Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Hepatology ; 67(2): 636-650, 2018 02.
Article in English | MEDLINE | ID: mdl-28913935

ABSTRACT

Hepatocellular carcinomas (HCC) contain a subpopulation of cancer stem cells (CSCs), which exhibit stem cell-like features and are responsible for tumor relapse, metastasis, and chemoresistance. The development of effective treatments for HCC will depend on a molecular-level understanding of the specific pathways driving CSC emergence and stemness. MacroH2A1 is a variant of the histone H2A and an epigenetic regulator of stem-cell function, where it promotes differentiation and, conversely, acts as a barrier to somatic-cell reprogramming. Here, we focused on the role played by the histone variant macroH2A1 as a potential epigenetic factor promoting CSC differentiation. In human HCC sections we uncovered a significant correlation between low frequencies of macroH2A1 staining and advanced, aggressive HCC subtypes with poorly differentiated tumor phenotypes. Using HCC cell lines, we found that short hairpin RNA-mediated macroH2A1 knockdown induces acquisition of CSC-like features, including the growth of significantly larger and less differentiated tumors when injected into nude mice. MacroH2A1-depleted HCC cells also exhibited reduced proliferation, resistance to chemotherapeutic agents, and stem-like metabolic changes consistent with enhanced hypoxic responses and increased glycolysis. The loss of macroH2A1 increased expression of a panel of stemness-associated genes and drove hyperactivation of the nuclear factor kappa B p65 pathway. Blocking phosphorylation of nuclear factor kappa B p65 on Ser536 inhibited the emergence of CSC-like features in HCC cells knocked down for macroH2A1. Conclusion: The absence of histone variant macroH2A1 confers a CSC-like phenotype to HCC cells in vitro and in vivo that depends on Ser536 phosphorylation of nuclear factor kappa B p65; this pathway may hold valuable targets for the development of CSC-focused treatments for HCC. (Hepatology 2018;67:636-650).


Subject(s)
Carcinoma, Hepatocellular/pathology , Histones/physiology , Liver Neoplasms/pathology , Neoplastic Stem Cells/pathology , Cell Proliferation , Gene Expression Profiling , Hep G2 Cells , Humans , Phosphorylation , Transcription Factor RelA/metabolism
2.
Front Microbiol ; 8: 1157, 2017.
Article in English | MEDLINE | ID: mdl-28690599

ABSTRACT

A mutual interplay exists between adaptive immune system and gut microbiota. Altered gut microbial ecosystems are associated with the metabolic syndrome, occurring in most obese individuals. However, it is unknown why 10-25% of obese individuals are metabolically healthy, while normal weight individuals can develop inflammation and atherosclerosis. We modeled these specific metabolic conditions in mice fed with a chow diet, an obesogenic but not inflammatory diet-mimicking healthy obesity, or Paigen diet-mimicking inflammation in the lean subjects. We analyzed a range of markers and cytokines in the aorta, heart, abdominal fat, liver and spleen, and metagenomics analyses were performed on stool samples. T lymphocytes infiltration was found in the aorta and in the liver upon both diets, however a significant increase in CD4+ and CD8+ cells was found only in the heart of Paigen-fed animals, paralleled by increased expression of IL-1, IL-4, IL-6, IL-17, and IFN-γ. Bacteroidia, Deltaproteobacteria, and Verrucomicrobia dominated in mice fed Paigen diet, while Gammaproteobacteria, Delataproteobacteria, and Erysipelotrichia were more abundant in obese mice. Mice reproducing human metabolic exceptions displayed gut microbiota phylogenetically distinct from normal diet-fed mice, and correlated with specific adaptive immune responses. Diet composition thus has a pervasive role in co-regulating adaptive immunity and the diversity of microbiota.

3.
Stem Cells ; 34(4): 913-23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26840742

ABSTRACT

The cellular and molecular basis of vascular calcification (VC) in atherosclerosis is not fully understood. Here, we investigate role of resident/circulating progenitor cells in VC and contribution of inflammatory plaque environment to this process. Vessel-derived stem/progenitor cells (VSCs) and mesenchymal stem cells (MSCs) isolated from atherosclerotic ApoE(-/-) mice showed significantly more in vitro osteogenesis and chondrogenesis than cells generated from control C57BL/6 mice. To assess their ability to form bone in vivo, cells were primed chondrogenically or cultured in control medium on collagen glycosaminoglycan scaffolds in vitro prior to subcutaneous implantation in ApoE(-/-) and C57BL/6 mice using a crossover study design. Atherosclerotic ApoE(-/-) MSCs and VSCs formed bone when implanted in C57BL/6 mice. In ApoE(-/-) mice, these cells generated more mature bone than C57BL/6 cells. The atherosclerotic in vivo environment alone promoted bone formation by implanted C57BL/6 cells. Un-primed C57BL/6 VSCs were unable to form bone in either mouse strain. Treatment of ApoE(-/-) VSC chondrogenic cultures with interleukin (IL)-6 resulted in significantly increased glycosaminoglycan deposition and expression of characteristic chondrogenic genes at 21 days. In conclusion, resident vascular cells from atherosclerotic environment respond to the inflammatory milieu and undergo calcification. IL-6 may have a role in aberrant differentiation of VSCs contributing to vascular calcification in atherosclerosis.


Subject(s)
Atherosclerosis/genetics , Cytokines/metabolism , Mesenchymal Stem Cells , Osteogenesis/genetics , Plaque, Atherosclerotic/genetics , Animals , Apolipoproteins E/genetics , Atherosclerosis/pathology , Atherosclerosis/therapy , Blood Vessels/cytology , Cell Differentiation/genetics , Chondrogenesis/genetics , Glycosaminoglycans/metabolism , Humans , Interleukin-6/metabolism , Mice , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/therapy , Vascular Calcification/genetics , Vascular Calcification/metabolism , Vascular Calcification/pathology
4.
Am J Physiol Renal Physiol ; 307(12): F1412-26, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25339699

ABSTRACT

Mesenchymal stem cells (MSCs) suppress T helper (Th)17 cell differentiation and are being clinically pursued for conditions associated with aberrant Th17 responses. Whether such immunomodulatory effects are enhanced by coadministration of MSCs with other agents is not well known. In the present study, individual and combined effects of MSCs and the vitamin D receptor (VDR) agonist paricalcitol on Th17 induction were investigated in vitro and in a mouse model of sterile kidney inflammation (unilateral ureteral obstruction). In vitro, MSCs and paricalcitol additively suppressed Th17 differentiation, although only MSCs suppressed expression of Th17-associated transcriptions factors. Combined administration of MSCs and paricalcitol resulted in an early (day 3) reduction of intrarenal CD4(+) and CD8(+) T cells, CD11b(+)/lymphocyte antigen 6G(+) neutrophils, and inflammatory (lymphocyte antigen 6C(hi)) monocytes as well as reduced transcript for IL-17 compared with untreated animals. Later (day 8), obstructed kidneys of MSC/paricalcitol double-treated mice, but not mice treated with either intervention alone, had reduced tubular injury and interstitial fibrosis as well as lower numbers of neutrophils and inflammatory monocytes and an increase in the ratio between M2 (CD206(+)) and M1 (CD206(-)) macrophages compared with control mice. Adjunctive therapy with VDR agonists may enhance the immunosuppressive properties of MSCs in the setting of pathogenic Th17-type immune responses and related inflammatory responses.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Ergocalciferols/pharmacology , Immunosuppressive Agents/pharmacology , Kidney/drug effects , Mesenchymal Stem Cell Transplantation , Nephritis/prevention & control , Receptors, Calcitriol/agonists , Th17 Cells/drug effects , Animals , Biomarkers/metabolism , Cells, Cultured , Disease Models, Animal , Female , Fibrosis , Interleukin-17/genetics , Interleukin-17/metabolism , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Nephritis/etiology , Nephritis/immunology , Nephritis/metabolism , Nephritis/pathology , Neutrophil Infiltration/drug effects , Receptors, Calcitriol/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Time Factors , Ureteral Obstruction/complications
6.
Kidney Int ; 81(4): 379-90, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21975862

ABSTRACT

Interleukin 17A-secreting T-helper 17 (Th17) cells are pathogenic in inflammatory kidney diseases, but their intrarenal regulation is poorly understood. In order to better define Th17 cell dynamics during interstitial inflammation, we utilized the mouse unilateral ureteral obstruction model to analyze inflammatory cell subtypes by multicolor flow cytometry and cell sorting and by effects on in vitro-generated Th17 cells. Interleukin 17A expression localized to CCR6(+)CCR4(+/-)CD4(+) T-cells and progressively increased in obstructed kidneys. The number of CCR6(+)CD4(+) T-cells increased over 10-fold by 72 h, were enriched for interleukin 17A production, and were highly proliferative based on in vivo bromodeoxyuridine incorporation. Secreted products of leukocytes isolated from obstructed kidneys enhanced the interleukin 17A production of in vitro-generated Th17 cells. This Th17-enhancing activity was identified as interleukin-1 produced by renal dendritic cells and monocytes. The in vivo validity of these findings was confirmed in mice lacking the interleulin-1 receptor and in mice treated with a recombinant interleukin-1 receptor antagonist, each of which exhibited reduced intrarenal Th17 activity compared with control mice. Thus, the inflamed kidney accumulates CCR6(+) Th17 cells that undergo activation and proliferation. Production of interleukin 1 family cytokines by resident dendritic cells and infiltrating monocytes enhances intrarenal Th17 activation in acute kidney injury.


Subject(s)
Interleukin-17/metabolism , Interleukin-1/immunology , Nephritis/immunology , Th17 Cells/immunology , Ureteral Obstruction/immunology , Animals , CD4 Antigens/analysis , Cell Proliferation , Cells, Cultured , Dendritic Cells/metabolism , Disease Models, Animal , Female , Flow Cytometry , Interleukin-1/metabolism , Lymphocyte Count , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Nephritis/metabolism , Receptors, CCR4/analysis , Receptors, CCR6/analysis , Ureteral Obstruction/complications , Ureteral Obstruction/metabolism
7.
Kidney Int ; 80(5): 447-50, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21841834

ABSTRACT

Circulating neutrophils are essential for innate immunity and undergo rapid, stepwise adhesion to and transmigration through the endothelium following tissue injury and microbial invasion. Neutrophil dysfunction may contribute to morbidity and mortality in acute kidney injury but has not frequently been studied at a mechanistic level. Rossaint et al. provide experimental evidence in mice and humans that acute uremia causes discrete intracellular signaling abnormalities that interfere with specific stages of neutrophil trafficking during inflammation.


Subject(s)
Acute Kidney Injury/physiopathology , Cell Movement , Leukocyte Rolling , Signal Transduction , Animals
8.
Eur J Immunol ; 41(10): 2840-51, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21710489

ABSTRACT

Mesenchymal stem cells (MSCs) inhibit T-cell activation and proliferation but their effects on individual T-cell-effector pathways and on memory versus naïve T cells remain unclear. MSC influence on the differentiation of naïve and memory CD4(+) T cells toward the Th17 phenotype was examined. CD4(+) T cells exposed to Th17-skewing conditions exhibited reduced CD25 and IL-17A expression following MSC co-culture. Inhibition of IL-17A production persisted upon re-stimulation in the absence of MSCs. These effects were attenuated when cell-cell contact was prevented. Th17 cultures from highly purified naïve- and memory-phenotype responders were similarly inhibited. Th17 inhibition by MSCs was reversed by indomethacin and a selective COX-2 inhibitor. Media from MSC/Th17 co-cultures contained increased prostaglandin E2 (PGE2) levels and potently suppressed Th17 differentiation in fresh cultures. MSC-mediated Th17 inhibition was reversed by a selective EP4 antagonist and was mimicked by synthetic PGE2 and a selective EP4 agonist. Activation-induced IL-17A secretion by naturally occurring, effector-memory Th17 cells from a urinary obstruction model was also inhibited by MSC co-culture in a COX-dependent manner. Overall, MSCs potently inhibit Th17 differentiation from naïve and memory T-cell precursors and inhibit naturally-occurring Th17 cells derived from a site of inflammation. Suppression entails cell-contact-dependent COX-2 induction resulting in direct Th17 inhibition by PGE2 via EP4.


Subject(s)
Dinoprostone/metabolism , Mesenchymal Stem Cells/physiology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/metabolism , Cell Communication , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/biosynthesis , Female , Flow Cytometry , Indomethacin/pharmacology , Interleukin-17/antagonists & inhibitors , Interleukin-17/biosynthesis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Lymphocyte Activation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Polymerase Chain Reaction , Receptors, Prostaglandin E, EP4 Subtype/agonists , Th17 Cells/drug effects
9.
Transplantation ; 81(12): 1708-15, 2006 Jun 27.
Article in English | MEDLINE | ID: mdl-16794538

ABSTRACT

BACKGROUND: Production of nitric oxide (NO) by graft infiltrating macrophages has been proposed as an important effector mechanism of allograft rejection. Although high levels of NO are generated during allograft rejection, undetectable or only limited amounts of NO were found in rejected skin xenografts. METHODS: BALB/c mice were grafted with skin transplants from syngeneic, allogeneic or xenogeneic (rat) donors. The production of NO, cytokines and arginase in the grafts was determined by spectrophotometry, enzyme-linked immunosorbent assay, or polymerase chain reaction. Effects of depletion of CD4+ cells, neutralization of interleukin (IL)-4 or application of arginase inhibitors N(omega)-hydroxy-L-arginine (L-NOHA) and L-valine on production of NO in rejected xenografts were evaluated. RESULTS: Rejection of rat skin xenografts, on the contrary to rejection of allografts, was associated with a local high production of Th2 cytokines IL-4 and IL-10, overexpression of arginase genes, strongly enhanced arginase activity and attenuated NO generation in the graft. The supernatants obtained after cultivation of skin xenograft (but not allograft or syngeneic graft) explants contained a high arginase activity and strongly suppressed NO production by activated macrophages. This suppression was completely inhibited by L-NOHA or was overcome by an excess of exogenous L-arginine, a substrate for NO synthesis. Cocultivation of xenograft explants that did not produce NO with arginase inhibitors L-NOHA or L-valine restored NO generation in the graft. CONCLUSION: The results suggest that upregulation of arginase activity by Th2 cytokines during xenograft rejection limits the bioavailability of L-arginine for the inducible NO synthase and thus attenuates generation of NO by the graft-infiltrating macrophages.


Subject(s)
Arginase/metabolism , Arginine/metabolism , Graft Rejection/immunology , Graft Rejection/metabolism , Nitric Oxide/biosynthesis , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Arginase/antagonists & inhibitors , Arginase/genetics , Female , Gene Expression Regulation, Enzymologic , Graft Rejection/enzymology , Interleukin-10/biosynthesis , Interleukin-4/biosynthesis , Macrophages/metabolism , Male , Mice , Rats , Skin/metabolism , Skin Transplantation , Tissue Culture Techniques , Transplantation, Heterologous , Transplantation, Homologous
10.
Transpl Int ; 18(7): 854-62, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15948866

ABSTRACT

Corneal xenotransplantation may be an alternative approach to overcome shortage of allografts for clinical transplantation. Orthotopic corneal rat-to-mouse xenotransplantation and syngeneic transplantation was performed and the effects of anti-CD4 and anti-CD8 treatments on corneal xenograft survival and production of cytokines, interleukin (IL)-2, IL-4, IL-10, gamma-interferon (IFN-gamma) and nitric oxide (NO) were evaluated. RT-PCR was used to determine the expression of genes for cytokines and inducible nitric oxide synthase (iNOS) in the grafts. The presence of iNOS protein in grafts was detected by immunofluorescent staining. We found that corneal xenotransplantation was associated with a strong upregulation of genes for both Th1 and Th2 cytokines and with NO production in the graft. Treatment of xenograft recipients with mAb anti-CD4, but not anti-CD8, resulted in a profound inhibition of IL-2, IL-4 and IL-10 production, and in a significant prolongation of corneal xenograft survival. The results show that upregulation of Th2 cytokines after corneal xenotransplantation does not correlate with xenograft rejection. Rather, corneal graft rejection is associated with the expression of genes for IFN-gamma and iNOS and with NO production.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD4 Antigens/immunology , CD8 Antigens/immunology , Corneal Transplantation , Cytokines/biosynthesis , Nitric Oxide/biosynthesis , Transplantation, Heterologous , Animals , Cornea/metabolism , Female , Gene Expression , Graft Survival/drug effects , Male , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Rats , Rats, Inbred Lew
11.
Xenotransplantation ; 12(3): 227-34, 2005 May.
Article in English | MEDLINE | ID: mdl-15807773

ABSTRACT

BACKGROUND: Production of nitric oxide (NO) by graft infiltrating macrophages has been suggested as an important effector mechanism of allograft rejection. Expression of the gene for the inducible NO synthase (iNOS) and the production of NO in rejected graft has been demonstrated in various models of allotransplantation. However, whether NO plays a role in rejection of skin xenografts has not been documented. METHODS: Explants of rejected skin allografts or xenografts (rat to mouse) were cultivated in vitro and the production of NO, interleukin (IL)-2, IL-4, IL-10 and interferon-gamma (IFN-gamma) by graft infiltrating cells was determined by the Griess reaction or ELISA. Effects of supernatants from cultures of xenograft explants on the expression of gene for iNOS, accumulation of iNOS protein and NO production were determined by RT-PCR or Western blots. Molecular mass of the factor with the suppressive activity was characterized by filtration on chromatography Sephacryl S-200 Superfine column. In addition, the effects of 2-amino-5,6-dihydro-6-methyl-4H-1,3-thiazine (AMT), a selective iNOS inhibitor, on survival of skin xenografts were tested. RESULTS: While explants of rejected mouse skin allografts produced substantial amounts of NO, undetectable or only very low levels of NO were found in supernatants from cultured rat skin xenografts. Cocultivation of bacterial lipopolysaccharide (LPS)-stimulated mouse macrophages which produce high quantities of NO, with pieces of rejected xenografts, but not of syngeneic grafts, allografts or normal rat skin, completely inhibited production of NO. Production of IL-6 and IL-10 by LPS-stimulated macrophages was not inhibited under the same conditions. The inhibition of NO production was mediated by a factor which was produced by rejected rat xenograft and which was eluted from chromatography Sephacryl S-200 Superfine column in a fraction representing a molecular mass of 67 kDa. The factor did not inhibit the expression of the gene for iNOS, reduce the level of iNOS protein in stimulated macrophages, or function as a scavenger of NO. Rather, the factor inhibited the function of iNOS. The finding that NO does not play an important role during rejection of skin xenografts is supported by the observation that treatment of graft recipients with AMT, a specific iNOS inhibitor, did not enhance xenograft survival, while the same treatment resulted in prolongation of survival of skin allografts. CONCLUSION: The results thus demonstrate that a 67-kDa molecule produced by rejected rat skin xenografts selectively inhibits iNOS activity in graft infiltrating macrophages. We suggest that NO does not play a significant role in rejection of skin xenografts as it does in the case of allograft rejection.


Subject(s)
Graft Rejection/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Skin Transplantation , Skin/metabolism , Transplantation, Heterologous , Animals , Cytokines/biosynthesis , Enzyme Inhibitors/pharmacology , Female , Graft Rejection/pathology , Graft Survival/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Weight , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II , Rats , Rats, Inbred Lew , Skin/enzymology , Skin/pathology
12.
Mol Immunol ; 38(12-13): 989-95, 2002 May.
Article in English | MEDLINE | ID: mdl-12009578

ABSTRACT

Nitric oxide (NO) as a small ubiquitous molecule influencing a great variety of biological processes in the organism. Within the immune system, increased levels of NO were observed in various immunopathological situations, inflammatory reactions and during the response to transplantation and tumour antigens. It appears that NO can influence various facets of immune response. We studied involvement and the role of NO in immune response to skin allograft in mice. The production of NO at the site of graft rejection correlated well with the kinetic of rejection reaction and with the fate of the allograft. Graft infiltrating macrophages were identified as a principal cell population producing NO and the production of NO by macrophages was dependent on the presence of activated CD4(+) T cells. Survival of skin allografts was significantly prolonged by the treatment of graft recipients with 2-amino-5,6-dihydro-6-methyl-4H-1,3-thiazine (AMT), a specific inhibitor of inducible NO synthase (iNOS). These results suggest a role for NO as the effector cytotoxic molecule involved in the graft rejection. Experiments in vitro demonstrated that NO, in addition to its effector function, acts as a modulator of cytokine production. Spleen cells stimulated with alloantigens in the presence of AMT or S-ethylisothiourea (EIT), an another selective iNOS inhibitor, produced considerably more interleukin (IL)-4 and IL-10 than the cells stimulated in the absence of iNOS inhibitors. The production of Th1 cytokines IL-2 and interferon (IFN)-gamma was not enhanced by the inhibition of NO synthesis. The results altogether show that NO can act in transplantation reactions as an immunomodulator on cytokine production level and as an effector molecule involved in the graft destruction.


Subject(s)
Graft Rejection/immunology , Nitric Oxide/physiology , Skin Transplantation/immunology , Animals , Cells, Cultured , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Graft Survival/drug effects , Graft Survival/immunology , Kinetics , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Thiazines/pharmacology , Transplantation Tolerance
SELECTION OF CITATIONS
SEARCH DETAIL
...