Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Immunol ; 203(7): 1753-1765, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31462505

ABSTRACT

Chronic neuroinflammation is a key pathological hallmark of multiple sclerosis (MS) that suggests that resolution of inflammation by specialized proresolving molecules is dysregulated in the disease. Annexin A1 (ANXA1) is a protein induced by glucocorticoids that facilitates resolution of inflammation through several mechanisms that include an inhibition of leukocyte recruitment and activation. In this study, we investigated the ability of ANXA1 to influence T cell effector function in relapsing/remitting MS (RRMS), an autoimmune disease sustained by proinflammatory Th1/Th17 cells. Circulating expression levels of ANXA1 in naive-to-treatment RRMS subjects inversely correlated with disease score and progression. At the cellular level, there was an impaired ANXA1 production by CD4+CD25- conventional T and CD4+RORƎĀ³t+ T (Th17) cells from RRMS subjects that associated with an increased migratory capacity in an in vitro model of blood brain barrier. Mechanistically, ANXA1 impaired monocyte maturation secondarily to STAT3 hyperactivation and potently reduced T cell activation, proliferation, and glycolysis. Together, these findings identify impaired disease resolution pathways in RRMS caused by dysregulated ANXA1 expression that could represent new potential therapeutic targets in RRMS.


Subject(s)
Annexin A1/immunology , Gene Expression Regulation/immunology , Lymphocyte Activation , Multiple Sclerosis/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Adult , Cell Proliferation , Female , Glycolysis/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Male , Middle Aged , Monocytes/immunology , Monocytes/pathology , Multiple Sclerosis/pathology , STAT3 Transcription Factor/immunology , Severity of Illness Index , Th1 Cells/pathology , Th17 Cells/pathology
2.
J Immunol ; 189(6): 2941-53, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22904304

ABSTRACT

The sensing by T cells of metabolic and energetic changes in the microenvironment can determine the differentiation, maturation, and activation of these cells. Although it is known that mammalian target of rapamycin (mTOR) gauges nutritonal and energetic signals in the extracellular milieu, it is not known how mTOR and metabolism influence CD4+CD25-FOXP3- effector T cell (Teff) responses. In this article, we show that leptin-induced activation of mTOR, which, in turn, controls leptin production and signaling, causes a defined cellular, biochemical, and transcriptional signature that determine the outcome of Teff responses, both in vitro and in vivo. The blockade of leptin/leptin receptor signaling, induced by genetic means or by starvation, leads to impaired mTOR activity that inhibits the proliferation of Teffs in vivo. Notably, the transcriptional signature of Teffs in the presence of leptin blockade appears similar to that observed in rapamycin-treated Teffs. These results identify a novel link between nutritional status and Teff responses through the leptin-mTOR axis and define a potential target for Teff modulation in normal and pathologic conditions.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Leptin/physiology , TOR Serine-Threonine Kinases/metabolism , Transcriptional Activation/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/pathology , Cell Line , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Down-Regulation/immunology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mice, Transgenic , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transcriptional Activation/drug effects
3.
Sci Rep ; 14(1): 17571, 2024 07 30.
Article in English | MEDLINE | ID: mdl-39080325

ABSTRACT

The molecular mechanisms that govern differential T cell development from CD4+CD25-conventional T (Tconv) into CD4+CD25+ forkhead-box-P3+ (FoxP3+) inducible regulatory T (iTreg) cells remain unclear. Herein, we investigated the relative contribution of protein kinase A (PKA) in this process. Mechanistically, we found that PKA controlled the efficiency of human iTreg cell generation through the expression of different FoxP3 splicing variants containing or not the exon 2. We found that transient PKA inhibition reduced the recruitment of cAMP-responsive element-binding protein (CREB) on regulatory regions of the FoxP3 gene, a condition that is associated with an impaired acquisition of their suppressive capacity in vitro. To corroborate our findings in a human model of autoimmunity, we measured CREB phosphorylation and FoxP3 levels in iTreg cells from treatment-naĆÆve relapsing-remitting (RR)-multiple sclerosis (MS) subjects. Interestingly, both phospho-CREB and FoxP3 induction directlyĀ correlated and were significantly reduced in RR-MS patients, suggesting a previously unknown mechanism involved in the induction and function of human iTreg cells.


Subject(s)
Autoimmunity , Cyclic AMP Response Element-Binding Protein , Cyclic AMP-Dependent Protein Kinases , Forkhead Transcription Factors , T-Lymphocytes, Regulatory , Humans , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Phosphorylation , Gene Expression Regulation , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Female , Male
4.
J Biol Chem ; 285(31): 24141-53, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20495008

ABSTRACT

Nerve growth factor (NGF) induces terminal differentiation in PC12, a pheochromocytoma-derived cell line. NGF binds a specific receptor on the membrane and triggers the ERK1/2 cascade, which stimulates the transcription of neural genes. We report that NGF significantly affects mitochondrial metabolism by reducing mitochondrial-produced reactive oxygen species and stabilizing the electrochemical gradient. This is accomplished by stimulation of mitochondrial manganese superoxide dismutase (MnSOD) both transcriptionally and post-transcriptionally via Ki-Ras and ERK1/2. Activation of MnSOD is essential for completion of neuronal differentiation because 1) expression of MnSOD induces the transcription of a neuronal specific promoter and neurite outgrowth, 2) silencing of endogenous MnSOD by small interfering RNA significantly reduces transcription induced by NGF, and 3) a Ki-Ras mutant in the polylysine stretch at the COOH terminus, unable to stimulate MnSOD, fails to induce complete differentiation. Overexpression of MnSOD restores differentiation in cells expressing this mutant. ERK1/2 is also downstream of MnSOD, as a SOD mimetic drug stimulates ERK1/2 with the same kinetics of NGF and silencing of MnSOD reduces NGF-induced late ERK1/2. Long term activation of ERK1/2 by NGF requires SOD activation, low levels of hydrogen peroxide, and the integrity of the microtubular cytoskeleton. Confocal immunofluorescence shows that NGF stimulates the formation of a complex containing membrane-bound Ki-Ras, microtubules, and mitochondria. We propose that active NGF receptor induces association of mitochondria with plasma membrane. Local activation of ERK1/2 by Ki-Ras stimulates mitochondrial SOD, which reduces reactive oxygen species and produces H(2)O(2). Low and spatially restricted levels of H(2)O(2) induce and maintain long term ERK1/2 activity and ultimately differentiation of PC12 cells.


Subject(s)
Genes, ras/genetics , Nerve Growth Factor/metabolism , Reactive Oxygen Species , Superoxide Dismutase/metabolism , ras Proteins/metabolism , Animals , Cell Differentiation , Cytoskeleton/metabolism , Humans , MAP Kinase Signaling System , Mitochondria/enzymology , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , PC12 Cells , Rats
5.
J Leukoc Biol ; 74(6): 1117-24, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12972513

ABSTRACT

The ability of the envelope glycoprotein gp120 [human immunodeficiency virus (HIV) env] to induce intracellular signals is thought to contribute to HIV-1 pathogenesis. In the present study, we found that the exposure of CD4+ CD45RA+ naive T cells to HIVenv results in a long-lasting hyporesponsiveness to antigen stimulation. This phenomenon is not dependent on CD4-mediated signals and also can be generated by the exposure of naive T cell to soluble CD4-HIVenv complexes. The analysis of the proximal signaling reveals that HIVenv does not activate Lck as well as the mitogen-activated protein kinase intermediate cascade. Conversely, the envelope glycoprotein stimulates the cyclic adenosine monophosphate (cAMP)-dependent protein kinase A (PKA) activity and induces the progressive accumulation of the phosphorylated form of the cAMP-responsive element binding. Of note, the ligation of CXCR4 by stromal cell-derived factor-1alpha but not the engagement of CD4 by monoclonal antibody stimulates the PKA activity and induces a long-lasting hyporesponsivity state in naive CD4+ lymphocytes. The pretreatment of lymphocytes with H89, a cell-permeable PKA inhibitor, prevents the induction of anergy. These findings reveal a novel mechanism by which HIVenv may modulate the processes of clonal expansion, homeostatic proliferation, and terminal differentiation of the naive T lymphocyte subset.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cyclic AMP-Dependent Protein Kinases/metabolism , HIV Envelope Protein gp120/pharmacology , HIV-1/immunology , Sulfonamides , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Cell Differentiation , Cells, Cultured , Clonal Anergy , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dendritic Cells/metabolism , Enzyme Inhibitors/pharmacology , Humans , Isoquinolines/pharmacology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Monocytes/metabolism
6.
Gene ; 291(1-2): 115-21, 2002 May 29.
Article in English | MEDLINE | ID: mdl-12095685

ABSTRACT

We have examined the effects of an adenosine 3',5'-cyclic monophosphate (cAMP) analog on human aldolase C gene expression in the rat pheochromocytoma cell line PC12. Incubation for 4 h with 500 microM 8-Br-cAMP increased aldolase C mRNA expression 2.5-fold and the expression was still above basal level 24 h later. Using transient transfection experiments we demonstrate that the distal element D in the promoter region of the human aldolase C gene, which binds a transcriptional activator (NGFI-B), is involved in this regulation. NGFI-B mRNA and protein expression were promptly (15 min) increased after 8-Br-cAMP treatment and precedes aldolase C mRNA increase (30 min). After 4 h of 8-Br-cAMP treatment, the binding of NGFI-B protein to the distal element D in the distal promoter region was increased twofold and this correlates with the increased expression of the clone that contains distal element D. These results indicate that the distal element D in the promoter region of the human aldolase C gene is the target of a cAMP-dependent regulation pathway.


Subject(s)
Cyclic AMP/pharmacology , Fructose-Bisphosphate Aldolase/genetics , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Chloramphenicol O-Acetyltransferase/drug effects , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression/genetics , Gene Expression Regulation, Enzymologic/drug effects , Humans , Nuclear Receptor Subfamily 4, Group A, Member 1 , PC12 Cells , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Cytoplasmic and Nuclear , Receptors, Steroid , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation/drug effects
7.
Metabolism ; 63(10): 1272-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25060689

ABSTRACT

OBJECTIVE: In this report we show that the adipocytokine leptin directly modulates autophagy in human CD4(+)CD25(-) conventional (Tconv) T cells. RESULTS: In vitro treatment with recombinant human leptin determined an inhibition of autophagy during T cell receptor (TCR) stimulation, and this phenomenon was dose- and time-dependent. The events were secondary to the activation of the mammalian-target of rapamycin (mTOR)-pathway induced by leptin, as testified by its reversion induced by mTOR inhibition with rapamycin. At molecular level these phenomena associated with Bcl-2 up-regulation and its interaction with Beclin-1, whose complex exerts a negative effect on autophagy. MATERIALS/METHODS: The impact of leptin on autophagy of Tconv cells was determined at biochemical level by western blotting and by flow cytometry; the interaction between BCL-2 and Beclin-1 by co-immunoprecipitation assays. CONCLUSIONS: Our results, suggest that in unconditioned, freshly-isolated human Tconv cells, autophagy and proliferation are controlled by leptin during TCR-engagement, and that both phenomena occur alternatively indicating a balance between these processes during immune activation.


Subject(s)
Autophagy/physiology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/physiology , Interleukin-2 Receptor alpha Subunit/metabolism , Leptin/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/physiology , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Beclin-1 , CD4-Positive T-Lymphocytes/drug effects , Cell Line , Cell Proliferation/drug effects , Humans , Immunoprecipitation/methods , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
8.
Expert Rev Clin Immunol ; 7(3): 287-94, 2011 May.
Article in English | MEDLINE | ID: mdl-21595595

ABSTRACT

For decades, obesity has been considered to be the result of the complex interaction between genes and the environment and its pathogenesis is still unresolved. The discovery of hormones and neural mediators responsible for the control of food intake and metabolism at the hypothalamic level has provided fundamental insights into the complicated pathways that control food intake. However, the molecular basis for the association between obesity and low-degree chronic inflammation is still unknown. More recently, the discovery of leptin, one of the most abundant adipocyte-derived hormones, has suggested that nutritional status, through leptin secretion, can control immune self-tolerance modulating Treg suppressive function and responsiveness. Furthermore, recent experimental evidence has shown the presence of an abundant adipose tissue-resident Treg population responsible for the control of metabolic parameters and glucose homeostasis. Better knowledge of the intricate network of interactions among leptin-related energy regulation, Treg activities and obesity could lead to valuable strategies for therapeutic intervention in obesity and obesity-associated insulin resistance.


Subject(s)
Autoimmune Diseases/complications , Obesity/complications , Autoimmune Diseases/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Genetic Predisposition to Disease , Humans , Leptin/genetics , Receptor, Melanocortin, Type 4/genetics , Repressor Proteins/genetics , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
9.
J Biol Chem ; 280(43): 36474-82, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-16081426

ABSTRACT

The levels of Ras proteins in human primary fibroblasts are regulated by PDGF (platelet-derived growth factor). PDGF induced post-transcriptionally Ha-Ras by stimulating reactive oxygen species (ROS) and ERK1/2. Activation of ERK1/2 and high ROS levels stabilize Ha-Ras protein, by inhibiting proteasomal degradation. We found a remarkable example in vivo of amplification of this circuitry in fibroblasts derived from systemic sclerosis (scleroderma) lesions, producing vast excess of ROS and undergoing rapid senescence. High ROS, Ha-Ras, and active ERK1/2 stimulated collagen synthesis, DNA damage, and accelerated senescence. Conversely ROS or Ras inhibition interrupted the signaling cascade and restored the normal phenotype. We conclude that in primary fibroblasts stabilization of Ras protein by ROS and ERK1/2 amplifies the response of the cells to growth factors and in systemic sclerosis represents a critical factor in the onset and progression of the disease.


Subject(s)
Fibroblasts/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Platelet-Derived Growth Factor/physiology , Reactive Oxygen Species , Scleroderma, Systemic/pathology , ras Proteins/metabolism , Apoptosis , Blotting, Northern , Cells, Cultured , DNA Damage , Flow Cytometry , Humans , Immunoblotting , MAP Kinase Kinase 1/metabolism , Microscopy, Fluorescence , Models, Biological , Oxidation-Reduction , Phenotype , Platelet-Derived Growth Factor/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL