Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Placenta ; 126: 175-183, 2022 08.
Article in English | MEDLINE | ID: mdl-35853410

ABSTRACT

INTRODUCTION: In preeclampsia (PE), inadequate remodelling of spiral arterioles in the decidua basalis causes oxidative stress and subsequent increased release of antiangiogenic soluble endoglin (sENG) into the maternal circulation. Decidual mesenchymal stem/stromal cells (DMSCs) reside adjacent to endothelial cells in this vascular niche. Surprisingly, DMSCs express membrane-bound ENG (CD105). PE-affected DMSCs (PE-DMSCs) are abnormal and due to reduced extravillous invasion, more of them are present, but the significance of this is not known. METHODS: DMSCs were isolated and characterised from normotensive control and severe-PE placentae. Extracellular vesicle (EV) types, shed microvesicles (sMV) and exosomes, were isolated from DMSC conditioned media (DMSCCM), respectively. Secretion of ENG by DMSCs was assessed by ELISA of DMSCCM, with and without EV depletion. The effects of reducing ENG concentration, by blocking antibody, on human umbilical vein endothelial cell (HUVEC) attachment were assessed by xCELLigence real-time functional assays. RESULTS: ENG was detected in DMSCCM and these levels significantly decreased when depleted of exosomes and sMV. There was no significant difference in the amount of ENG secreted by control DMSCs and PE-DMSCs. Blocking ENG in concentrated DMSCCM, used to treat HUVECs, improved endothelial cell attachment. DISCUSSION: In normotensive pregnancies, DMSC secretion of ENG likely has a beneficial effect on endothelial cells. However, in PE pregnancies, shallow invasion of the spiral arterioles exposes more PE-DMSC derived sources of ENG (soluble and EV). The presence of these PE-DMSCs in the vascular niche contributes to endothelial cell dysfunction.


Subject(s)
Mesenchymal Stem Cells , Pre-Eclampsia , Endoglin/metabolism , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Pregnancy
2.
Stem Cell Res Ther ; 10(1): 50, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30728068

ABSTRACT

BACKGROUND: Mesenchymal stem cells derived from the chorionic villi of human placentae (pMSCs) produce a unique array of mediators that regulate the essential cellular functions of their target cells. These properties make pMSCs attractive candidates for cell-based therapy. Here, we examined the effects of culturing human natural killer (NK) cells with pMSCs on NK cell functions. METHODS: pMSCs were cultured with IL-2-activated and non-activated NK cells. NK cell proliferation and cytolytic activities were monitored. NK cell expression of receptors mediating their cytolytic activity against pMSCs, and the mechanisms underlying this effect on pMSCs, were also investigated. RESULTS: Our findings show that IL-2-activated NK cells, but not freshly isolated NK cells, efficiently lyse pMSCs and that this response might involve the activating NK cell receptor CD69. Interestingly, although pMSCs expressed HLA class I molecules, they were nevertheless lysed by NK cells, suggesting that HLA class I antigens do not play a significant role in protecting pMSCs from NK cell cytolytic activity. Co-culturing NK cells with pMSCs also inhibited NK cell expression of receptors, including CD69, NKpG2D, CD94, and NKp30, although these co-cultured NK cells were not inhibited in lysing cancer cells in vitro. Importantly, co-cultured NK cells significantly increased their production of molecules with anti-tumor effects. CONCLUSIONS: These findings suggest that pMSCs might have potential applications in cancer therapy.


Subject(s)
Chorionic Villi/metabolism , Killer Cells, Natural/metabolism , Mesenchymal Stem Cells/metabolism , Cell Differentiation , Cell Proliferation , Humans
3.
Stem Cell Res Ther ; 9(1): 275, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30359307

ABSTRACT

BACKGROUND: Human decidua basalis mesenchymal stem/multipotent stromal cells (DBMSCs) inhibit endothelial cell activation by inflammation induced by monocytes. This property makes them a promising candidate for cell-based therapy to treat inflammatory diseases, such as atherosclerosis. This study was performed to examine the ability of DBMSCs to protect endothelial cell functions from the damaging effects resulting from exposure to oxidatively stress environment induced by H2O2 and monocytes. METHODS: DBMSCs were co-cultured with endothelial cells isolated from human umbilical cord veins in the presence of H2O2 and monocytes, and various functions of endothelial cell were then determined. The effect of DBMSCs on monocyte adhesion to endothelial cells in the presence of H2O2 was also examined. In addition, the effect of DBMSCs on HUVEC gene expression under the influence of H2O2 was also determined. RESULTS: DBMSCs reversed the effect of H2O2 on endothelial cell functions. In addition, DBMSCs reduced monocyte adhesion to endothelial cells and also reduced the stimulatory effect of monocytes on endothelial cell proliferation in the presence of H2O2. Moreover, DBMSCs modified the expression of many genes mediating important endothelial cell functions. Finally, DBMSCs increased the activities of glutathione and thioredoxin reductases in H2O2-treated endothelial cells. CONCLUSIONS: We conclude that DBMSCs have potential for therapeutic application in inflammatory diseases, such as atherosclerosis by protecting endothelial cells from oxidative stress damage. However, more studies are needed to elucidate this further.


Subject(s)
Human Umbilical Vein Endothelial Cells/drug effects , Hydrogen Peroxide/pharmacology , Mesenchymal Stem Cells/drug effects , Monocytes/metabolism , Oxidative Stress/drug effects , Adult , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers/metabolism , Cell Adhesion/drug effects , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Proliferation/drug effects , Coculture Techniques , Culture Media, Conditioned/pharmacology , Decidua/cytology , Decidua/metabolism , Female , Gene Expression , Glutathione Reductase/genetics , Glutathione Reductase/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Monocytes/cytology , Pregnancy , Thioredoxin-Disulfide Reductase/genetics , Thioredoxin-Disulfide Reductase/metabolism , Umbilical Cord/cytology , Umbilical Cord/metabolism
4.
Stem Cell Res Ther ; 9(1): 238, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30241570

ABSTRACT

BACKGROUND: Mesenchymal stem/stromal cells derived from chorionic villi of human term placentae (pMSCs) protect human endothelial cells from injury induced by hydrogen peroxide (H2O2). In diabetes, elevated levels of glucose (hyperglycaemia) induce H2O2 production, which causes the endothelial dysfunction that underlies the enhanced immune responses and adverse complications associated with diabetes, which leads to thrombosis and atherosclerosis. In this study, we examined the ability of pMSCs to protect endothelial cell functions from the negative impact of high level of glucose. METHODS: pMSCs isolated from the chorionic villi of human term placentae were cultured with endothelial cells isolated from human umbilical cord veins in the presence of glucose. Endothelial cell functions were then determined. The effect of pMSCs on gene expression in glucose-treated endothelial cells was also determined. RESULTS: pMSCs reversed the effect of glucose on key endothelial cell functions including proliferation, migration, angiogenesis, and permeability. In addition, pMSCs altered the expression of many genes that mediate important endothelial cell functions including survival, apoptosis, adhesion, permeability, and angiogenesis. CONCLUSIONS: This is the first comprehensive study to provide evidence that pMSCs protect endothelial cells from glucose-induced damage. Therefore, pMSCs have potential therapeutic value as a stem cell-based therapy to repair glucose-induced vascular injury and prevent the adverse complications associated with diabetes and cardiovascular disease. However, further studies are necessary to reveal more detailed aspects of the mechanism of action of pMSCs on glucose-induced endothelial damage in vitro and in vivo.


Subject(s)
Culture Media, Conditioned/pharmacology , Gene Expression Regulation/drug effects , Glucose/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Mesenchymal Stem Cells/metabolism , Apoptosis/drug effects , Cell Membrane Permeability/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemokines/genetics , Chemokines/metabolism , Chorionic Villi/metabolism , Coculture Techniques , Collagen/chemistry , Collagen/genetics , Collagen/metabolism , Drug Combinations , Endothelins/genetics , Endothelins/metabolism , Female , Glucose/antagonists & inhibitors , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukins/genetics , Interleukins/metabolism , Laminin/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mesenchymal Stem Cells/cytology , Pregnancy , Proteoglycans/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction , THP-1 Cells , Umbilical Cord/cytology , Umbilical Cord/metabolism
5.
Stem Cells Int ; 2018: 6480793, 2018.
Article in English | MEDLINE | ID: mdl-29795719

ABSTRACT

Stem cell-based therapies rely on stem cell ability to repair in an oxidative stress environment. Preconditioning of mesenchymal stem cells (MSCs) to a stress environment has beneficial effects on their ability to repair injured tissues. We previously reported that MSCs from the decidua basalis (DBMSCs) of human placenta have many important cellular functions that make them potentially useful for cell-based therapies. Here, we studied the effect of DBMSC preconditioning to a stress environment. DBMSCs were exposed to various concentrations of hydrogen peroxide (H2O2), and their functions were then assessed. DBMSC expression of immune molecules after preconditioning was also determined. DBMSC preconditioning with H2O2 enhanced their proliferation, colonogenicity, adhesion, and migration. In addition, DBMSCs regardless of H2O2 treatment displayed antiangiogenic activity. H2O2 preconditioning also increased DBMSC expression of genes that promote cellular functions and decreased the expression of genes, which have opposite effect on their functions. Preconditioning also reduced DBMSC expression of IL-1ß, but had no effects on the expression of other immune molecules that promote proliferation, adhesion, and migration. These data show that DBMSCs resist a toxic environment, which adds to their potential as a candidate stem cell type for treating various diseases in hostile environments.

6.
Stem Cell Res Ther ; 9(1): 102, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29650045

ABSTRACT

BACKGROUND: Human decidua parietalis mesenchymal stem/multipotent stromal cells (DPMSCs) have unique phenotypic and functional properties that make them promising candidates for cell-based therapy. Here, we investigated DPMSC interaction with natural killer (NK) cells, and the effects of this interaction on NK cell phenotypic characteristics and functional activities. METHODS: DPMSCs isolated from the decidua parietalis of human fetal membranes were cultured with interleukin (IL)-2-activated and IL-2-unactivated NK cells isolated from healthy human peripheral blood. NK cell proliferation and cytolytic activities were then examined using functional assays. NK cell expression of receptors mediating the cytolytic activity against DPMSCs, and the mechanism underlying this effect on DPMSCs, were also examined using flow cytometry and light microscopy, respectively. RESULTS: DPMSCs stimulated IL-2-induced proliferation of resting NK cells and the proliferation of activated NK cells. Moreover, IL-2-activated NK cells, but not freshly isolated NK cells, efficiently lysed DPMSCs. The induction of this NK cell cytolytic activity against DPMSCs was mediated by the activating NK cell receptors NKG2D, CD69, NKp30, and NKp44. However, DPMSCs showed a direct induction of NK cell cytolytic activity through CD69. We also found that DPMSCs expressed the ligands for these activating NK cell receptors including Nectin-2, ULBP-2, MICA, and MICB. Although DPMSCs expressed HLA class I molecules, they were susceptible to lysis by NK cells, suggesting that HLA class I antigens do not play a significant role in NK cell cytolytic action. In addition, DPMSCs did not inhibit NK cell cytolytic activity against cancer cells. Importantly, DPMSCs significantly increased NK expression of inflammatory molecules with anticancer activities. CONCLUSIONS: We conclude that DPMSCs have potential for therapeutic application in cancer therapy, but not in transplantation or immunological diseases.


Subject(s)
Decidua/metabolism , Killer Cells, Natural/immunology , Mesenchymal Stem Cells/immunology , Female , Humans , Male
7.
Biomater Sci ; 6(1): 60-78, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29184934

ABSTRACT

Tissue regeneration by stem cells is driven by the paracrine activity of shedding vesicles and exosomes, which deliver specific cargoes to the recipient cells. Proteins, RNA, cytokines and subsequent gene expression, orchestrate the regeneration process by improving the microenvironment to promote cell survival, controlling inflammation, repairing injury and enhancing the healing process. The action of microRNA is widely accepted as an essential driver of the regenerative process through its impact on multiple downstream biological pathways, and its ability to regulate the host immune response. Here, we present an overview of the recent potential uses of exosomes for regenerative medicine and tissue engineering. We also highlight the differences in composition between shedding vesicles and exosomes that depend on the various types of stem cells from which they are derived. The conditions that affect the production of exosomes in different cell types are deliberated. This review also presents the current status of candidate exosomal microRNAs for potential therapeutic use in regenerative medicine, and in applications involving widely studied organs and tissues such as heart, lung, cartilage and bone.


Subject(s)
Exosomes/chemistry , Extracellular Vesicles/chemistry , Wound Healing/physiology , Animals , Humans , MicroRNAs/genetics , Regenerative Medicine , Tissue Engineering/methods
8.
Placenta ; 59: 74-86, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28502524

ABSTRACT

Mesenchymal stem/stromal cells derived from chorionic villi of human term placentae (pMSCs) produce a unique combination of molecules, which modulate important cellular functions of their target cells while concurrently suppressing their immune responses. These properties make MSCs advantageous candidates for cell-based therapy. Our first aim was to examine the effect of high levels of oxidative stress on pMSC functions. pMSCs were exposed to hydrogen peroxide (H2O2) and their ability to proliferate and adhere to an endothelial cell monolayer was determined. Oxidatively stressed pMSCs maintained their proliferation and adhesion potentials. The second aim was to measure the ability of pMSCs to prevent oxidative stress-related damage to endothelial cells. Endothelial cells were exposed to H2O2, then co-cultured with pMSCs, and the effect on endothelial cell adhesion, proliferation and migration was determined. pMSCs were able to reverse the damaging effects of oxidative stress on the proliferation and migration but not on the adhesion of endothelial cells. These data indicate that pMSCs are not only inherently resistant to oxidative stress, but also protect endothelial cell functions from oxidative stress-associated damage. Therefore, pMSCs could be used as a therapeutic tool in inflammatory diseases by reducing the effects of oxidative stress on endothelial cells.


Subject(s)
Cell- and Tissue-Based Therapy , Endothelial Cells/physiology , Mesenchymal Stem Cells/physiology , Oxidative Stress , Placenta/cytology , Cell Adhesion , Cell Movement , Female , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Peroxide , Pregnancy
9.
Placenta ; 59: 87-95, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28411943

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) are isolated from various fetal and adult tissues such as bone marrow, adipose tissue, cord blood and placenta. Placental MSCs (pMSCs), the main focus of this review, are relatively new MSC types that are not as intensively studied compared with bone marrow-derived MSCs (BMMSCs). MSCs modulate the immune functions of important immune cells involved in alloantigen recognition and elimination, including antigen presenting cells (APCs), T cells, B cells and natural killer (NK) cells. Clinical trials, both completed and underway, employ MSCs to treat various human immunological diseases, such as multiple sclerosis (MS) and type 1 diabetes. However, the mechanisms that mediate the immunosuppressive effects of pMSCs are still largely unknown, and the safety of pMSC use in clinical settings needs further confirmation. Here, we review the current knowledge of the immunosuppressive properties of placental MSCs.


Subject(s)
Immune Tolerance , Mesenchymal Stem Cells/immunology , Placenta/cytology , Clinical Trials as Topic , Female , Humans , Immunotherapy , Lymphocytes/physiology , Mesenchymal Stem Cell Transplantation , Pregnancy
10.
Placenta ; 45: 58-62, 2016 09.
Article in English | MEDLINE | ID: mdl-27577711

ABSTRACT

Fetal growth restriction (FGR) is a leading cause of perinatal morbidity and mortality. FGR pregnancies are often associated with histological evidence of placental vascular thrombosis. The proteoglycans are important components and regulators of vascular homeostasis. Previous studies from our laboratory highlighted mRNA and protein expression differences in placental proteoglycan decorin (DCN), within a clinically well-characterised cohort of third-trimester idiopathic FGR compared with gestation-matched uncomplicated control pregnancies. We also showed that decorin contributes to abnormal angiogenesis and increased thrombin generation in vitro. These observations suggest that DCN gene expression may contribute to the etiology of FGR. Small for gestational age (SGA) is frequently used as a proxy for FGR and is defined as a birth weight below the 10th percentile of a birth weight curve. We therefore made use of a unique resource of first trimester tissues obtained via chorionic villus sampling during the first trimester to investigate the temporal relationship between altered DCN expression and any subsequent development of SGA. We hypothesized that placental DCN expression is decreased early in gestation in SGA pregnancies. Surplus chorionic villus specimens from 15 women subsequently diagnosed with FGR and 50 from women with uncomplicated pregnancies were collected. DCN mRNA and DCN protein were determined using real-time PCR and immunoblotting, respectively. Both DCN mRNA and protein were significantly decreased in placentae from first-trimester SGA-pregnancies compared with controls (p < 0.05). This is the first study to report a temporal relationship between altered placental DCN expression and subsequent development of SGA.


Subject(s)
Decorin/metabolism , Down-Regulation , Placenta/metabolism , Adult , Female , Humans , Infant, Small for Gestational Age , Maternal Age , Pregnancy , Pregnancy Trimester, First/metabolism
11.
Placenta ; 48 Suppl 1: S17-S20, 2016 12.
Article in English | MEDLINE | ID: mdl-27506263

ABSTRACT

Workshops are an important part of the IFPA annual meeting as they allow for discussion of specialised topics. At the 2015 IFPA annual meeting there were 12 themed workshops, three of which are summarized in this report. These workshops related to various aspects of placental biology and collectively covered areas of obesity and the placenta, stem cells of the feto-maternal interface, and placental immunobiology and infection.


Subject(s)
Obesity/metabolism , Placenta Diseases/metabolism , Placenta/metabolism , Stem Cells/metabolism , Female , Humans , Pregnancy
12.
Stem Cells Int ; 2016: 5184601, 2016.
Article in English | MEDLINE | ID: mdl-27087815

ABSTRACT

Mesenchymal stem cell (MSC) therapies for the treatment of diseases associated with inflammation and oxidative stress employ primarily bone marrow MSCs (BMMSCs) and other MSC types such as MSC from the chorionic villi of human term placentae (pMSCs). These MSCs are not derived from microenvironments associated with inflammation and oxidative stress, unlike MSCs from the decidua basalis of the human term placenta (DBMSCs). DBMSCs were isolated and then extensively characterized. Differentiation of DBMSCs into three mesenchymal lineages (adipocytes, osteocytes, and chondrocytes) was performed. Real-time polymerase chain reaction (PCR) and flow cytometry techniques were also used to characterize the gene and protein expression profiles of DBMSCs, respectively. In addition, sandwich enzyme-linked immunosorbent assay (ELISA) was performed to detect proteins secreted by DBMSCs. Finally, the migration and proliferation abilities of DBMSCs were also determined. DBMSCs were positive for MSC markers and HLA-ABC. DBMSCs were negative for hematopoietic and endothelial markers, costimulatory molecules, and HLA-DR. Functionally, DBMSCs differentiated into three mesenchymal lineages, proliferated, and migrated in response to a number of stimuli. Most importantly, these cells express and secrete a distinct combination of cytokines, growth factors, and immune molecules that reflect their unique microenvironment. Therefore, DBMSCs could be attractive, alternative candidates for MSC-based therapies that treat diseases associated with inflammation and oxidative stress.

13.
Exp Gerontol ; 80: 43-50, 2016 07.
Article in English | MEDLINE | ID: mdl-27090484

ABSTRACT

Pyrroloquinoline quinone (PQQ) is linked to fundamental biological processes such as mitochondrial biogenesis and lipid metabolism. PQQ may also function as an essential micronutrient during animal development. Recent studies have shown the therapeutic potential of PQQ for several age-related diseases due to its antioxidant capacity. However, whether PQQ can promote longevity is unknown. Here, we investigate the effects of PQQ on oxidative stress resistance as well as lifespan modulation in Caenorhabditis elegans. We find that PQQ enhances resistance to oxidative stress and extends the lifespan of C. elegans at optimal doses. The underlying molecular mechanism involves the increased activities of the primary lifespan extension transcriptional factors DAF-16/FOXO, the conserved oxidative stress-responsive transcription factor SKN-1/Nrf2, and upregulation of daf-16, skn-1 downstream targets including sod-3, hsp16.2, gst-1 and gst-10. Our findings uncover a novel role of PQQ in longevity, supporting PQQ as a possible dietary supplement for overall health improvement.


Subject(s)
Aging/physiology , Caenorhabditis elegans/physiology , Longevity/physiology , Oxidative Stress , PQQ Cofactor/pharmacology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , DNA-Binding Proteins/genetics , Forkhead Transcription Factors/genetics , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Transcription Factors/genetics
14.
Reprod Sci ; 23(9): 1193-207, 2016 09.
Article in English | MEDLINE | ID: mdl-26902429

ABSTRACT

Mesenchymal stem/multipotent stromal cells (MSCs) from the human placenta show stem cell-like properties useful for regenerative medicine. Previously, we reported that MSCs isolated from the fetal part of human term placentae have characteristics, which make them a potential candidate for regenerative medicine. In this study, we characterized MSC isolated from the maternal part of human term placenta. The MSCs were isolated from the decidua parietalis (DPMSCs) of human placenta using a digestion method and characterized by colony-forming unit assay and the expression of MSC markers by flow cytometry technique. In addition, DPMSC differentiation into the 3 mesenchymal lineages was also performed. Moreover, the gene and protein expression profiles of DPMSCs were identified by real-time polymerase chain reaction and flow cytometry techniques, respectively. Furthermore, proteins secreted by DPMSCs were detected by sandwich enzyme-linked immunosorbent assays. Finally, the proliferation and migration potentials of DPMSCs were also determined. The DPMSCs were positive for MSC markers and negative for hematopoietic and endothelial markers, as well as costimulatory molecules and HLA-DR. Functionally, DPMSCs formed colonies and differentiated into chondrocytes, osteocytes, and adipocytes. In addition, they proliferated and migrated in response to different stimuli. Finally, they expressed and secreted many biological and immunological factors with multiple functions. Here, we carry out an extensive characterization of DPMSCs of human placenta. We report that these cells express and secrete a wide range of molecules with multiple functions, and therefore, we suggest that these cells could be an attractive candidate for cell-based therapy.


Subject(s)
Decidua/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Cytokines/metabolism , Decidua/metabolism , Female , Gene Expression , Humans , Mesenchymal Stem Cells/metabolism , RNA, Messenger/metabolism , Receptors, Chemokine/metabolism
15.
Reproduction ; 149(5): 523-32, 2015 May.
Article in English | MEDLINE | ID: mdl-25713425

ABSTRACT

Abnormal trophoblast function is associated with fetal growth restriction (FGR). The JAK-STAT pathway is one of the principal signalling mechanisms by which cytokines and growth factors modulate cell proliferation, differentiation, cell migration and apoptosis. The expression of placental JAK-STAT genes in human idiopathic FGR is unknown. In this study, we propose the hypothesis that JAK-STAT pathway genes are differentially expressed in idiopathic FGR-affected pregnancies and contribute to abnormal feto-placental growth by modulating the expression of the amino acid transporter SNAT2, differentiation marker CGB/human chorionic gonadotrophin beta-subunit (ß-hCG) and apoptosis markers caspases 3 and 8, and TP53. Expression profiling of FGR-affected placentae revealed that mRNA levels of STAT3, STAT2 and STAT5B decreased by 69, 52 and 50%, respectively, compared with gestational-age-matched controls. Further validation by real-time PCR and immunoblotting confirmed significantly lower STAT3 mRNA and STAT3 protein (total and phosphorylated) levels in FGR placentae. STAT3 protein was localised to the syncytiotrophoblast (ST) in both FGR and control placentae. ST differentiation was modelled by in vitro differentiation of primary villous trophoblast cells from first-trimester and term placentae, and by treating choriocarcinoma-derived BeWo cells with forskolin in cell culture. Differentiation in these models was associated with increased STAT3 mRNA and protein levels. In BeWo cells treated with siRNA targeting STAT3, the mRNA and protein levels of CGB/ß-hCG, caspases 3 and 8, and TP53 were significantly increased, while that of SNAT2 was significantly decreased compared with the negative control siRNA. In conclusion, we report that decreased STAT3 expression in placentae may contribute to abnormal trophoblast function in idiopathic FGR-affected pregnancies.


Subject(s)
Apoptosis , Fetal Growth Retardation/pathology , Placenta/cytology , STAT3 Transcription Factor/metabolism , Trophoblasts/pathology , Adult , Blotting, Western , Case-Control Studies , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cohort Studies , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Infant, Low Birth Weight , Male , Placenta/metabolism , Pregnancy , Pregnancy Trimester, First , Pregnancy Trimester, Third , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , Trophoblasts/metabolism
16.
Placenta ; 36(3): 312-21, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25575436

ABSTRACT

INTRODUCTION: Maternal decidua basalis tissue attached to the placenta following delivery is a source of decidual mesenchymal stem cells (DMSCs). The in vitro characteristics of DMSCs have been partly defined but their in vivo function(s) are poorly understood. The anatomic location, or niche, provides clues regarding potential in vivo function(s) of DMSCs, but the niche has not been described. METHODS: Cells were isolated from the decidua basalis and flow cytometric analyses showed the expected phenotypic profile for MSC cell surface markers. In vitro, the cells differentiated into adipocytes, osteocytes, and chondrocytes. DMSCs were then stained with antibodies by immunofluorescence detection. RESULTS: Immunocytochemistry revealed that DMSCs were positive for FZD-9, STRO-1, 3G5, and α-SMA as expected and lacked expression of vWF and Ck7. Fluorescence in situ hybridization analysis showed the cultured cells were of maternal origin. Immunofluorescence was carried out on placental bed biopsies using the FZD-9, STRO-1, 3G5, and α-SMA antibodies. DMSCs were located in the vascular niche in decidua basalis. Immunofluorescence with antibodies to FZD-9, Ck7 and vWF revealed DMSCs in the vascular niche surrounding intact non-transformed spiral arterioles but DMSCs were absent in fully transformed spiral arterioles. DISCUSSION: Spiral arteriole remodelling is a critical feature of human pregnancy. The DMSC niche was investigated in fully transformed and non-transformed spiral arterioles. DMSCs have not been previously implicated in spiral arteriole remodelling. The absence of DMSCs around fully transformed spiral arterioles suggests they are a target for replacement or destruction by invading placental extravillous trophoblast cells, which carry out spiral arteriole remodelling.


Subject(s)
Arterioles/cytology , Decidua/cytology , Mesenchymal Stem Cells/cytology , Placentation , Stem Cell Niche , Vascular Remodeling , Adult , Antigens, Surface/metabolism , Arterioles/metabolism , Biomarkers/metabolism , Cells, Cultured , Decidua/metabolism , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mesenchymal Stem Cells/metabolism , Pregnancy
17.
Stem Cell Rev Rep ; 11(3): 423-41, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25287760

ABSTRACT

BACKGROUND: Mesenchymal stem cells derived from the chorionic villi of human term placenta (pMSCs) have drawn considerable interest because of their multipotent differentiation potential and their immunomodulatory capacity. These properties are the foundation for their clinical application in the fields of stem cell transplantation and regenerative medicine. Previously, we showed that pMSCs induce an anti-inflammatory phenotype in human macrophages. In this study, we determined whether pMSCs modify the differentiation and maturation of human monocytes into dendritic cells (DCs). The consequences on dendritic function and on T cell proliferation were also investigated. METHODS: Interleukin-4 (IL-4) and granulocyte-macrophage colony stimulating factor (GM-CSF) were used to stimulate the differentiation of monocytes into immature dendritic cells (iDCs), which were subsequently co-cultured with pMSCs. Lipopolysaccharide (LPS) was used to induce maturation of iDCs into mature dendritic cells (mDCs). Flow cytometry and enzyme-linked immunosorbent assays (ELISA) were used to quantify the effect pMSC co-culturing on DC differentiation using CD1a, a distinctive marker of DCs, as well as other molecules important in the immune functions of DCs. The phagocytic activity of iDCs co-cultured with pMSCs, and the effects of iDCs and mDC stimulation on T cell proliferation, were also investigated. RESULTS: Monocyte differentiation into iDCs was inhibited when co-cultured with pMSCs and maturation of iDCs by LPS treatment was also prevented in the presence of pMSCs as demonstrated by reduced expression of CD1a and CD83, respectively. The inhibitory effect of pMSCs on iDC differentiation was dose dependent. In addition, pMSC co-culture with iDCs and mDCs resulted in both phenotypic and functional changes as shown by reduced expression of costimulatory molecules (CD40, CD80, CD83 and CD86) and reduced capacity to stimulate CD4(+) T cell proliferation. In addition, pMSC co-culture increased the surface expression of major histocompatibility complex (MHC-II) molecules on iDCs but decreased MHC-II expression on mDCs. Moreover, pMSC co-culture with iDCs or mDCs increased the expression of immunosuppressive molecules [B7H3, B7H4, CD273, CD274 and indoleamine-pyrrole 2,3-dioxygenase (IDO). Additionally, the secretion of IL-12 and IL-23 by iDCs and mDCs co-cultured with pMSCs was decreased. Furthermore, pMSC co-culture with mDCs decreased the secretion of IL-12 and INF-γ whilst increasing the secretion of IL-10 in a T cell proliferation experiment. Finally, pMSC co-culture with iDCs induced the phagocytic activity of iDCs. CONCLUSIONS: We have shown that pMSCs have an inhibitory effect on the differentiation, maturation and function of DCs, as well as on the proliferation of T cells, suggesting that pMSCs can control the immune responses at multiple levels.


Subject(s)
Cell Differentiation/genetics , Dendritic Cells/cytology , Mesenchymal Stem Cells/cytology , Monocytes/cytology , Antigens, CD1/metabolism , Cell Differentiation/drug effects , Cell Proliferation/genetics , Chorionic Villi/metabolism , Coculture Techniques , Dendritic Cells/metabolism , Female , Gene Expression Regulation, Developmental , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Humans , Interleukin-4/administration & dosage , Mesenchymal Stem Cells/metabolism , Monocytes/metabolism , Placenta/cytology , Placenta/metabolism , Pregnancy
18.
Placenta ; 35(2): 117-24, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24331737

ABSTRACT

INTRODUCTION: Pre-eclampsia (PE) has a familial association, with daughters of women who had PE during pregnancy having more than twice the risk of developing PE themselves. Through genome-wide linkage and genetic association studies in PE-affected families and large population samples, we previously identified the following as positional candidate maternal susceptibility genes for PE; ACVR1, INHA, INHBB, ERAP1, ERAP2, LNPEP, COL4A1 and COL4A2. The aims of this study were to determine mRNA expression levels of previously identified candidate maternal pre-eclampsia susceptibility genes from normotensive and severe PE (SPE) pregnancies and correlate mRNA expression levels with the clinical severity of SPE. METHODS: Third trimester decidual tissues were collected from both normotensive (n = 21) and SPE pregnancies (n = 24) and mRNA expression levels were determined by real-time PCR. Gene expression was then correlated with several parameters of clinical severity in SPE. Statistical significance was determined by Mann-Whitney U test and Spearman's Correlation. RESULTS: The data demonstrate significantly increased decidual mRNA expression levels of ACVR1, INHBB, ERAP1, ERAP2, LNPEP, COL4A1 and COL4A2 in SPE (p < 0.05). Increased mRNA expression levels of several genes - INHA, INHBB, COL4A1 and COL4A2 were correlated with earlier onset of PE and earlier delivery of the fetus (p < 0.05). CONCLUSION: These results suggest altered expression of maternal susceptibility genes may play roles in PE development and the course of disease severity.


Subject(s)
Decidua/metabolism , Genetic Predisposition to Disease/genetics , Inhibin-beta Subunits/biosynthesis , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , RNA, Messenger/metabolism , Activin Receptors, Type I/biosynthesis , Adult , Collagen Type IV/biosynthesis , Female , Humans , Inhibins/biosynthesis , Pregnancy , Pregnancy Trimester, Third
19.
Placenta ; 35 Suppl: S4-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24321782

ABSTRACT

Workshops are an important part of the IFPA annual meeting as they allow for discussion of specialized topics. At IFPA meeting 2013 there were twelve themed workshops, four of which are summarized in this report. These workshops related to various aspects of placental biology but collectively covered areas of pregnancy pathologies and placental metabolism: 1) diabetes in pregnancy; 2) lipids, fatty acids and the placenta; 3) oxygen in placental development and pathologies; 4) stem cells and pathologies.


Subject(s)
Diabetes, Gestational/metabolism , Dyslipidemias/physiopathology , Oxygen/physiology , Placentation , Animals , Female , Fetal Development , Humans , Pregnancy , Signal Transduction , Stem Cells/physiology
20.
Stem Cell Rev Rep ; 9(5): 620-41, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23812784

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) have a therapeutic potential in tissue repair because of capacity for multipotent differentiation and their ability to modulate the immune response. In this study, we examined the ability of human placental MSCs (pMSCs) to modify the differentiation of human monocytes into macrophages and assessed the influence of pMSCs on important macrophage functions. METHODS: We used GM-CSF to stimulate the differentiation of monocytes into the M1 macrophage pathway and then co-cultured these cells with pMSCs in the early stages of macrophage differentiation. We then evaluated the effect on differentiation by microscopic examination and by quantification of molecules important in the differentiation and immune functions of macrophages using flow cytometry and ELISA. The mechanism by which pMSCs could mediate their effects on macrophage differentiation was also studied. RESULTS: The co-culture of pMSCs with monocytes stimulated to follow the inflammatory M1 macrophage differentiation pathway resulted in a shift to anti-inflammatory M2-like macrophage differentiation. This transition was characterized by morphological of changes typical of M2 macrophages, and by changes in cell surface marker expression including CD14, CD36, CD163, CD204, CD206, B7-H4 and CD11b, which are distinctive of M2 macrophages. Co-culture with pMSCs reduced the expression of the costimulatory molecules (CD40, CD80 and CD86) and increased the expression of co-inhibitory molecules (CD273, CD274 and B7-H4) as well as the surface expression of major histocompatibility complex (MHC-II) molecules. Furthermore, the secretion of IL-10 was increased while the secretion of IL-1ß, IL-12 (p70) and MIP-1α was decreased; a profile typical of M2 macrophages. Finally, pMSCs induced the phagocytic activity and the phagocytosis of apoptotic cells associated with M2- like macrophages; again a profile typical of M2 macrophages. We found that the immunoregulatory effect of pMSCs on macrophage differentiation was mediated by soluble molecules acting partially via glucocorticoid and progesterone receptors. CONCLUSIONS: We have shown that pMSCs can transition macrophages from an inflammatory M1 into an anti-inflammatory M2 phenotype. Our findings suggest a new immunosuppressive property of pMSCs that may be employed in the resolution of inflammation associated with inflammatory diseases and in tissue repair.


Subject(s)
Cell Differentiation/immunology , Macrophages/immunology , Mesenchymal Stem Cells/immunology , Monocytes/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , Apoptosis/drug effects , Apoptosis/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Coculture Techniques , Cytochalasin B/immunology , Cytochalasin B/pharmacology , Cytokines/immunology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Macrophages/cytology , Macrophages/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Monocytes/cytology , Monocytes/metabolism , Phagocytosis/drug effects , Phagocytosis/immunology , Placenta/cytology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...