Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Type of study
Language
Publication year range
1.
Front Cell Dev Biol ; 11: 1089817, 2023.
Article in English | MEDLINE | ID: mdl-36875761

ABSTRACT

Type 1 diabetes mellitus (T1DM) is an autoimmune disorder specifically targeting pancreatic islet beta cells. Despite many efforts focused on identifying new therapies able to counteract this autoimmune attack and/or stimulate beta cells regeneration, TD1M remains without effective clinical treatments providing no clear advantages over the conventional treatment with insulin. We previously postulated that both the inflammatory and immune responses and beta cell survival/regeneration must be simultaneously targeted to blunt the progression of disease. Umbilical cord-derived mesenchymal stromal cells (UC-MSC) exhibit anti-inflammatory, trophic, immunomodulatory and regenerative properties and have shown some beneficial yet controversial effects in clinical trials for T1DM. In order to clarify conflicting results, we herein dissected the cellular and molecular events derived from UC-MSC intraperitoneal administration (i.p.) in the RIP-B7.1 mouse model of experimental autoimmune diabetes. Intraperitoneal (i.p.) transplantation of heterologous mouse UC-MSC delayed the onset of diabetes in RIP-B7.1 mice. Importantly, UC-MSC i. p. transplantation led to a strong peritoneal recruitment of myeloid-derived suppressor cells (MDSC) followed by multiple T-, B- and myeloid cells immunosuppressive responses in peritoneal fluid cells, spleen, pancreatic lymph nodes and the pancreas, which displayed significantly reduced insulitis and pancreatic infiltration of T and B Cells and pro-inflammatory macrophages. Altogether, these results suggest that UC-MSC i. p. transplantation can block or delay the development of hyperglycemia through suppression of inflammation and the immune attack.

2.
Cell Death Differ ; 17(6): 1025-33, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20075941

ABSTRACT

Exposure of mouse embryonic stem (mES) cells to high concentrations of chemical nitric oxide (NO) donors promotes differentiation, but the mechanisms involved in this process at the gene expression level are poorly defined. In this study we report that culture of mES cells in the presence of 0.25-1.0 mM diethylenetriamine nitric oxide adduct (DETA-NO) leads to downregulation of Nanog and Oct4, the two master genes involved in the control of the pluripotent state. This action of NO was also apparent in the human ES cell line, HS 181. The suppressive action of NO on Nanog gene depends on the activation of p53 repressor protein by covalent modifications, such as pSer15, pSer315, pSer392 and acetyl Lys 379. NO-induced repression of Nanog is also associated with binding of trimethylated histone H3 and pSer315 p53 to its promoter region. In addition, exposure to 0.5 mM DETA-NO induces early differentiation events of cells with acquisition of epithelial morphology and expression of markers of definitive endoderm, such as FoxA2, Gata4, Hfn1-beta and Sox 17. This phenotype was increased when cells were treated with valproic acid (VPA) for 10 days.


Subject(s)
Embryonic Stem Cells/metabolism , Homeodomain Proteins/genetics , Nitric Oxide/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Line , Down-Regulation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Endoderm/cytology , Histones/metabolism , Homeodomain Proteins/metabolism , Mice , Nanog Homeobox Protein , Nitric Oxide Donors/pharmacology , Octamer Transcription Factor-3/metabolism , Phenotype , Promoter Regions, Genetic , Protein Processing, Post-Translational , Tumor Suppressor Protein p53/metabolism
3.
Cell Death Dis ; 1: e80, 2010 Oct 07.
Article in English | MEDLINE | ID: mdl-21368853

ABSTRACT

Nitric oxide (NO) is an intracellular messenger in several cell systems, but its contribution to embryonic stem cell (ESC) biology has not been characterized. Exposure of ESCs to low concentrations (2-20 µM) of the NO donor diethylenetriamine NO adduct confers protection from apoptosis elicited by leukaemia inhibitory factor (LIF) withdrawal. NO blocked caspase 3 activation, PARP degradation, downregulation of the pro-apoptotic genes Casp7, Casp9, Bax and Bak1 and upregulation of the anti-apoptotic genes Bcl-2 111, Bcl-2 and Birc6. These effects were also observed in cells overexpressing eNOS. Exposure of LIF-deprived mESCs to low NO prevented the loss of expression of self-renewal genes (Oct4, Nanog and Sox2) and the SSEA marker. Moreover, NO blocked the differentiation process promoted by the absence of LIF and bFGF in mouse and human ESCs. NO treatment decreased the expression of differentiation markers, such as Brachyury, Gata6 and Gata4. Constitutive overexpression of eNOS in cells exposed to LIF deprivation maintained the expression of self-renewal markers, whereas the differentiation genes were repressed. These effects were reversed by addition of the NOS inhibitor L-NMMA. Altogether, the data suggest that low NO has a role in the regulation of ESC differentiation by delaying the entry into differentiation, arresting the loss of self-renewal markers and promoting cell survival by inhibiting apoptosis.


Subject(s)
Embryonic Stem Cells/cytology , Nitric Oxide/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Survival , Embryonic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Humans , Leukemia Inhibitory Factor/metabolism , Lewis X Antigen/metabolism , Mice , Nitric Oxide Synthase Type III/metabolism , Octamer Transcription Factor-3/metabolism , Polyamines/pharmacology , SOXB1 Transcription Factors/metabolism
4.
Cell Signal ; 13(11): 809-17, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11583916

ABSTRACT

The mechanism by which nitric oxide (NO) protects from apoptosis is a matter of debate. We have shown previously that phosphorylation of tyrosine residues participates in the protection from apoptosis in insulin-producing RINm5F cells (Inorg. Chem. Commun. 3 (2000) 32). Since NO has been reported to activate the tyrosine kinase c-Src and this kinase is involved in the activation of protein kinase G (PKG) in some cell systems, we aimed at studying the contribution of c-Src and PKG systems in anti-apoptotic actions of NO in serum-deprived RINm5F cells. Here we report that exposure of serum-deprived cells to 10 microM DETA/NO results in protection from degradation of the anti-apoptotic protein Bcl-2, together with a reduction of cytochrome c release from mitochondria and caspase-3 inhibition. Studies with the inhibitors ODQ and KT-5823 revealed that these actions are dependent on both activation of guanylate cyclase and PKG. DETA/NO was also able to induce autophosphorylation and activation c-Src protein both in vivo and in vitro and active c-Src was able to induce tyrosine phosphorylation of Bcl-2 in vitro. The c-Src kinase inhibitor PP1 abrogated the actions of DETA/NO on cGMP formation, PKG activation, caspase activation, cytochrome c release from mitochondria, and Bcl-2 phosphorylation and degradation in serum-deprived cells. We thus propose that activation of c-Src is an early step in the chain of events that signal cGMP-dependent anti-apoptotic actions of NO in mitocohondria.


Subject(s)
Apoptosis , Islets of Langerhans/enzymology , Nitric Oxide/physiology , Proto-Oncogene Proteins pp60(c-src)/physiology , Animals , Cell Line , Cell Survival , Culture Media, Serum-Free , Cyclic GMP/biosynthesis , Cyclic GMP-Dependent Protein Kinases/physiology , Guanylate Cyclase/physiology , Islets of Langerhans/cytology , Nitric Oxide Donors/pharmacology , Triazenes/pharmacology
5.
Exp Cell Res ; 269(2): 222-9, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11570814

ABSTRACT

Exposure of insulin-secreting RINm5F cells to the chemical nitric oxide donor sodium nitroprusside (SNP) resulted in apoptotic cell death, as detected by cytochrome c release from mitochondria and caspase 3 activation. SNP exposure also leads to phosphorylation and activation of enzymes involved in cellular response to stress such as signal-regulated kinase 2 (ERK2) and c-Jun NH(2)-terminal kinase 46 (JNK46). Both cytochrome c release and caspase 3 activation were abrogated in cells exposed to MEK and p38 inhibitors. Treatment of cells with the NO donors SNP, DETA-NO, GEA 5024, and SNAP resulted in phosphorylation of the antiapoptotic protein Bcl-2, which was resistant to blockade of MEK, p38, and JNK pathways and sensitive to phosphoinositide 3-kinase (PI3K) inhibition. In addition, transient transfection of cells with the wild-type PI3K gamma gene mimics the increased rate of Bcl-2 phosphorylation detected in NO-treated cells. The generation of phosphoinositides seems to participate in the process since Bcl-2 phosphorylation was not observed in cells overexpressing lipid-kinase-deficient PI3Kgamma. The potential of SNP toxicity directly from NO was supported by our finding that the NO scavenger carboxy-PTIO prevented cell death. We found no evidence to support the contention that oxygen radicals generated during cellular SNP metabolism mediate cell toxicity in RINm5F cells, since neither addition of catalase/superoxide dismutase nor transfection with superoxide dismutase prevented SNP-induced cell death. Thus, we propose that exposure to apoptotic concentrations of NO triggers ERK- and p38-dependent cytochrome c release, caspase 3 activation, and PI3K-dependent Bcl-2 phosphorylation.


Subject(s)
Apoptosis , Insulin/metabolism , MAP Kinase Signaling System , Mitochondria/pathology , Nitroprusside/pharmacology , Animals , Blotting, Western , Caspase 3 , Caspases/metabolism , Catalase/metabolism , Cell Line , Cell Survival , Cells, Cultured , Cytochrome c Group/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Free Radicals , Imidazoles/pharmacology , Indicators and Reagents/pharmacology , Islets of Langerhans/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Precipitin Tests , Protein Binding , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyridines/pharmacology , Rats , Signal Transduction , Superoxide Dismutase/metabolism , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL