Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Intervalo de año de publicación
1.
Tissue Eng Regen Med ; 16(6): 615-630, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31824824

RESUMEN

Background: Mesenchymal Stem/Stromal Cells (MSCs) from the decidua parietalis (DPMSCs) of human term placenta express several molecules with important biological and immunological properties. DPMSCs induce natural killer cell expression of inflammatory receptors and their cytotoxic activity against cancer cells. These properties make DPMSCs promising therapeutical agent for cancer. The successful development of MSCs as an anti-cancer therapeutic cells rely on their ability to function in a hostile inflammatory and oxidative stress cancer environment. Here, we studied the effects of conditioned medium obtained from the culture of breast cancer cells (CMMDA-231) on the functional and phenotypic properties of DPMSCs. Methods: DPMSCs were cultured with CMMDA-231 and important functions of DPMSCs were measured. The effect of CMMDA-231 on DPMSC expression of several genes with different functions was also evaluated. Results: DPMSCs were able to function in response to CMMDA-231, but with reduced proliferative and adhesive potentials. Preconditioning of DPMSCs with CMMDA-231 enhanced their adhesion while reducing their invasion. In addition, CMMDA-231 modulated DPMSC expression of many genes with various functional (i.e., proliferation, adhesion, and invasion) properties. DPMSCs also showed increased expression of genes with anti-cancer property. Conclusion: These data show the ability of DPMSCs to survive and function in cancer environment. In addition, preconditioning of DPMSCs with CMMDA-231 enhanced their anti-cancer properties and thus demonstrating their potential as an anti-cancer therapeutic agent. However, future studies are essential to reveal the mechanism underlying the effects of MDA-231 on DPMSC functional activities and also to confirm the anti-cancer therapeutic potential of DPMSCs.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados/química , Decidua/citología , Femenino , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Embarazo
2.
Stem Cell Res Ther ; 10(1): 50, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30728068

RESUMEN

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.


Asunto(s)
Vellosidades Coriónicas/metabolismo , Células Asesinas Naturales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Diferenciación Celular , Proliferación Celular , Humanos
3.
Stem Cell Res Ther ; 9(1): 275, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30359307

RESUMEN

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.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Monocitos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Adulto , Antígenos CD/genética , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Decidua/citología , Decidua/metabolismo , Femenino , Expresión Génica , Glutatión Reductasa/genética , Glutatión Reductasa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Monocitos/citología , Embarazo , Reductasa de Tiorredoxina-Disulfuro/genética , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Cordón Umbilical/citología , Cordón Umbilical/metabolismo
4.
Stem Cell Res Ther ; 9(1): 238, 2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-30241570

RESUMEN

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.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Apoptosis/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quimiocinas/genética , Quimiocinas/metabolismo , Vellosidades Coriónicas/metabolismo , Técnicas de Cocultivo , Colágeno/química , Colágeno/genética , Colágeno/metabolismo , Combinación de Medicamentos , Endotelinas/genética , Endotelinas/metabolismo , Femenino , Glucosa/antagonistas & inhibidores , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Interleucinas/genética , Interleucinas/metabolismo , Laminina/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Células Madre Mesenquimatosas/citología , Embarazo , Proteoglicanos/química , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Células THP-1 , Cordón Umbilical/citología , Cordón Umbilical/metabolismo
5.
Stem Cell Res Ther ; 9(1): 102, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29650045

RESUMEN

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.


Asunto(s)
Decidua/metabolismo , Células Asesinas Naturales/inmunología , Células Madre Mesenquimatosas/inmunología , Femenino , Humanos , Masculino
6.
Placenta ; 59: 74-86, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28502524

RESUMEN

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.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Células Endoteliales/fisiología , Células Madre Mesenquimatosas/fisiología , Estrés Oxidativo , Placenta/citología , Adhesión Celular , Movimiento Celular , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Peróxido de Hidrógeno , Embarazo
7.
Placenta ; 59: 87-95, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28411943

RESUMEN

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.


Asunto(s)
Tolerancia Inmunológica , Células Madre Mesenquimatosas/inmunología , Placenta/citología , Ensayos Clínicos como Asunto , Femenino , Humanos , Inmunoterapia , Linfocitos/fisiología , Trasplante de Células Madre Mesenquimatosas , Embarazo
8.
Stem Cells Int ; 2016: 5184601, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27087815

RESUMEN

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.

9.
Placenta ; 36(3): 312-21, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25575436

RESUMEN

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.


Asunto(s)
Arteriolas/citología , Decidua/citología , Células Madre Mesenquimatosas/citología , Placentación , Nicho de Células Madre , Remodelación Vascular , Adulto , Antígenos de Superficie/metabolismo , Arteriolas/metabolismo , Biomarcadores/metabolismo , Células Cultivadas , Decidua/metabolismo , Femenino , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Células Madre Mesenquimatosas/metabolismo , Embarazo
10.
Stem Cell Rev Rep ; 11(3): 423-41, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25287760

RESUMEN

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.


Asunto(s)
Diferenciación Celular/genética , Células Dendríticas/citología , Células Madre Mesenquimatosas/citología , Monocitos/citología , Antígenos CD1/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/genética , Vellosidades Coriónicas/metabolismo , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Humanos , Interleucina-4/administración & dosificación , Células Madre Mesenquimatosas/metabolismo , Monocitos/metabolismo , Placenta/citología , Placenta/metabolismo , Embarazo
11.
Stem Cell Rev Rep ; 9(5): 620-41, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23812784

RESUMEN

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.


Asunto(s)
Diferenciación Celular/inmunología , Macrófagos/inmunología , Células Madre Mesenquimatosas/inmunología , Monocitos/inmunología , Antígenos CD/inmunología , Antígenos CD/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Citocalasina B/inmunología , Citocalasina B/farmacología , Citocinas/inmunología , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Macrófagos/citología , Macrófagos/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Monocitos/citología , Monocitos/metabolismo , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Placenta/citología , Embarazo
12.
Stem Cell Rev Rep ; 9(1): 16-31, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22628114

RESUMEN

BACKGROUND: Bone marrow derived mesenchymal stem cells (BM-MSCs) are used extensively in transplantation but their use is associated with many problems including low abundance in BM, low overall number, decreased differentiation potential with age and the invasive isolation procedures needed to obtain BM. We report a novel method of isolating placental MSCs (pMSCs) from chorionic villi, which exhibit the phenotypic and functional characteristics that will make them an attractive source of MSCs for cell-based therapy. METHODS: A novel explant approach was used to isolate pMSCs from chorionic villi of human placentae. These pMSCs were characterized by flow cytometry and were differentiated into adipocytes, osteocytes and chondrocytes using differentiation medium as demonstrated by cytochemical staining. The gene and protein expression profiles of pMSCs were also characterized using real time polymerase chain reaction (PCR) and flow cytometry, respectively. In addition, cytokine secretion by pMSCs was also analysed using sandwich enzyme-linked immunosorbent assay (ELISA) technique. Moreover, the migration and proliferation potentials of pMSCs were also determined. RESULTS: pMSCs were isolated from fetal part of the chorionic villi and these pMSCs expressed CD44, CD90, CD105, CD146, CD166 and HLA-ABC but not CD14, CD19, CD40, CD45, CD80, CD83, CD86 and HLA-DR. In addition, these pMSCs differentiated into osteocytes, chondrocytes and adipocytes and they also expressed several adhesion molecules, chemokines/receptors, growth factor receptors and cytokines/receptors. Moreover, they secreted many cytokines (IL-1Ra, IL6, IL8, IL10, IL11 and IL15) and they were able to proliferate. Furthermore, they migrated in response to chemotactic factors including stromal cell-derived factor-1 (SDF-1), platelet derived growth factor (PDGF), hepatocyte growth factor (HGF), and monocyte chemotactic protein-1 (MCP-1). CONCLUSIONS: We devised a novel explant method of isolating pMSCs that expressed many biological factors responsible for mediating cellular processes such as migration/homing, immune modulation and angiogenesis. Therefore, we suggest that pMSCs prepared from human term placental chorionic villous explants are an attractive source of MSCs for cell therapy.


Asunto(s)
Vellosidades Coriónicas/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Placenta/citología , Adipocitos/metabolismo , Antígenos CD/biosíntesis , Moléculas de Adhesión Celular/biosíntesis , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Quimiocinas/biosíntesis , Condrocitos/metabolismo , Citocinas/biosíntesis , Femenino , Citometría de Flujo , Humanos , Osteocitos/metabolismo , Embarazo , Receptores de Quimiocina/biosíntesis , Receptores de Citocinas/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis
13.
J Reprod Immunol ; 94(2): 131-41, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22542910

RESUMEN

Interactions between maternal immune cells and the placenta are of substantial interest since diseases of pregnancy, such as recurrent miscarriage, villitis of unknown etiology and preeclampsia may arise due to inadequate adaptation of the maternal immune system. During normal pregnancy trophoblast debris is shed from the placenta into the maternal blood in large quantities. This trophoblast debris is then rapidly cleared from the maternal circulation. In this study, we exposed trophoblast debris generated from an in vitro placental explant model to peripheral blood-derived macrophages and quantified a variety of molecules that are important in immune responses by ELISA or flow cytometry. Phagocytosis of trophoblast debris resulted in reduced cell-surface expression of MHC-II molecules, the costimulatory molecules (CD80, CD86, CD40 and B7H3), monocyte chemoattractant protein-1 (MCP-1), inter-cellular adhesion molecule 1 (ICAM-1) and IL-8 receptors in macrophages while the expression of programmed death-1 ligand 1 (PD-L1) was upregulated. In addition, phagocytosis of trophoblast debris induced the secretion of the anti-inflammatory cytokines IL-10, IL6 and IL1Ra and decreased the secretion of pro-inflammatory cytokines IL-1ß, IL12p70 and IL-8 by macrophages. Phagocytosis of trophoblast debris also increased macrophage expression of the immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO). We have shown that phagocytosis of trophoblast debris from normal placentae alters the phenotype of macrophages such that they are likely to deviate maternal immune responses towards tolerance and away from inflammation. This may be one of the mechanisms that allow the human fetal allograft to survive in direct contact with the maternal immune system.


Asunto(s)
Estructuras Celulares/inmunología , Feto/inmunología , Macrófagos/inmunología , Tolerancia al Trasplante , Trasplante Homólogo/inmunología , Trofoblastos/inmunología , Antígenos CD/genética , Antígenos CD/metabolismo , Células Cultivadas , Estructuras Celulares/citología , Estructuras Celulares/metabolismo , Citocinas/genética , Citocinas/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Tolerancia Inmunológica , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Mediadores de Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Circulación Placentaria/inmunología , Embarazo , Trofoblastos/citología , Trofoblastos/metabolismo
14.
Reprod Sci ; 19(6): 597-606, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22138542

RESUMEN

The outer layer of the human placenta is the multinucleated syncytiotrophoblast. The syncytiotrophoblast is formed by the fusion of mononuclear cytotrophoblasts, and aged syncytiotrophoblast nuclei are extruded into the maternal blood as membrane-enclosed "syncytial nuclear aggregates" that are then eliminated from the maternal circulation. Apoptosis proteins are hypothesized to be involved in both of these processes, but the mechanism of death in the syncytiotrophoblast is unclear and death processes in this multinucleated layer are likely to differ from related processes in mononuclear cells. We have used a combination of villous explant culture and immunohistochemical staining of semi-serial sections from the explants to study the changing expression of 4 proteins that are markers of apoptotic processes in first-trimester human placentae. These studies show that Bcl-2 expression is limited to the syncytiotrophoblast and syncytial nuclear aggregates, while conversely Bax is expressed in some cytotrophoblasts. Activated caspase 3 and the M30 cytokeratin neoepitope were localized to isolated regions of the syncytiotrophoblast and some syncytial nuclear aggregates but were never present in the same area. Combining our results with those of others, we suggest a refined scheme whereby proteins of the apoptosis cascade participate in both the processes of syncytial formation and death.


Asunto(s)
Apoptosis/fisiología , Trofoblastos/fisiología , Caspasa 3/análisis , Femenino , Humanos , Inmunohistoquímica , Queratinas/análisis , Embarazo , Primer Trimestre del Embarazo , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Técnicas de Cultivo de Tejidos , Trofoblastos/química , Proteína X Asociada a bcl-2/análisis
15.
Mol Hum Reprod ; 12(11): 687-94, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16988002

RESUMEN

Deportation of trophoblast shed from the placenta into the maternal circulation was first described over 100 years ago. Despite this, little is known about the quantity or nature of the shed and deported trophoblasts. Neither do we have a clear understanding of the fate of deported trophoblasts nor do we have a clear understanding of their effects on the maternal physiology. This deficiency is largely due to the inaccessibility of deported trophoblasts in vivo. This study aimed to produce a model that would allow us to study deported trophoblasts. We devised a system for culturing placental explants of 12-week gestation in cell culture inserts with a stainless steel mesh bottom that allowed the ready harvesting of shed/deported trophoblasts. Immunohistochemical and morphologic investigations demonstrated that these in vitro shed/deported trophoblasts are similar to those found in vivo and that apoptotic, necrotic and viable trophoblasts were shed from the explants. Inhibiting caspases induced a change from predominantly apoptotic to predominantly necrotic trophoblast shedding. We have devised an in vitro model that allows the collection of shed/deported trophoblasts which will significantly enhance our ability to study these cells. Our preliminary investigations confirm that apoptosis plays an important role in trophoblast shedding/deportation.


Asunto(s)
Técnicas de Cultivo de Órganos/métodos , Trofoblastos/citología , Apoptosis , Supervivencia Celular , Inhibidores de Cisteína Proteinasa/farmacología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Femenino , Edad Gestacional , Humanos , Leucocitos , Necrosis , Oligopéptidos/farmacología , Técnicas de Cultivo de Órganos/instrumentación , Embarazo , Trofoblastos/efectos de los fármacos
16.
J Reprod Immunol ; 72(1-2): 33-45, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16843534

RESUMEN

During pregnancy, trophoblasts are shed into maternal blood from the placenta as they die. Trophoblasts are fetal cells and are therefore immunologically foreign to the maternal immune system, but the effects of shed trophoblasts on the maternal immune system are poorly characterized. We have used an in vitro villous explant model to harvest shed trophoblasts. These shed trophoblasts consist of multinucleated syncytial knots as well as mononuclear cells, and approximately 90% are apoptotic as determined by immunostaining with antibodies recognizing activated caspase-3 and the M30 cytokeratin neoepitope. U937 cells phagocytosed the shed apoptotic trophoblasts and, subsequently, secretion of the anti-inflammatory cytokine IL-10 was increased. In contrast, secretion of the proinflammatory cytokine Il-1beta by U937 cells was decreased after phagocytosis of apoptotic trophoblasts and the changes in both IL-10 and IL-1beta secretion were blocked by co-incubation with the phagocytosis inhibitor cytochalasin B. Shed trophoblasts caused a significant increase also in expression of the, immunosuppressive, tryptophan-metabolizing enzyme indoleamine 2,3-dioxygenase. We speculate that the shedding of trophoblasts may not be simply a mechanism the fetus uses to dispose of aged trophoblasts but rather shed apoptotic trophoblasts may provide a chronic source of tolerizing paternally derived antigens to regulate maternal immune responses to the fetus.


Asunto(s)
Apoptosis , Macrófagos/inmunología , Fagocitosis , Placenta/inmunología , Trofoblastos/inmunología , Citocalasina B/farmacología , Dioxigenasas/metabolismo , Femenino , Humanos , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Antígenos Comunes de Leucocito/análisis , Fagocitosis/efectos de los fármacos , Placenta/citología , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA