Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
J Biomed Sci ; 31(1): 49, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38735943

ABSTRACT

BACKGROUND: The impact of global overconsumption of simple sugars on bone health, which peaks in adolescence/early adulthood and correlates with osteoporosis (OP) and fracture risk decades, is unclear. Mesenchymal stromal/stem cells (MSCs) are the progenitors of osteoblasts/bone-forming cells, and known to decrease their osteogenic differentiation capacity with age. Alarmingly, while there is correlative evidence that adolescents consuming greatest amounts of simple sugars have the lowest bone mass, there is no mechanistic understanding on the causality of this correlation. METHODS: Bioinformatics analyses for energetics pathways involved during MSC differentiation using human cell information was performed. In vitro dissection of normal versus high glucose (HG) conditions on osteo-/adipo-lineage commitment and mitochondrial function was assessed using multi-sources of non-senescent human and murine MSCs; for in vivo validation, young mice was fed normal or HG-added water with subsequent analyses of bone marrow CD45- MSCs. RESULTS: Bioinformatics analyses revealed mitochondrial and glucose-related metabolic pathways as integral to MSC osteo-/adipo-lineage commitment. Functionally, in vitro HG alone without differentiation induction decreased both MSC mitochondrial activity and osteogenesis while enhancing adipogenesis by 8 h' time due to depletion of nicotinamide adenine dinucleotide (NAD+), a vital mitochondrial co-enzyme and co-factor to Sirtuin (SIRT) 1, a longevity gene also involved in osteogenesis. In vivo, HG intake in young mice depleted MSC NAD+, with oral NAD+ precursor supplementation rapidly reversing both mitochondrial decline and osteo-/adipo-commitment in a SIRT1-dependent fashion within 1 ~ 5 days. CONCLUSIONS: We found a surprisingly rapid impact of excessive glucose, a single dietary factor, on MSC SIRT1 function and osteogenesis in youthful settings, and the crucial role of NAD+-a single molecule-on both MSC mitochondrial function and lineage commitment. These findings have strong implications on future global OP and disability risks in light of current worldwide overconsumption of simple sugars.


Subject(s)
Glucose , Mesenchymal Stem Cells , Mitochondria , NAD , Osteogenesis , Sirtuin 1 , Mesenchymal Stem Cells/metabolism , Sirtuin 1/metabolism , Sirtuin 1/genetics , Osteogenesis/physiology , Mice , Humans , Animals , Mitochondria/metabolism , Glucose/metabolism , NAD/metabolism , Cell Differentiation
3.
STAR Protoc ; 4(4): 102728, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37979177

ABSTRACT

To better implement mesenchymal stem cell (MSC)-based therapy toward cartilage diseases, a more efficient and less off-target chondrogenesis protocol is needed. Here, we present a protocol to induce human MSC chondrogenesis via Wnt antagonism. We describe steps for pellet formation, Wnt antagonism-based chondrogenic induction, and refreshing the differentiation medium. We detail procedures for characterizing MSC chondrogenesis. By using Wnt antagonism instead of conventional transforming growth factor ß-based induction, this protocol avoids the potential for induction of chondrocyte hypertrophy/osteogenesis or other lineages. For complete details on the use and execution of this protocol, please refer to Hsieh et al. (2023).1.


Subject(s)
Chondrogenesis , Transforming Growth Factor beta , Humans , Transforming Growth Factor beta/metabolism , Cell Differentiation , Immunologic Factors
4.
Cells ; 12(16)2023 08 11.
Article in English | MEDLINE | ID: mdl-37626861

ABSTRACT

Three-dimensional (3D) in vitro spheroid/organoid culture increasingly appears to better mimic physiological states than standard 2D systems. The biological consequence of 3D spheroids, however, differs for different cell types: for pluripotent embryonic stem cells (ESCs), differentiation and loss of stemness occur, while the converse is true for somatic and cancer cells. Despite such diverse consequences, there are likely conserved mechanisms governing 3D spheroid formation across cell types that are unknown but could be efficiently targeted for translational application. To elucidate such processes, we performed transcriptome analysis with functional validation on 2D- and 3D-cultured mouse ESCs, mesenchymal stromal/stem cells (MSCs), and cancer cells. At both the transcriptomic and functional levels, 3D spheroid formation resulted in commitment towards known cell-specific functional outcomes. Surprisingly in all cell types, downregulation of the cholesterol synthesis pathway was found during 3D spheroid formation, with modulation concomitantly affecting 3D spheroid formation and cell-specific consequences; similar results were seen with human cell types. Furthermore, improved antioxidant capacity after 3D spheroid formation across cell types was further enhanced with modulation of the pathway. These findings demonstrate the profound cell-specific consequences and the translational value of understanding conserved mechanisms across diverse cell types after 3D spheroid formation.


Subject(s)
Antioxidants , Embryonic Stem Cells , Humans , Animals , Mice , Antioxidants/pharmacology , Down-Regulation , Cell Differentiation , Gene Expression Profiling
5.
Stem Cells Transl Med ; 12(5): 235-244, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37184894

ABSTRACT

As invaluable as the standard 2-dimensional (2D) monolayer in vitro cell culture system has been, there is increasing evidence that 3-dimensional (3D) non-adherent conditions are more relevant to the in vivo condition. While one of the criteria for human mesenchymal stem cells (MSCs) has been in vitro plastic adherence, such 2D culture conditions are not representative of in vivo cell-cell and cell-extracellular matrix (ECM) interactions, which may be especially important for this progenitor/stem cell of skeletal and connective tissues. The 3D spheroid, a multicellular aggregate formed under non-adherent 3D in vitro conditions, may be particularly suited as an in vitro method to better understand MSC physiological processes, since expression of ECM and other adhesion proteins are upregulated in such a cell culture system. First used in embryonic stem cell in vitro culture to recapitulate in vivo developmental processes, 3D spheroid culture has grown in popularity as an in vitro method to mimic the 3-dimensionality of the native niche for MSCs within tissues/organs. In this review, we discuss the relevance of the 3D spheroid culture for understanding MSC biology, summarize the biological outcomes reported in the literature based on such this culture condition, as well as contemplate limitations and future considerations in this rapidly evolving and exciting area.


Subject(s)
Mesenchymal Stem Cells , Humans , Stem Cells , Spheroids, Cellular , Cell Differentiation/physiology
6.
iScience ; 26(1): 105713, 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36582823

ABSTRACT

Human mesenchymal stem cells (MSCs) remain one of the best cell sources for cartilage, a tissue without regenerative capacity. However, MSC chondrogenesis is commonly induced through TGFß, a pleomorphic growth factor without specificity for this lineage. Using tissue- and induced pluripotent stem cell-derived MSCs, we demonstrate an efficient and precise approach to induce chondrogenesis through Wnt/ß-catenin antagonism alone without TGFß. Compared to TGFß, Wnt/ß-catenin antagonism more rapidly induced MSC chondrogenesis without eliciting off-target lineage specification toward smooth muscle or hypertrophy; this was mediated through increasing N-cadherin levels and ß-catenin interactions-key components of the adherens junctions (AJ)-and increasing cytoskeleton-mediated condensation. Validation with transcriptomic analysis of human chondrocytes compared to MSCs and osteoblasts showed significant downregulation of Wnt/ß-catenin and TGFß signaling along with upregulation of α-catenin as an upstream regulator. Our findings underscore the importance of understanding developmental pathways and structural modifications in achieving efficient MSC chondrogenesis for translational application.

7.
Opt Lett ; 46(24): 6039-6042, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34913913

ABSTRACT

A platinum (Pt) nanolayer was successfully deposited on indium titanium oxide (ITO) as a buffer layer for polymer light-emitting diodes (PLEDs) using a rapid and low-cost sputtering system at room temperature. With a 5 s Pt-dispersed ITO as the anode window substrate of the PLED, a maximum current efficiency of 4.00 cd/A was realized, which is notably higher than that of a typical PLED (1.13 cd/A). It was determined that the average current efficiency and electroluminescence intensity of the proposed PLED were enhanced by 2.5 times and 290%, respectively, compared to a typical PLED.

8.
Stem Cells Transl Med ; 10(5): 711-724, 2021 05.
Article in English | MEDLINE | ID: mdl-33506633

ABSTRACT

Multipotent human mesenchymal stromal cells (MSCs) from multiple organs including the bone marrow (BM) and placenta harbor clinically relevant immunomodulation best demonstrated toward T lymphocytes. Surprisingly, there is limited knowledge on interactions with B lymphocytes, which originate from the BM where there is a resident MSC. With increasing data demonstrating MSC tissue-specific propensities impacting therapeutic outcome, we therefore investigated the interactions of BM-MSCs-its resident and "niche" MSC-and placental MSCs (P-MSCs), another source of MSCs with well-characterized immunomodulatory properties, on the global functional outcomes of pan-peripheral B cell populations. We found that P-MSCs but not BM-MSCs significantly inhibit proliferation and further differentiation of stimulated human peripheral B populations in vitro. Moreover, although BM-MSCs preserve multiple IL-10-producing regulatory B cell (Breg) subsets, P-MSCs significantly increase all subsets. To corroborate these in vitro findings in vivo, we used a mouse model of B-cell activation and found that adoptive transfer of P-MSCs but not BM-MSCs significantly decreased activated B220+ B cells. Moreover, adoptive transfer of P-MSCs but not BM-MSCs significantly decreased the overall B220+ B-cell proliferation and further differentiation, similar to the in vitro findings. P-MSCs also increased two populations of IL-10-producing murine Bregs more strongly than BM-MSCs. Transcriptome analyses demonstrated multifactorial differences between BM- and P-MSCs in the profile of relevant factors involved in B lymphocyte proliferation and differentiation. Our results highlight the divergent outcomes of tissue-specific MSCs interactions with peripheral B cells, and demonstrate the importance of understanding tissue-specific differences to achieve more efficacious outcome with MSC therapy.


Subject(s)
B-Lymphocytes , Mesenchymal Stem Cells , Pluripotent Stem Cells , Animals , B-Lymphocytes/cytology , B-Lymphocytes, Regulatory , Bone Marrow Cells , Cell Communication , Cell Differentiation , Cell Proliferation , Female , Interleukin-10 , Mesenchymal Stem Cells/classification , Mesenchymal Stem Cells/cytology , Mice , Placenta/cytology , Pluripotent Stem Cells/cytology , Pregnancy
9.
Int J Mol Sci ; 21(14)2020 Jul 18.
Article in English | MEDLINE | ID: mdl-32708387

ABSTRACT

Multipotent human mesenchymal stem cells (MSCs) harbor clinically relevant immunomodulation, and HLA-G, a non-classical MHC class I molecule with highly restricted tissue expression, is one important molecule involved in these processes. Understanding of the natural regulatory mechanisms involved in expression of this elusive molecule has been difficult, with near exclusive reliance on cancer cell lines. We therefore studied the transcriptional control of HLA-G in primary isolated human bone marrow- (BM), human embryonic stem cell-derived (hE-), as well as placenta-derived MSCs (P-MSCs), and found that all 3 types of MSCs express 3 of the 7 HLA-G isoforms at the gene level; however, fibroblasts did not express HLA-G. Protein validation using BM- and P-MSCs demonstrated expression of 2 isoforms including a larger HLA-G-like protein. Interferon-γ (IFN-γ) stimulation upregulated both gene and protein expression in MSCs but not the constitutively expressing JEG-3 cell line. Most interestingly in human MSCs and placental tissue, hypomethylation of CpG islands not only occurs on the HLA-G proximal promoter but also on the gene body as well, a pattern not seen in either of the 2 commonly used choriocarcinoma cell lines which may contribute to the unique HLA-G expression patterns and IFN-γ-responsiveness in MSCs. Our study implicates the importance of using normal cells and tissues for physiologic understanding of tissue-specific transcriptional regulation, and highlight the utility of human MSCs in unraveling the transcriptional regulation of HLA-G for better therapeutic application.


Subject(s)
Bone Marrow Cells/metabolism , DNA Methylation/genetics , DNA/metabolism , Embryonic Stem Cells/metabolism , HLA-G Antigens/metabolism , Mesenchymal Stem Cells/metabolism , Placenta/cytology , Azacitidine/pharmacology , Cell Line, Tumor , CpG Islands , DNA Methylation/drug effects , Demethylation/drug effects , Female , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HLA-G Antigens/genetics , Humans , Interferon-gamma/pharmacology , Placenta/metabolism , Pregnancy , Promoter Regions, Genetic , Protein Isoforms , Tandem Mass Spectrometry
10.
Stem Cells ; 36(6): 903-914, 2018 06.
Article in English | MEDLINE | ID: mdl-29396902

ABSTRACT

Multilineage tissue-source mesenchymal stem cells (MSCs) possess strong immunomodulatory properties and are excellent therapeutic agents, but require constant isolation from donors to combat replicative senescence. The differentiation of human induced pluripotent stem cells (iPSCs) into MSCs offers a renewable source of MSCs; however, reports on their immunomodulatory capacity have been discrepant. Using MSCs differentiated from iPSCs reprogrammed using diverse cell types and protocols, and in comparison to human embryonic stem cell (ESC)-MSCs and bone marrow (BM)-MSCs, we performed transcriptome analyses and assessed for functional immunomodulatory properties. Differentiation of MSCs from iPSCs results in decreased c-Myc expression and its downstream pathway along with a concomitant downregulation in the DNA replication pathway. All four lines of iPSC-MSCs can significantly suppress in vitro activated human peripheral blood mononuclear cell (PBMC) proliferation to a similar degree as ESC-MSCs and BM-MSCs, and modulate CD4 T lymphocyte fate from a type 1 helper T cell (Th1) and IL-17A-expressing (Th17) cell fate to a regulatory T cell (Treg) phenotype. Moreover, iPSC-MSCs significantly suppress cytotoxic CD8 T proliferation, activation, and differentiation into type 1 cytotoxic T (Tc1) and IL-17-expressing CD8 T (Tc17) cells. Coculture of activated PBMCs with human iPSC-MSCs results in an overall shift of secreted cytokine profile from a pro-inflammatory environment to a more immunotolerant milieu. iPSC-MSC immunomodulation was also validated in vivo in a mouse model of induced inflammation. These findings support that iPSC-MSCs possess low oncogenicity and strong immunomodulatory properties regardless of cell-of-origin or reprogramming method and are good potential candidates for therapeutic use. Stem Cells 2018;36:903-914.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Differentiation , Down-Regulation , Humans , Immunomodulation , Mice
11.
Oncotarget ; 7(28): 43949-43959, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27304057

ABSTRACT

Mesenchymal stem cells (MSCs) are paraxial mesodermal progenitors with potent immunomodulatory properties. Reports also indicate that MSCs can undergo neural-like differentiation, offering hope for use in neurodegenerative diseases. However, ex vivo expansion of these rare somatic stem cells for clinical use leads to cellular senescence. A newer source of MSCs derived from human pluripotent stem cells (PSC) can offer the 'best-of-both-worlds' scenario, abrogating the concern of teratoma formation while preserving PSC proliferative capacity. PSC-derived MSCs (PSC-MSCs) also represent MSCs at the earliest developmental stage, and we found that these MSCs harbor stronger neuro-differentiation capacity than post-natal MSCs. PSC-MSCs express higher levels of neural stem cell (NSC)-related genes and transcription factors than adult bone marrow MSCs at baseline, and rapidly differentiate into neural-like cells when cultured in either standard neurogenic differentiation medium (NDM) or when the cytoskeletal modulator RhoA kinase (ROCK) is inhibited. Interestingly, when NDM is combined with ROCK inhibition, PSC-MSCs undergo further commitment, acquiring characteristics of post-mitotic neurons including nuclear condensation, extensive dendritic growth, and neuron-restricted marker expression including NeuN, ß-III-tubulin and Doublecortin. Our data demonstrates that PSC-MSCs have potent capacity to undergo neural differentiation and also implicate the important role of the cytoskeleton in neural lineage commitment.


Subject(s)
Cytoskeleton , Mesenchymal Stem Cells/cytology , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Cell Differentiation/physiology , Cell Lineage , Cytoskeleton/enzymology , Humans , rho-Associated Kinases/metabolism
12.
Stem Cell Reports ; 5(3): 392-404, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26321145

ABSTRACT

Multipotent human mesenchymal stromal cells (hMSCs) harbor immunomodulatory properties that are therapeutically relevant. One of the most clinically important populations of leukocytes is the interleukin-17A (IL-17A)-secreting T (Th17) lymphocytes. However, mechanisms of hMSC and Th17 cell interactions are incompletely resolved. We found that, along with Th1 responses, hMSCs strongly suppressed Th17 responses and this required both IL-25--also known as IL--17E-as well as programmed death ligand-1 (PD-L1), a potent cell surface ligand for tolerance induction. Knockdown of IL-25 expression in hMSCs abrogated Th17 suppression in vitro and in vivo. However, IL-25 alone was insufficient to significantly suppress Th17 responses, which also required surface PD-L1 expression. Critically, IL-25 upregulated PD-L1 surface expression through the signaling pathways of JNK and STAT3, with STAT3 found to constitutively occupy the proximal region of the PD-L1 promoter. Our findings demonstrate the complexities of hMSC-mediated Th17 suppression, and highlight the IL-25/STAT3/PD-L1 axis as a candidate therapeutic target.


Subject(s)
B7-H1 Antigen/immunology , Interleukin-17/immunology , Mesenchymal Stem Cells/immunology , STAT3 Transcription Factor/immunology , Th17 Cells/immunology , Transcription, Genetic/immunology , B7-H1 Antigen/genetics , Gene Expression Regulation/immunology , Gene Knockdown Techniques , Humans , Mesenchymal Stem Cells/cytology , STAT3 Transcription Factor/genetics , Th17 Cells/cytology
13.
J Vis Exp ; (106): e53265, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26780482

ABSTRACT

The immunomodulatory properties of multilineage human mesenchymal stem cells (MSCs) appear to be highly relevant for clinical use towards a wide-range of immune-related diseases. Mechanisms involved are increasingly being elucidated and in this article, we describe the basic experiment to assess MSC immunomodulation by assaying for suppression of effector leukocyte proliferation. Representing activation, leukocyte proliferation can be assessed by a number of techniques, and we describe in this protocol the use of the fluorescent cellular dye carboxyfluorescein succinimidyl ester (CFSE) to label leukocytes with subsequent flow cytometric analyses. This technique can not only assess proliferation without radioactivity, but also the number of cell divisions that have occurred as well as allowing for identification of the specific population of proliferating cells and intracellular cytokine/factor expression. Moreover, the assay can be tailored to evaluate specific populations of effector leukocytes by magnetic bead surface marker selection of single peripheral blood mononuclear cell populations prior to co-culture with MSCs. The flexibility of this co-culture assay is useful for investigating cellular interactions between MSCs and leukocytes.


Subject(s)
Mesenchymal Stem Cells/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/physiology , Cells, Cultured , Coculture Techniques , Cytokines/immunology , Humans , Immunomodulation , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Mesenchymal Stem Cells/cytology
14.
J Leukoc Biol ; 96(2): 295-303, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24714552

ABSTRACT

Monocytes are a population of leukocytes that terminally differentiate into macrophages and DCs. Whereas these differentiated progeny have inflammatory and resident--which are more immunomodulatory--phenotypes, less has been reported on the plasticity of monocytes themselves. We found that MSCs, a population of somatic stem cells, can rapidly induce human and murine monocytes through secretion of HGF to acquire an immunomodulatory phenotype to suppress T cell effector function. MSCs are multilineage postnatal progenitor cells with strong immunomodulatory effects toward T lymphocytes, NK lymphocytes, and DCs, but less is known regarding their interactions with monocytes. We found that CD14(+) human monocytes express c-Met, the receptor for HGF, and both depletion of HGF-treated CD14(+) monocytes and knockdown of HGF secretion in MSCs abrogate the suppression of anti-CD3/28-activated T cell proliferation. HGF-treated monocytes remain undifferentiated and can alter activated T cell cytokine expression from a Th1 toward Th2 profile. Moreover, monocytes cocultured with MSCs or treated with HGF alone can produce high levels of IL-10, a potent immunomodulatory cytokine. Injection of HGF to WT mice also results in an increase in IL-10(+)-expressing monocytes from the spleen, a known reservoir for circulating monocytes. Mechanistically, HGFs modulate IL-10 production in monocytes through the ERK1/2 pathway. Our data demonstrate further the pleomorphic nature of MSC immunomodulation, as well as highlight the important role of immunomodulatory monocytes in altering T cell effector function.


Subject(s)
Hepatocyte Growth Factor/immunology , MAP Kinase Signaling System/immunology , Mesenchymal Stem Cells/immunology , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/immunology , Monocytes/immunology , Animals , Cell Differentiation/immunology , Cell Line , Cell Proliferation , Dendritic Cells/cytology , Dendritic Cells/immunology , Female , Humans , Interleukin-10/immunology , Male , Mesenchymal Stem Cells/cytology , Mice , Monocytes/cytology , Proto-Oncogene Proteins c-met/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology
15.
Stem Cell Reports ; 1(2): 139-51, 2013.
Article in English | MEDLINE | ID: mdl-24052949

ABSTRACT

Mesenchymal stromal cells (MSCs) are multilineage progenitors with immunomodulatory properties, including expansion of immunomodulatory leukocytes such as regulatory T lymphocytes (Tregs) and tolerogenic dendritic cells. We report that human MSCs can expand CD14(-)CD11b(+)CD33(+) human myeloid-derived suppressor cells (MDSCs). MSC-expanded MDSCs suppress allogeneic lymphocyte proliferation, express arginase-1 and inducible nitric oxide synthase, and increase the number of Tregs. This expansion occurs through the secretion of hepatocyte growth factor (HGF), with effects replicated by adding HGF singly and abrogated by HGF knockdown in MSCs. In wild-type mice, the liver, which secretes high levels of HGF, contains high numbers of Gr-1(+)CD11b(+) MDSCs, and injection of HGF into mice significantly increases the number of MDSCs. Expansion of MDSCs by MSC-secreted HGF involves c-Met (its receptor) and downstream phosphorylation of STAT3, a key factor in MDSC expansion. Our data further support the strong immunomodulatory nature of MSCs and demonstrate the role of HGF, a mitogenic molecule, in the expansion of MDSCs.


Subject(s)
Hepatocyte Growth Factor/metabolism , Liver/cytology , Mesenchymal Stem Cells/immunology , Myeloid Cells/immunology , Proto-Oncogene Proteins c-met/metabolism , STAT3 Transcription Factor/metabolism , Animals , Antigens, CD/metabolism , Bone Marrow Cells/metabolism , Cells, Cultured , Female , Gene Knockdown Techniques , Hepatocyte Growth Factor/genetics , Humans , Liver/immunology , Liver/metabolism , Mesenchymal Stem Cells/cytology , Mice , Myeloid Cells/metabolism , Placenta/cytology , Pregnancy , T-Lymphocytes, Regulatory/metabolism
16.
J Nanosci Nanotechnol ; 13(4): 2538-48, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23763127

ABSTRACT

The decomposition of CO2 over oxygen deficient nickel ferrite nanoparticles (NFNs) and zinc ferrite nanoparticles (ZFNs) at 573 K was studied. The oxidation states with fine structure of Fe/Ni or Fe/Zn species were also measured in NFNs and ZFNs catalysts, respectively. Oxygen deficiency of catalysts was obtained by reduction in hydrogen. Decomposition of CO2 into carbon and oxygen has been carried out within few minutes when it comes into contact with oxygen deficient catalysts through incorporation of oxygen into ferrite nanoparticles. Oxygen and carbon rather than CO were produced in the decomposition process. The complete decomposition of CO2 was possible because of higher degree of oxygen deficiency andsurface-to-volume ratio of the catalysts. The pre-edge XANES spectra of Fe species in both catalysts exhibit an absorbance feature at 7114 eV for the 1s to 3d transition which is forbidden by the selection rule in case of perfect octahedral symmetry. The EXAFS data showed that the NFNs had two central Fe atoms coordinated by primarily Fe-O and Fe-Fe with bond distances of 1.871 and 3.051 angstroms, respectively. In case of ZFNs these values are 1.889 and 3.062 A, respectively. Methane gas was produced during the reactivation of NFNs by flowing hydrogen gas. Decomposition of CO2, moreover, recovery of valuable methane using heat energy of offgas produced from power generation plant or steel industry is an appealing alternative for energy recovery.

17.
J Nanosci Nanotechnol ; 13(4): 2675-81, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23763143

ABSTRACT

The physical and chemical properties of porous zero-valent iron nanoparticles (ZVINs) have highly been acknowledged in the decontamination of heavy metal containing wastes and groundwater. In the present work, the treatment of Cr-contaminant through adsorption onto the ZVINs has been studied. The morphology, crystal structure, and surface composition of Fe(O) nanoparticles were investigated by field emission scanning electron microscopy/energy dispersive X-ray spectroscopy (FE-SEM/EDS), transmission electron microscope (TEM), X-ray diffraction (XRD), and X-ray photoelectron spectroscopy (XPS), respectively. X-ray absorption near edge structure (XANES) revealed that the Cr(VI) species reduce to Cr(III) while oxidizing the ZVINs to Fe2O3, Fe3O4 or FeO electrochemically. Furthermore, the nitrogen adsorption-desorption isotherm of the porous nanoparticles was similar to a type IV curve with an obvious mesopore-characteristic H3 hysteresis loop, whereas the sizes of mesopores were in the range of 30-50 nm. Experimentally, the efficiency for Cr(VI) removal in the range of 150-300 ppm was found to be > 99.9%. Remarkably, the reduction reaction was completed within 10 min in the absence of additional pH controls. This work also highlights the utility of X-ray absorption spectroscopy (XAS) coupled with conventional characterization methods to comprehensively study the speciation and possible reaction pathway in adsorption process.

18.
Cell Transplant ; 20(11-12): 1721-30, 2011.
Article in English | MEDLINE | ID: mdl-21669042

ABSTRACT

Interactions between maternal natural killer lymphocytes (NKs) and fetal tissues are important in mediating maternal-fetal tolerance. We therefore investigated the interactions of NKs to placenta-derived multipotent cells (PDMCs) isolated from the term human placenta. PDMCs have similar cell surface marker expression as bone marrow mesenchymal stem cells (BMMSCs) and additionally express human embryonic stem cell markers SSEA-4 and CD-9. Differentiation into the tri-mesodermal lineages of osteoblastic, adipocytic, and chondrogenic phenotypes can be readily achieved under the appropriate conditions. We found that PDMCs are more resistant to NK-mediated lysis than the major histocompatibility complex (MHC) class-I null target cell K562, and can suppress NK secretion of interferon-γ (IFN-γ). Moreover, as third-party cells, PDMCs suppressed the cytotoxic effects of cytokine-stimulated NKs on K562. Pretreatment of PDMCs with IFN-γ, a proinflammatory cytokine, surprisingly enhanced such immunosuppressive effects. Cell-cell contact between NKs and PDMCs is required for suppressive effects, which are partially mediated by slight upregulation of the NK inhibitory receptor killer inhibitory receptor and downregulation of the activating receptor NKp30. Moreover, enhancement of PDMC suppressive effects is also mediated by IFN-γ-induced surface expression of HLA-G--an immunomodulatory nonclassical MHC class I molecule--on PDMCs, as seen by partial reversibility with HLA-G neutralizing antibodies. With its broad immunosuppressive properties, PDMCs may represent a potential cell source for therapeutic use.


Subject(s)
HLA-G Antigens/metabolism , Killer Cells, Natural/immunology , Multipotent Stem Cells/metabolism , Placenta/cytology , Cell Differentiation , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Female , HLA-G Antigens/immunology , Humans , Immunosuppression Therapy , Interferon-gamma/pharmacology , Interleukin-2/pharmacology , K562 Cells , Multipotent Stem Cells/cytology , Multipotent Stem Cells/immunology , Natural Cytotoxicity Triggering Receptor 3/metabolism , Pregnancy , Receptors, Cell Surface/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...