenchymal Stem Cells/cytology , Muscle, Skeletal/cytology , Umbilical Cord/cytology , Antigens, CD34/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cells, Cultured , Endoglin/metabolism , Female , Fetal Blood/immunology , Gene Expression Regulation , Genetic Markers , Humans , Leukocyte Common Antigens/metabolism , Mesenchymal Stem Cells/immunology , Muscle Development , Muscle, Skeletal/chemistry , Pregnancy , Sequence Analysis, RNA , Term Birth , Thy-1 Antigens/metabolism , Umbilical Cord/immunology
8.
Int J Mol Sci ; 21(15)2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32731615

ABSTRACT

Mesenchymal stem cells (MSCs) have become a promising tool in cellular therapy for restoring immune system haemostasis; however, the success of clinical trials has been impaired by the lack of standardized manufacturing processes. This study aims to determine the suitability of source tissues and culture media for the production of MSC-based advanced therapy medicinal products (ATMPs) and to define parameters to extend the set of release criteria. MSCs were isolated from umbilical cord (UC), bone marrow and lipoaspirate and expanded in three different culture media. MSC phenotype, proliferation capacity and immunosuppressive parameters were evaluated in normal MSCs compared to primed MSCs treated with cytokines mimicking an inflammatory environment. Compared to bone marrow and lipoaspirate, UC-derived MSCs (UC-MSCs) showed the highest proliferative capacity, which was further enhanced by media supplemented with bFGF, while the cells maintained their immunosuppressive characteristics. Moreover, UC-MSCs expanded in the bFGF-enriched medium were the least sensitive to undesirable priming-induced changes in the MSC phenotype. Surface markers and secreted factors were identified to reflect the cell response to inflammatory priming and to be variable among MSCs from different source tissues. This study demonstrates that UC is a favorable cell source for manufacturing MSC-based ATMPs for immunosuppressive applications. UC-MSCs are able to use the bFGF-enriched medium for higher cell yields without the impairment of immunosuppressive parameters and undesirable phenotype changes after inflammatory preconditioning of MSCs before transplantation. Additionally, immunosuppressive parameters were identified to help finding predictors of clinically efficient MSCs in the following clinical trials.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Fibroblast Growth Factor 2/pharmacology , Immunosuppression Therapy , Mesenchymal Stem Cells/immunology , Umbilical Cord/immunology , Cell Differentiation/immunology , Fibroblast Growth Factor 2/immunology , Humans , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology
9.
Allergol. immunopatol ; 48(3): 295-305, mayo-jun. 2020. tab, graf
Article in English | IBECS | ID: ibc-192033

ABSTRACT

INTRODUCTION: In newborns, dramatic changes occur in the blood and bone marrow during the first hours; there are rapid fluctuations in the quantities of leukocytes populations. In this work, we investigated leukocytes subsets counts in two types of blood samples (cord blood and capillary blood) extracted from healthy newborns. METHODS: Blood samples from Mexican neonates were collected by Instituto Nacional de Pediatría with written informed consent. For all samples we determined leukocytes populations; neutrophils, monocytes, total lymphocytes, and populations: T CD3+ cells, TCD4+ cells, T CD8+ cells, B CD19+ cells and NK CD16+56 cells by flow cytometry. We used the Mann-Whitney U test to compare leukocytes of cord and capillary blood; also to analyze the differences between gender and we obtained reference values of the cord and capillary blood in neonates. RESULTS: We observed higher absolute counts and frequencies of total lymphocyte in capillary blood compared with cord blood. In absolute numbers, the capillary blood showed significant differences in neutrophils, monocytes, lymphocytes, T CD3+ cells, T CD4+ cells, T CD8+ cells, B CD19+ cells, and NK cells; no significant differences were observed between genders. DISCUSSION: Our data contribute to newborn Mexican reference values for all these populations of leukocytes. We found that the dispersal range differs between the two types of blood, suggesting a different fate in the immune response. Immunophenotyping of the blood cell population to identify these cells is an essential tool in the diagnosis and follow-up of neonates with immunodeficiencies and other immune disorders


No disponible


Subject(s)
Humans , Male , Female , Infant, Newborn , T-Lymphocyte Subsets , Reference Values , Fetal Blood , Mexico , Umbilical Cord/immunology , Flow Cytometry , Neutrophils , Monocytes , Lymphocyte Count/methods
10.
Stem Cell Res Ther ; 11(1): 207, 2020 05 27.
Article in English | MEDLINE | ID: mdl-32460839

ABSTRACT

The novel coronavirus disease 2019 (COVID-19) has grown to be a global public health emergency since patients were first detected in Wuhan, China. Thus far, no specific drugs or vaccines are available to cure the patients with COVID-19 infection. The immune system and inflammation are proposed to play a central role in COVID-19 pathogenesis. Mesenchymal stem cells (MSCs) have been shown to possess a comprehensive powerful immunomodulatory function. Intravenous infusion of MSCs has shown promising results in COVID-19 treatment. Here, we report a case of a severe COVID-19 patient treated with human umbilical cord Wharton's jelly-derived MSCs (hWJCs) from a healthy donor in Liaocheng People's Hospital, China, from February 24, 2020. The pulmonary function and symptoms of the patient with COVID-19 pneumonia was significantly improved in 2 days after hWJC transplantation, and recovered and discharged in 7 days after treatment. After treatment, the percentage and counts of lymphocyte subsets (CD3+, CD4+, and CD8+ T cell) were increased, and the level of IL-6, TNF-α, and C-reactive protein is significantly decreased after hWJC treatment. Thus, the intravenous transplantation of hWJCs was safe and effective for the treatment of patients with COVID-19 pneumonia, especially for the patients in a critically severe condition. This report highlights the potential of hWJC infusions as an effective treatment for COVID-19 pneumonia.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Pneumonia, Viral/therapy , Betacoronavirus/genetics , C-Reactive Protein/immunology , C-Reactive Protein/metabolism , COVID-19 , COVID-19 Testing , Clinical Laboratory Techniques , Coronavirus Infections/diagnosis , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Humans , Immunomodulation , Infusions, Intravenous , Interleukin-6/blood , Interleukin-6/immunology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/virology , Male , Mesenchymal Stem Cells/immunology , Middle Aged , Pandemics , Pneumonia, Viral/diagnosis , Pneumonia, Viral/immunology , SARS-CoV-2 , Treatment Outcome , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology , Umbilical Cord/cytology , Umbilical Cord/immunology , Wharton Jelly/cytology , Wharton Jelly/immunology , COVID-19 Drug Treatment
11.
Autoimmunity ; 53(4): 201-209, 2020 06.
Article in English | MEDLINE | ID: mdl-32321315

ABSTRACT

Mesenchymal stem cells (MSCs) are identified as a promising tool for the treatment of autoimmune diseases, and several microRNAs (miRNAs) are shown to exhibit vital roles in immune diseases. However, their function and mechanism in systemic lupus erythematosus (SLE) is still unclear. The qRT-PCR analysis was employed to investigate level of miR-153-3p. Subsequently, western blot and luciferase reporter assays were carried out to determine miR-153-3p targets. Cell proliferation and migration were determined using EdU proliferation assays and transwell migration assays. Apoptosis levels were evaluated by annexin V staining and flow cytometry. We used human umbilical cord-derived mesenchymal stem cells (UC-MSCs) transplantation to treat MRL/lpr mice. It was observed that miR-153-3p was upregulated in patients with SLE, and was closely related to SLE disease activity. Overexpression of miR-153-3p decreased UC-MSCs proliferation and migration, and weakened UC-MSCs-mediated decrease of follicular T helper (Tfh) cells and increase of regulatory T (Treg) cells through repressing PELI1 in vitro. We also found that PELI1 overexpression abolished the function of miR-153-3p on UC-MSCs. Furthermore, miR-153-3p overexpression weakened the therapeutic effect of UC-MSCs in MRL/lpr mice in vivo. Taken together, all data suggested that miR-153-3p is a mediator of SLE UC-MSCs regulation and may function as a new therapeutic target for the treatment of lupus.


Subject(s)
Lupus Erythematosus, Systemic/immunology , Mesenchymal Stem Cells/immunology , MicroRNAs/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Female , Humans , Mice , Mice, Inbred MRL lpr , Proteinuria/urine , T Follicular Helper Cells/immunology , T-Lymphocytes, Regulatory/immunology , Umbilical Cord/immunology
12.
Stem Cell Rev Rep ; 15(6): 900-918, 2019 12.
Article in English | MEDLINE | ID: mdl-31741193

ABSTRACT

Therapeutic options for end-stage organ failure are often limited to whole organ transplantation. The tolerance or rejection of the transplanted organ is driven by both early non-specific innate and specific adaptive responses. The use of mesenchymal stromal cells (MSCs) is considered a promising tool in regenerative medicine. Human umbilical cord (HUC) is an easily available source of MSCs, without relevant ethical issues. Moreover, Wharton's jelly-derived MSCs (WJ-MSCs), showed consistent immunomodulatory features that may be useful to promote immune tolerance in the host after transplantation. Few data are available on the phenotype of WJ-MSCs in situ. We investigated the expression of immune-related molecules, such as HLAs, IDO, CD276/B7-H3, and others, both in situ (HUC) and in in vitro-cultured WJ-MSCs. Morphological and biochemical techniques were used to define the expression of such molecules. In addition, we focused on the possible role of CD276/B7-H3 on T cells proliferation inhibition. We assessed CD276/B7-H3 expression by WJ-MSCs both in situ and alongside cell culture. WJ-MSCs were able to suppress T cell proliferation in mixed lymphocyte reaction (MLR). Moreover, we describe for the first time a specific role for CD276/B7-H3, since the immunomodulatory ability of WJ-MSCs was abolished upon anti-CD276/B7-H3 antibody addition to the MLR. These results further detail the immune regulation properties and tolerance induction exerted by human WJ-MSCs, in particular pointing to CD276/B7-H3 as one of the main involved factors. These data further suggest WJ-MSCs as potent tools to modulate local immune response in "support-type" regenerative medicine approaches.


Subject(s)
B7 Antigens/antagonists & inhibitors , Cell Differentiation , Lymphocyte Activation/immunology , Mesenchymal Stem Cells/immunology , Umbilical Cord/immunology , Wharton Jelly/immunology , B7 Antigens/immunology , Cell Proliferation , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Humans , In Vitro Techniques , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Wharton Jelly/cytology
13.
Zhonghua Nei Ke Za Zhi ; 58(11): 819-822, 2019 Nov 01.
Article in Chinese | MEDLINE | ID: mdl-31665857

ABSTRACT

The efficacy and safety of co-transplantation of unrelated donor peripheral blood stem cells (UD-PBSCs) combined with umbilical cord mesenchymal stem cells (UC-MSCs) in refractory severe aplastic anemia-Ⅱ(RSAA-Ⅱ) were analyzed retrospectively. Fifteen patients with RSAA-Ⅱ underwent UD-PBSCs and UC-MSCs co-transplantation, among whom 14 cases had hematopoietic reconstitution without severe graft versus-host disease (GVHD). The 5-year overall survival rate was 78.57%. Combination of UD-PBSCs and UC-MSCs transplantation could be a safe and effective option for RSAA-Ⅱ.


Subject(s)
Anemia, Aplastic/surgery , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Umbilical Cord/physiology , Unrelated Donors , Anemia, Aplastic/immunology , Anemia, Aplastic/mortality , Anemia, Aplastic/pathology , China/epidemiology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/mortality , Hematopoietic Stem Cells/immunology , Humans , Mesenchymal Stem Cells , Peripheral Blood Stem Cells , Retrospective Studies , Survival Rate , Tissue Donors , Transplantation Conditioning/methods , Treatment Outcome , Umbilical Cord/immunology
14.
Sci Rep ; 9(1): 14415, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31595012

ABSTRACT

Cell-based therapies are increasingly focused on allogeneic stem cell sources because of several advantages in eliminating donor variability (e.g., aging and disease pathophysiology) affecting stem cell quality and in cell-banked sourcing of healthy donors to enable "off-the-shelf" products. However, allogeneic cell therapy is limited by host patient immunologic competence and inconsistent performance due to cell delivery methods. To address allogeneic cell therapy limitations, this study developed a new allogeneic stem cell sheet using human umbilical cord mesenchymal stem cells (hUC-MSC) that present low antigenicity (i.e., major histocompatibility complex, MHC). Optimal conditions including cell density, passage number, and culture time were examined to fabricate reliable hUC-MSC sheets. MHC II antigens correlated to alloimmune rejection were barely expressed in hUC-MSC sheets compared to other comparator MSC sheets (hBMSC and hADSC). hUC-MSC sheets easily graft spontaneously onto subcutaneous tissue in immune-deficient mice within 10 minutes of placement. No sutures are required to secure sheets to tissue because sheet extracellular matrix (ECM) actively facilitates cell-target tissue adhesion. At 10 days post-transplantation, hUC-MSC sheets remain on ectopic target tissue sites and exhibit new blood vessel formation. Furthermore, implanted hUC-MSC sheets secrete human HGF continuously to the murine target tissue. hUC-MSC sheets described here should provide new insights for improving allogenic cell-based therapies.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Transplantation, Homologous , Animals , Culture Media/pharmacology , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Humans , Immunocompetence/drug effects , Immunocompetence/immunology , Mesenchymal Stem Cells/immunology , Mice , Regenerative Medicine/methods , Tissue Engineering/methods , Umbilical Cord/cytology , Umbilical Cord/growth & development , Umbilical Cord/immunology
15.
Int Immunopharmacol ; 74: 105687, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31295689

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease which is lack of effective therapies. Abnormal activation, proliferation, and differentiation of T lymphocytes are closely related to RA. Mesenchymal stem cells (MSCs) can be used for RA treatment due to their immunoregulatory effects. However, the specific molecular mechanisms have not been fully elucidated and the therapeutic effect has been inconsistent. This study investigated the immunomodulatory effect of human umbilical cord MSCs (hUCMSCs) on T lymphocytes in collagen-induced arthritis (CIA) rats and RA patients to clarify the possible mechanism of hUCMSCs in RA treatment. The effects of hUCMSCs on arthritis index, radiological and synovial pathological changes, T lymphocyte proliferation and apoptosis, RORγt and Foxp3 expression, Th17 and Treg cell ratios, and IL-17 and TGF-ß levels were assessed in CIA rats. Further, we verified the effect of hUCMSCs in RA patients, and compared the effect of hUCMSCs with that of hUCMSC derived extracellular vesicles (EVs). The results showed that hUCMSCs inhibited the proliferation and promoted apoptosis in T lymphocytes, downregulated RORγt mRNA and protein expression, decreased Th17 cell ratio, upregulated Foxp3 mRNA and protein expression, and increased Treg cell ratio in the spleen. Furthermore, they downregulated RORγt and Foxp3 expression in the joints, and inhibited IL-17 and promoted TGF-ß expression in the serum, thereby improving arthritis, delaying radiological progression, and inhibiting synovial hyperplasia in CIA rats. In vitro the effects of hUCMSCs and EVs were consistent with those in vivo. Therefore, hUCMSCs may be expected to serve as a new therapy for RA.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Umbilical Cord/immunology , Animals , Apoptosis , Arthritis, Experimental/therapy , Arthritis, Rheumatoid/therapy , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Immunomodulation , Interleukin-17/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Rats , Rats, Wistar , Transforming Growth Factor beta/metabolism
16.
Stem Cell Res Ther ; 10(1): 79, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30845980

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs) are considered potential candidates that hold great promise in the treatment of immune-related diseases. For therapeutic applications, it is necessary to isolate and expand MSCs with procedures complying with good manufacturing practice (GMP). Recent studies reported the use of human serum (HS) instead of fetal bovine serum (FBS) for the expansion of bone marrow-derived MSCs. Nevertheless, there are only limited data on HS as an alternative to FBS for the isolation and expansion of umbilical (UC-MSCs) and placenta-derived MSCs (PL-MSCs). In this study, we evaluate the effect of HS compared to FBS on the proliferative and immunosuppressive capacities of these MSCs. METHODS: PL-MSCs and UC-MSCs were isolated and cultured in HS- or FBS-supplemented media. The MSC characteristics, including morphology, immunophenotype, and differentiation ability, were verified. The proliferative and immunosuppressive capacities were also examined. In addition, the proliferative-enhancing factors in both sera were explored using proteomic analysis. RESULTS: PL-MSCs and UC-MSCs proliferated faster in HS-supplemented medium than in equivalent levels of FBS-supplemented medium. Adipogenic and osteogenic differentiations occurred at nearly identical levels in HS- and FBS-supplemented media. Interestingly, MSCs cultured in HS-supplemented medium had a similar immunosuppressive effect as MSCs cultured in FBS-supplemented medium. Proteomic analysis revealed that Con-A binding glycoproteins with a molecular weight > 100 kDa in FBS could significantly enhance MSC proliferation. In contrast, the proliferative enhancing factors in HS were found in the Con-A non-binding fraction and WGA binding fraction with a molecular weight > 100 kDa. CONCLUSIONS: Taken together, our results suggest applications for the use of HS instead of FBS for the isolation and expansion of PL-MSCs and UC-MSCs for cell therapy in the future. Furthermore, this study identifies factors in HS that are responsible for its proliferative and immunosuppressive effects and might thus lead to the establishment of GMPs for the therapeutic use of MSCs.


Subject(s)
Cell Proliferation , Immunomodulation , Mesenchymal Stem Cells/immunology , Placenta/immunology , Serum , Umbilical Cord/immunology , Female , Humans , Mesenchymal Stem Cells/cytology , Placenta/cytology , Pregnancy , Umbilical Cord/cytology
17.
J Steroid Biochem Mol Biol ; 186: 154-160, 2019 02.
Article in English | MEDLINE | ID: mdl-30359690

ABSTRACT

Clinical and epidemiological evidence supports that pregnancies carrying a male fetus are more vulnerable to infections and preterm birth, probably due to testosterone immunosuppressive properties. In human placentas, testosterone lowers the expression of CYP27B1, the vitamin D (VD)-activating enzyme, diminishing cathelicidin synthesis, a potent VD-dependent antimicrobial peptide (AMP). VD also stimulates other AMPs, including defensins. To get insights into the increased male vulnerability mechanisms, we investigated the relationship between fetal sex and the immunoendocrine milieu at the fetoplacental unit. For this, umbilical vein serum and placental samples were collected from healthy newborns. In males' serum, testosterone levels were significantly higher and negatively associated with TNF-α, a cytokine that strengthens the immune response. Males showed lower serum TNF-α and increased levels and gene expression of the immunosuppressive cytokine IL-10. Only in female samples there was a positive association (P < 0.05) between AMPs and both TNF-α and CYP27B1 and between 25-hydroxyvitamin D3 and IL-1ß serum levels. Accordingly, VD-metabolites (25-hydroxyvitamin D3, calcitriol) significantly stimulated IL-1ß gene expression in cultured trophoblasts. Interestingly, IL-1ß mRNA correlated positively with defensins (P < 0.05) in males, but not with cathelicidin expression, which was significantly diminished in comparison to females. Our data suggest that high umbilical serum testosterone and IL-10 in males could explain reduced TNF-α levels and lack of association between VD-dependent innate immunity markers and proinflammatory cytokines expression in the fetoplacental unit. Altogether, our observations imply a restricted basal immune milieu in males compared to females, which may help understand the higher male susceptibility to adverse perinatal outcomes.


Subject(s)
Testosterone/blood , Tumor Necrosis Factor-alpha/blood , Adult , Female , Humans , Immunity, Innate , Infant, Newborn , Interleukin-10/blood , Interleukin-10/immunology , Male , Placenta/chemistry , Placenta/immunology , Pregnancy , Testosterone/immunology , Tumor Necrosis Factor-alpha/immunology , Umbilical Cord/blood supply , Umbilical Cord/immunology , Young Adult
18.
Stem Cell Res Ther ; 9(1): 237, 2018 09 17.
Article in English | MEDLINE | ID: mdl-30223894

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) play an anti-inflammatory role by secreting certain bioactive molecules to exert their therapeutic effects for disease treatment. However, the underlying mechanism of MSCs in chronic autoimmune liver diseases-primary biliary cholangitis (PBC), for example-remains to be elucidated. METHODS: Human umbilical cord-derived MSCs (UC-MSCs) were injected intravenously into 2-octynoic acid coupled to bovine serum albumin (2OA-BSA)-induced autoimmune cholangitis mice. Serum levels of biomarkers and autoantibodies, histologic changes in the liver, diverse CD4+ T-cell subsets in different tissues, and chemokine activities were analyzed. Moreover, we investigated galectin-9 (Gal-9) expression and its function in UC-MSCs. RESULTS: In this study, UC-MSC transplantation (UC-MSCT) significantly ameliorated liver inflammation, primarily by diminishing T helper 1 (Th1) and Th17 responses as well as modifying liver chemokine activities in experimental autoimmune cholangitis mice. Mechanistically, UC-MSCs significantly repressed the proliferation of CD4+ T cells and suppressed the differentiation of Th1 and Th17 cells, which was likely dependent on Gal-9. Furthermore, the signal transducer and activator of transcription (STAT) and c-Jun N-terminal kinase (JNK) signaling pathways were involved in the production of Gal-9 in UC-MSCs. CONCLUSIONS: These results suggest that Gal-9 contributes significantly to UC-MSC-mediated therapeutic effects and improve our understanding of the immunomodulatory mechanisms of MSCs in the treatment of PBC.


Subject(s)
Autoimmune Diseases/therapy , Galectins/genetics , Immunomodulation , Liver Cirrhosis, Biliary/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Bile Ducts/immunology , Bile Ducts/pathology , Coculture Techniques , Epithelial Cells/immunology , Epithelial Cells/pathology , Fatty Acids, Monounsaturated/chemistry , Female , Galectins/immunology , Gene Expression Regulation , Humans , Immunoconjugates/administration & dosage , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/immunology , Liver Cirrhosis, Biliary/chemically induced , Liver Cirrhosis, Biliary/immunology , Liver Cirrhosis, Biliary/pathology , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , STAT Transcription Factors/genetics , STAT Transcription Factors/immunology , Serum Albumin, Bovine/administration & dosage , Serum Albumin, Bovine/chemistry , Signal Transduction , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Transplantation, Heterologous , Umbilical Cord/cytology , Umbilical Cord/immunology
19.
Stem Cell Res Ther ; 9(1): 220, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30107860

ABSTRACT

BACKGROUND: Mesenchymal stromal cell (MSC)-based therapy is a promising strategy for preventing the progression of chronic kidney disease (CKD), with the potential to induce tissue regeneration. In search of the best cellular source we compared, in the rat model of adriamycin (ADR) nephropathy, the regenerative potential of human stromal cells of non-renal origin, such as bone marrow (bm) MSCs and umbilical cord (uc) MSCs, with that of newly discovered stromal cells of renal origin, the kidney perivascular cells (kPSCs) known to exhibit tissue-specific properties. METHODS: The therapeutic effect of repeated infusions of human bmMSCs, ucMSCs, kPSCs (1.5 × 106 cells/rats) or conditioned medium from ucMSCs was studied in athymic rats with ADR-induced nephropathy (7.9 mg/kg). The ability of the three stromal cell populations to engraft the damaged kidney was evaluated by detecting the presence of human nuclear antigenpos cells. Glomerular podocyte loss and endothelial damage, sclerotic lesions and inflammation were assessed at 14 and 28 days. In-vitro experiments with a transwell system were performed to investigate the effects of different stromal cell populations on parietal epithelial cells (PECs) activated or not with albumin or angiotensin II for 24 h. RESULTS: Infusions of non-renal and renal stromal cells resulted in a comparable engraftment into the kidney, in the peritubular areas and around the glomerular structures. All three cell populations limited podocyte loss and glomerular endothelial cell injury, and attenuated the formation of podocyte and PEC bridges. This translated into a reduction of glomerulosclerosis and fibrosis. Human ucMSCs had an anti-inflammatory effect superior to that of the other stromal cells, reducing macrophage infiltration and inducing polarisation towards the M2 macrophage phenotype. Conditioned medium from ucMSCs shared the same renoprotective effects of the cells. Consistent with in-vivo data, bmMSCs and kPSCs, but even more so ucMSCs, limited proliferation, migratory potential and extracellular matrix production of activated PECs, when cultured in a transwell system. CONCLUSIONS: Our data indicate that either non-renal or renal stromal cells induce renal tissue repair, highlighting ucMSCs and their conditioned medium as the most reliable clinical therapeutic tool for CKD patients.


Subject(s)
Glomerulosclerosis, Focal Segmental/therapy , Graft Survival , Mesenchymal Stem Cell Transplantation , Renal Insufficiency, Chronic/therapy , Umbilical Cord/cytology , Animals , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Proliferation , Coculture Techniques , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Doxorubicin/administration & dosage , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/pathology , Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/immunology , Glomerulosclerosis, Focal Segmental/pathology , Humans , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Podocytes/drug effects , Podocytes/immunology , Podocytes/pathology , Rats , Rats, Nude , Regeneration , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/pathology , Transplantation, Heterologous , Umbilical Cord/immunology , Umbilical Cord/transplantation
20.
Zhonghua Shao Shang Za Zhi ; 34(8): 556-558, 2018 Aug 20.
Article in Chinese | MEDLINE | ID: mdl-30157561

ABSTRACT

Objective: To explore the effect of debridement combined with vacuum sealing drainage (VSD) on the treatment of severe infection in abdominal wall due to allogeneic umbilical cord embedded in abdominal wall for immunotherapy. Methods: From January 2015 to December 2016, 12 patients with severe infection in abdominal wall due to allogeneic umbilical cord embedded in abdominal wall for immunotherapy were admitted to our department. They were conducted with systemic anti-infective treatment, local debridement, and VSD. The wounds were continuously washed for 3 to 5 days after the VSD device installed, with negative pressure value from -16.0 to -12.0 kPa. The VSD device was removed 5 to 7 days later. Continue wound dressing by aseptic ribbon gauze was stuffed in the cavity, and the incision was sutured after the granulation tissue grew well in the cavity. Results: In all patients, allogeneic umbilical cords were completely removed and abdominal infection was cured. The wounds healed well, the sensory function of abdominal was normal, and the activity was not restricted. All the patients were followed up for 3 to 6 months with no reinfection or incisional hernia. Conclusions: Embeding the whole allogeneic umbilical cord in abdominal wall for immunotherapy can lead to severe infection in abdominal wall. Abdominal infection can be cured by debridement combined with VSD with good clinical results.


Subject(s)
Abdominal Wall/physiopathology , Debridement , Drainage , Infections/therapy , Negative-Pressure Wound Therapy , Umbilical Cord/immunology , Vacuum , Humans , Immunotherapy
SELECTION OF CITATIONS
SEARCH DETAIL
...