Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Clin Sci (Lond) ; 135(3): 575-588, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33496327

ABSTRACT

Hemolytic Uremic Syndrome (HUS), a disease triggered by Shiga toxin (Stx), is characterized by hemolytic anemia, thrombocytopenia and renal failure. The inflammatory response mediated by polymorphonuclear neutrophils (PMNs) and monocytes is essential to HUS onset. Still, the role of anti-inflammatory cytokines is less clear. The deficiency of IL-10, an anti-inflammatory cytokine, leads to severe pathology in bacterial infections but also to beneficial effects in models of sterile injury. The aim of this work was to analyze the role of IL-10 during HUS. Control and IL-10 lacking mice (IL-10-/-) were intravenously injected with Stx type 2 (Stx2) and survival rate was evaluated. PMN and circulating and renal pro- and anti-inflammatory factors were analyzed by FACS and enzyme-linked immunosorbent assay (ELISA) respectively. IL-10-/- mice showed a higher survival associated with lower renal damage reflected by reduced plasma urea and creatinine levels than control mice. Circulating PMN increased at 72 h in both mouse strains accompanied by an up-regulation of CD11b in control mice. In parallel, renal PMN were significantly increased only in control mice after toxin. Plasma TNF-α, IL-6 and corticosterone levels were higher increased in IL-10-/- than control mice. Simultaneously renal TNF-α raised constantly but was accompanied by increased TGF-ß levels in IL-10-/- mice. These results demonstrate that the profile of circulating and renal cytokines after Stx2 differed between strains suggesting that balance of these factors could participate in renal protection. We conclude that IL-10 absence has a protective role in an experimental model of HUS by reducing PMN recruitment into kidney and renal damage, and increasing mice survival.


Subject(s)
Hemolytic-Uremic Syndrome/chemically induced , Interleukin-10/metabolism , Shiga Toxin 2/toxicity , Animals , Corticosterone/blood , Hemolytic-Uremic Syndrome/pathology , Interleukin-10/genetics , Interleukin-6/blood , Kidney/chemistry , Kidney/pathology , Mice, Inbred BALB C , Mice, Knockout , Neutrophils , Survival Rate , Transforming Growth Factor beta , Tumor Necrosis Factor-alpha/blood
2.
Immunobiology ; 225(1): 151856, 2020 01.
Article in English | MEDLINE | ID: mdl-31744627

ABSTRACT

Sepsis is characterized by an early pro-inflammatory phase followed by compensatory anti-inflammatory mechanisms that lead to a late generalized immunosuppression, period where most deaths occur. Immunotherapy approaches to recover the immunocompetence in sepsis are similar to those used in cancer. Meta-tyrosine (m-Tyr) is a product of oxidative stress present in circulation during the sepsis and cancer-associated pro-inflammatory stages. In this work, considering its potential participation in pro-inflammatory processes, we evaluate the effect of m-Tyr during LPS induced immunosuppression phase in a murine model. In addition, we examine the effect of m-Tyr in a vaccination strategy using a weakly immunogenic tumor model. Our results showed that m-Tyr could prevent the establishment of immunosuppression and rescue the host from an installed immunosuppression induced by LPS. These effects were parallel to the ability of m-Tyr to improve the pro-inflammatory effects induced by LPS and inhibit the anti-inflammatory action of dexamethasone. Also, m-Tyr treatment prevents both the reduction of splenic lymphocytes and the increase of the expression of programmed death ligand-1 in splenic myeloid cells associated with immunosuppression. Besides, treatment with m-Tyr increased the protective effect of an anti-tumor vaccine, suggesting that m-Tyr could improve the immune response. In summary, we suggest that m-Tyr can modulate critical immunological indicators through the inflammatory context, which could improve the management of diseases, such as sepsis and cancer, in which immunosuppression is a significant clinical problem.


Subject(s)
Cancer Vaccines/immunology , Immunologic Factors/therapeutic use , Mammary Neoplasms, Animal/therapy , Myeloid-Derived Suppressor Cells/physiology , Sepsis/therapy , Tyrosine/therapeutic use , Animals , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Lipopolysaccharides/immunology , Mammary Neoplasms, Animal/immunology , Mice , Mice, Inbred BALB C , Myeloid-Derived Suppressor Cells/drug effects , Neoplasm Metastasis , Sepsis/immunology
3.
Shock ; 52(2): 264-273, 2019 08.
Article in English | MEDLINE | ID: mdl-30044324

ABSTRACT

Sepsis constitutes one of the major causes of death in ICUs. In sepsis induced by gram-negative, although lipopolysaccharide (LPS) initially induces an exacerbated secretion of proinflammatory cytokines leading to endotoxic shock and death resembling a septic shock, it is also capable of inducing refractoriness to subsequent challenge with LPS, a state known as endotoxin tolerance, which is considered the initial step of the immunosuppression found in septic patients. As we previously demonstrated the importance of glucocorticoids in endotoxin tolerance, the aim of this study was to evaluate the contribution of Interleukin-10 (IL-10) both in the endotoxic shock and in the development of the tolerance and its relationship with glucocorticoids. Our results show that, upon LPS challenge, IL-10 knockout mice (KO) mice had an enhanced LPS sensitivity, along with elevated levels of proinflammatory cytokines as tumor necrosis factor-α, IL-12 and interferon-γ, and enhanced tissue damage, despite the high levels of glucocorticoids. This effect may be because, in part, of the higher expression of tumor necrosis factor receptors in IL-10 KO mice. Further, the injection of dexamethasone did not protect IL-10 KO mice from a LPS lethal challenge. Although tolerance was achieved in the absence of IL-10, it was weaker and the elevated levels of glucocorticoids were not able to reverse the high sensitivity of IL-10 KO mice to LPS. Nevertheless, glucocorticoids would play a pivotal role in the establishment and maintenance of this partial tolerance in IL-10 KO mice. Finally, our results show that IL-10 and glucocorticoids could act in a bidirectional way influencing the inflammatory and anti-inflammatory periods.


Subject(s)
Endotoxins/toxicity , Glucocorticoids/pharmacology , Interleukin-10/deficiency , Shock, Septic/drug therapy , Animals , Cytokines/metabolism , Dexamethasone/pharmacology , Immune Tolerance/drug effects , Interleukin-10/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mifepristone/therapeutic use , Shock, Septic/chemically induced , Tumor Necrosis Factor-alpha/metabolism
4.
Clin Sci (Lond) ; 130(4): 259-71, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26582821

ABSTRACT

Secondary infections due to post-sepsis immunosuppression are a major cause of death in patients with sepsis. Repetitive inoculation of increasing doses of lipopolysaccharide (LPS) into mice mimics the immunosuppression associated with sepsis. Myeloid-derived suppressor cells (MDSCs, Gr-1(+) CD11b(+)) are considered a major component of the immunosuppressive network, interfering with T-cell responses in many pathological conditions. We used LPS-immunosuppressed (IS) mice to address whether MDSCs acquired their suppressive ability in the bone marrow (BM) and whether they could migrate to lymph nodes (LNs) to exert their suppressive function. Our results showed that Gr-1(+) CD11b(+) cells of IS mice already had the potential to inhibit T-cell proliferation in the BM. Moreover, soluble factors present in the BM from IS mice were responsible for inducing this inhibitory ability in control BM cells. In addition, migration of Gr-1(+) CD11b(+) to LNs in vivo was maximal when cells obtained from the BM of IS mice were inoculated into an IS context. In this regard, we found chemoattractant activity in cell-free LN extracts (LNEs) from IS mice and an increased expression of the LN-homing chemokine receptor C-C chemokine receptor type 7 (CCR7) in IS BM Gr-1(+) CD11b(+) cells. These results indicate that Gr-1(+) CD11b(+) cells found in BM from IS mice acquire their suppressive activity in the same niche where they are generated, and migrate to LNs to exert their inhibitory role. A better understanding of MDSC generation and/or regulation of factors able to induce their inhibitory function may provide new and more effective tools for the treatment of sepsis-associated immunosuppression.


Subject(s)
Antigens, Ly/immunology , Bone Marrow Cells/immunology , CD11b Antigen/immunology , Chemotaxis/drug effects , Immunocompromised Host , Lipopolysaccharides , Lymph Nodes/immunology , Myeloid Cells/immunology , Sepsis/immunology , Animals , Antigens, Ly/metabolism , Bone Marrow Cells/metabolism , CD11b Antigen/metabolism , Cells, Cultured , Cellular Microenvironment , Coculture Techniques , Disease Models, Animal , Lymph Nodes/metabolism , Lymphocyte Activation , Mice, Inbred BALB C , Myeloid Cells/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Sepsis/chemically induced , Sepsis/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
5.
Innate Immun ; 20(6): 585-97, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24048770

ABSTRACT

Prior exposure to endotoxins renders the host temporarily refractory to subsequent endotoxin challenge (endotoxin tolerance). Clinically, this state has also been pointed out as the initial cause of the non-specific humoral and cellular immunosuppression described in these patients. We recently demonstrated the restoration of immune response with mifepristone (RU486), a receptor antagonist of glucocorticoids. Here we report the treatment with other modulators of glucocorticoids, i.e. dehydroepiandrosterone (DHEA), a hormone with anti-glucocorticoid properties, or metyrapone (MET) an inhibitor of corticosterone synthesis. These drugs were able to partially, but significantly, restore the humoral immune response in immunosuppressed mice. A significant recovery of proliferative responsiveness was also observed when splenocytes were obtained from DHEA- or MET-treated immunosuppressed mice. In addition, these treatments restored the hypersensitivity response in immunosuppressed mice. Finally, although neither DHEA nor MET improved the reduced CD4 lymphocyte count in spleen from immunosuppressed mice, both treatments promoted spleen architecture reorganization, partially restoring the distinct cellular components and their localization in the spleen. The results from this study indicate that DHEA and MET could play an important role in the restoration of both adaptive humoral and cellular immune response in LPS-immunosuppressed mice, reinforcing the concept of a central involvement of endogenous glucocorticoids on this phenomenon.


Subject(s)
Adaptive Immunity/drug effects , Dehydroepiandrosterone/pharmacology , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Immunosuppression Therapy , Lipopolysaccharides/pharmacology , Metyrapone/pharmacology , Animals , CD4 Lymphocyte Count , Cell Proliferation , Glucocorticoids/antagonists & inhibitors , Hypersensitivity, Delayed/immunology , Macrophages, Peritoneal/drug effects , Mice , Mice, Inbred BALB C , Phagocytosis/drug effects , Receptors, Glucocorticoid/antagonists & inhibitors , Respiratory Burst/drug effects , Spleen/cytology , Spleen/immunology
6.
Clin Sci (Lond) ; 126(5): 355-65, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24053409

ABSTRACT

Secondary infections due to post-sepsis immunosuppression are a major cause of death in patients with sepsis. Strategies aimed at restoring immune functions offer a new perspective in the treatment of sepsis. In the present study, we used LPS (lipopolysaccharide)-immunosuppressed mice to analyse the effects of ATRA (all-trans retinoic acid) on different immune parameters. The IS (immunocompromised) group had decreased lymphocyte and increased MDSC (myeloid-derived suppressor cell) counts in lymph nodes. They also had an impaired in vitro T-cell proliferation, mediated by MDSCs. ATRA administration restored T-cell proliferation, which was associated with a decreased number of live MDSCs. The IS group treated with ATRA had an increased number of CD4+ and CD8+ T-cells. ATRA partially improved the primary humoral immune response, even when immunosuppression was established first and ATRA was administered subsequently. Our results demonstrate that ATRA restores immunocompetence by modulating the number of leucocytes and the survival of MDSCs, and thus represents an additional potential strategy in the treatment of the immunosuppressive state of sepsis.


Subject(s)
Immunocompetence/drug effects , Immunosuppression Therapy , Lipopolysaccharides/pharmacology , Models, Animal , Tretinoin/pharmacology , Animals , Cell Proliferation/drug effects , Flow Cytometry , Mice , Mice, Inbred BALB C , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
7.
Cytokine ; 62(3): 426-32, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23602200

ABSTRACT

Large amounts of anti-inflammatory mediators, such as interleukin (IL)-10, are produced and found early in the course of sepsis. We explore the role of IL-10 on neutrophil (PMN) activation/function using an in vitro model. Isolated human PMN were pre-incubated with lipopolysaccharide (LPS) and/or IL-10 for 18h. Subsequently, a second LPS exposure was performed and CD11b and CD66b up-regulation, and the reactive oxygen species (ROS) generation were measured 2h later. We found that IL-10 prevented PMN activation and the secretion of TNF-α and IL-8 induced by the first LPS contact. In the absence of IL-10, a second LPS exposure induced additive effects that were prevented by IL-10. Only ROS generation was highly affected by the blockade of PMN-secreted TNF-α or IL-8. Additionally, IL-10 prevented other possible mechanisms of LPS priming. Therefore, IL-10 modulates PMN activation preventing autocrine activating loops and priming mechanisms, rendering PMN less responsive to a second LPS exposure.


Subject(s)
Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , Neutrophil Activation/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Autocrine Communication/drug effects , Cell Degranulation/drug effects , Cell Separation , Humans , Interleukin-8/metabolism , Neutrophils/physiology , Reactive Oxygen Species/metabolism , Teichoic Acids/pharmacology
8.
PLoS One ; 8(2): e57128, 2013.
Article in English | MEDLINE | ID: mdl-23451160

ABSTRACT

Shiga toxins (Stx) are the main virulence factors in enterohemorrhagic Escherichia coli (EHEC) infections, causing diarrhea and hemolytic uremic syndrome (HUS). The genes encoding for Shiga toxin-2 (Stx2) are located in a bacteriophage. The toxin is formed by a single A subunit and five B subunits, each of which has its own promoter sequence. We have previously reported the expression of the B subunit within the eukaryotic environment, probably driven by their own promoter. The aim of this work was to evaluate the ability of the eukaryotic machinery to recognize stx2 sequences as eukaryotic-like promoters. Vero cells were transfected with a plasmid encoding Stx2 under its own promoter. The cytotoxic effect on these cells was similar to that observed upon incubation with purified Stx2. In addition, we showed that Stx2 expression in Stx2-insensitive BHK eukaryotic cells induced drastic morphological and cytoskeletal changes. In order to directly evaluate the capacity of the wild promoter sequences of the A and B subunits to drive protein expression in mammalian cells, GFP was cloned under eukaryotic-like putative promoter sequences. GFP expression was observed in 293T cells transfected with these constructions. These results show a novel and alternative way to synthesize Stx2 that could contribute to the global understanding of EHEC infections with immediate impact on the development of treatments or vaccines against HUS.


Subject(s)
Promoter Regions, Genetic , Shiga Toxin/genetics , Animals , Base Sequence , Cell Differentiation , Cell Line , Chlorocebus aethiops , Cricetinae , Sequence Homology, Nucleic Acid , Vero Cells
9.
PLoS Pathog ; 8(3): e1002632, 2012.
Article in English | MEDLINE | ID: mdl-22479186

ABSTRACT

The hemolytic uremic syndrome (HUS) is characterized by hemolytic anemia, thrombocytopenia and renal dysfunction. The typical form of HUS is generally associated with infections by Gram-negative Shiga toxin (Stx)-producing Escherichia coli (STEC). Endothelial dysfunction induced by Stx is central, but bacterial lipopolysaccharide (LPS) and neutrophils (PMN) contribute to the pathophysiology. Although renal failure is characteristic of this syndrome, neurological complications occur in severe cases and is usually associated with death. Impaired blood-brain barrier (BBB) is associated with damage to cerebral endothelial cells (ECs) that comprise the BBB. Astrocytes (ASTs) are inflammatory cells in the brain and determine the BBB function. ASTs are in close proximity to ECs, hence the study of the effects of Stx1 and LPS on ASTs, and the influence of their response on ECs is essential. We have previously demonstrated that Stx1 and LPS induced activation of rat ASTs and the release of inflammatory factors such as TNF-α, nitric oxide and chemokines. Here, we demonstrate that rat ASTs-derived factors alter permeability of ECs with brain properties (HUVECd); suggesting that functional properties of BBB could also be affected. Additionally, these factors activate HUVECd and render them into a proagregant state promoting PMN and platelets adhesion. Moreover, these effects were dependent on ASTs secreted-TNF-α. Stx1 and LPS-induced ASTs response could influence brain ECs integrity and BBB function once Stx and factors associated to the STEC infection reach the brain parenchyma and therefore contribute to the development of the neuropathology observed in HUS.


Subject(s)
Astrocytes/drug effects , Endothelium, Vascular/drug effects , Lipopolysaccharides/pharmacology , Shiga Toxin 1/toxicity , Tumor Necrosis Factor-alpha/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Blood-Brain Barrier , Brain/blood supply , Cell Membrane Permeability/drug effects , Cell Survival/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Endothelium, Vascular/pathology , Human Umbilical Vein Endothelial Cells , Humans , Rats
10.
J Clin Immunol ; 32(3): 622-31, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22228570

ABSTRACT

PURPOSE: The interaction of Shiga toxin (Stx) and/or lipopolysaccharide (LPS) with monocytes (Mo) may be central to the pathogenesis of hemolytic uremic syndrome (HUS), providing the cytokines necessary to sensitize endothelial cells to Stx action. We have previously demonstrated phenotypical alterations in Mo from HUS patients, including increased number of CD16+ Mo. Our aim was to investigate cytokine production in Mo from HUS patients. METHODS: We evaluated TNF-α and IL-10 intracellular contents and secretion in the different Mo subsets in mild (HUS 1) and moderate/severe (HUS 2 + 3) patients. As controls, we studied healthy (HC) and infected children (IC). We also studied Mo responsive capacity towards LPS, measuring the modulation of Mo surface molecules and cytokine production. RESULTS: In basal conditions, the intracellular measurement of TNF-α and IL-10 revealed that the highest number of cytokine-producing Mo was found in HUS 2 + 3 and IC, whereas LPS caused a similar increase in TNF-α and IL-10-producing Mo for all groups. However, when evaluating the release of TNF-α and IL-10, we found a diminished secretion capacity in the entire HUS group and IC compared to HC in basal and LPS conditions. Similarly, a lower Mo response to LPS in HUS 2 + 3 and IC groups was observed when surface markers were studied. CONCLUSION: These results indicate that Mo from severe cases of HUS, similar to IC but different to mild HUS cases, present functional changes in Mo subpopulations and abnormal responses to LPS.


Subject(s)
Hemolytic-Uremic Syndrome/immunology , Interleukin-10/immunology , Monocytes/immunology , Tumor Necrosis Factor-alpha/immunology , Child , Child, Preschool , Female , Humans , Infant , Interleukin-10/blood , Lipopolysaccharides/immunology , Male , Tumor Necrosis Factor-alpha/blood
11.
Cancer Res ; 71(22): 7113-24, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22084446

ABSTRACT

Concomitant tumor resistance (CR) is a phenomenon originally described in 1906 in which a tumor-bearing host is resistant to the growth of secondary tumor implants and metastasis. Although recent studies have indicated that T-cell-dependent processes mediate CR in hosts bearing immunogenic small tumors, manifestations of CR induced by immunogenic and nonimmunogenic large tumors have been associated with an elusive serum factor. In this study, we identify this serum factor as tyrosine in its meta and ortho isoforms. In three different murine models of cancer that generate CR, both meta-tyrosine and ortho-tyrosine inhibited tumor growth. In addition, we showed that both isoforms of tyrosine blocked metastasis in a fourth model that does not generate CR but is sensitive to CR induced by other tumors. Mechanistic studies showed that the antitumor effects of the tyrosine isoforms were mediated, in part, by early inhibition of mitogen-activated protein/extracellular signal-regulated kinase pathway and inactivation of STAT3, potentially driving tumor cells into a state of dormancy. By revealing a molecular basis for the classical phenomenon of CR, our findings may stimulate new generalized approaches to limit the development of metastases that arise after resection of primary tumors, an issue of pivotal importance to oncologists and their patients.


Subject(s)
Neoplasm Transplantation/immunology , Neoplasms, Experimental/immunology , Tyrosine/physiology , Animals , Chromatography, High Pressure Liquid , Disease Resistance , Extracellular Signal-Regulated MAP Kinases/physiology , Female , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/pathology , Phenylalanine/pharmacology , STAT3 Transcription Factor/physiology
12.
PLoS One ; 6(4): e19136, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21559455

ABSTRACT

Shiga toxin (Stx)-producing Escherichia coli (STEC) infection is associated with a broad spectrum of clinical manifestations that include diarrhea, hemorrhagic colitis, and hemolytic uremic syndrome (HUS). Systemic Stx toxemia is considered to be central to the genesis of HUS. Distinct methods have been used to evaluate anti-Stx response for immunodiagnostic or epidemiological analysis of HUS cases. The development of enzyme-linked immunosorbent assay (ELISA) and western blot (WB) assay to detect the presence of specific antibodies to Stx has introduced important advantages for serodiagnosis of HUS. However, application of these methods for seroepidemiological studies in Argentina has been limited. The aim of this work was to develop an ELISA to detect antibodies against the B subunit of Stx2, and a WB to evaluate antibodies against both subunits of Stx2 and Stx1, in order to analyze the pertinence and effectiveness of these techniques in the Argentinean population. We studied 72 normal healthy children (NHC) and 105 HUS patients of the urban pediatric population from the surrounding area of Buenos Aires city. Using the WB method we detected 67% of plasma from NHC reactive for Stx2, but only 8% for Stx1. These results are in agreement with the broad circulation of Stx2-expressing STEC in Argentina and the endemic behavior of HUS in this country. Moreover, the simultaneous evaluation by the two methods allowed us to differentiate acute HUS patients from NHC with a great specificity and accuracy, in order to confirm the HUS etiology when pathogenic bacteria were not isolated from stools.


Subject(s)
Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/microbiology , Shiga Toxins/immunology , Antibodies/immunology , Argentina , Case-Control Studies , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay/methods , Female , Follow-Up Studies , Hemolytic-Uremic Syndrome/drug therapy , Humans , Male , Serologic Tests , Shiga Toxins/chemistry , Time Factors , Treatment Outcome
13.
Exp Hematol ; 39(7): 763-72, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21549176

ABSTRACT

OBJECTIVE: Megakaryo/thrombopoiesis is a complex process regulated by multiple signals provided by the bone marrow microenvironment. Because macrophages are relevant components of the bone marrow stroma and their activation induces an upregulation of molecules that can regulate hematopoiesis, we analyzed the impact of these cells on the control of megakaryocyte development and platelet biogenesis. MATERIALS AND METHODS: The different stages of megakaryo/thrombopoiesis were analyzed by flow cytometry using an in vitro model of human cord blood CD34(+) cells stimulated with thrombopoietin in either a transwell system or conditioned media from monocyte-derived macrophages isolated from peripheral blood. Cytokines secreted from macrophages were characterized by protein array and enzyme-linked immunosorbent assay. RESULTS: Resting macrophages released soluble factors that promoted megakaryocyte growth, cell ploidy, a size increase, proplatelet production, and platelet release. Lipopolysaccharide stimulation triggered the secretion of cytokines that exerted opposite effects together with a dramatic switch of CD34(+) commitment to the megakaryocytic lineage toward the myeloid lineage. Neutralization of interleukin-8 released by stimulated macrophages partially reversed the inhibition of megakaryocyte growth. Activation of nuclear factor κB had a major role in the synthesis of molecules involved in the megakaryocyte inhibition mediated by lipopolysaccharide-stimulated macrophages. CONCLUSIONS: Our study extends our understanding about the role of the bone marrow microenvironment in the regulation of megakaryo/thrombopoiesis by showing that soluble factors derived from macrophages positively or negatively control megakaryocyte growth, differentiation, maturation, and their ability to produce platelets.


Subject(s)
Cytokines/pharmacology , Macrophages/metabolism , Paracrine Communication , Thrombopoiesis/drug effects , Antigens, CD34/metabolism , Blood Platelets/cytology , Blood Platelets/drug effects , Blood Platelets/metabolism , Cells, Cultured , Chemokines/pharmacology , Coculture Techniques , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Enzyme-Linked Immunosorbent Assay , Female , Fetal Blood/cytology , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Lipopolysaccharides/pharmacology , Macrophages/cytology , Megakaryocytes/cytology , Megakaryocytes/drug effects , Megakaryocytes/metabolism , NF-kappa B/metabolism , Thrombopoietin/pharmacology
14.
Infect Immun ; 79(3): 1280-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21173307

ABSTRACT

Severe sepsis is associated with early release of inflammatory mediators that contribute to the morbidity and mortality observed during the first stages of this syndrome. Although sepsis is a deadly, acute disease, high mortality rates have been observed in patients displaying evidence of sepsis-induced immune deactivation. Although the contribution of experimental models to the knowledge of pathophysiological and therapeutic aspects of human sepsis is undeniable, most of the current studies using animal models have focused on the acute, proinflammatory phase. We developed a murine model that reproduces the early acute phases but also the long-term consequences of human sepsis. We induced polymicrobial acute peritonitis (AP) by establishing a surgical connection between the cecum and the peritoneum, allowing the exit of intestinal bacteria. Using this model, we observed an acute phase with high mortality, leukopenia, increased interleukin-6 levels, bacteremia, and neutrophil activation. A peak of leukocytosis on day 9 or 10 revealed the persistence of the infection within the lung and liver, with inflammatory hepatic damage being shown by histological examination. Long-term (20 days) derangements in both innate and adaptive immune responses were found, as demonstrated by impaired systemic tumor necrosis factor alpha production in response to an inflammatory stimulus; a decreased primary humoral immune response and T cell proliferation, associated with an increased number of myeloid suppressor cells (Gr-1(+) CD11b(+)) in the spleen; and a low clearance capacity. This model provides a good approach to attempt novel therapeutic interventions directed to augmenting host immunity during late sepsis.


Subject(s)
Disease Models, Animal , Peritonitis/complications , Peritonitis/immunology , Sepsis/etiology , Sepsis/immunology , Animals , Cecum/microbiology , Cecum/surgery , Cytokines/immunology , Inflammation/immunology , Mice , Mice, Inbred BALB C , Peritoneum/surgery , Peritonitis/microbiology , Sepsis/microbiology
15.
Immunology ; 129(4): 600-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20102414

ABSTRACT

The membrane-anchored form of the chemokine fractalkine (CX(3)CL1) has been identified as a novel adhesion molecule that interacts with its specific receptor (CX(3)CR1) expressed in monocytes, T cells and natural killer cells to induce adhesion. In addition, CX(3)CL1 can be cleaved from the cell membrane to induce chemotaxis of CX(3)CR1-expressing leucocytes. Recently, marked variations in CX(3)CR1 monocyte expression have been observed during several pathological conditions. Regulation of CX(3)CR1 in monocytes during basal or inflammatory/anti-inflammatory conditions is poorly understood. The aim of this study was therefore to examine CX(3)CR1 expression during monocyte maturation and the effect of soluble mediators on this process. We found that basal expression of CX(3)CR1 in fresh monocytes was reduced during culture, and that lipopolysacchairde accelerated this effect. In contrast, interleukin-10 and interferon-gamma treatment abrogated CX(3)CR1 down-modulation, through a phosphatidylinositol 3 kinase-dependent pathway. Most importantly, CX(3)CR1 membrane expression correlated with monocyte CX(3)CL1-dependent function. Taken together, our data demonstrate that CX(3)CR1 expression in monocytes can be modulated, and suggest that alterations in their environment are able to influence CX(3)CL1-dependent functions, such as chemotaxis and adhesion, leading to changes in the kinetics, composition and/or functional status of the leucocyte infiltrate.


Subject(s)
Interferon-gamma/pharmacology , Interleukin-10/pharmacology , Monocytes/drug effects , Monocytes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Chemokine/biosynthesis , CX3C Chemokine Receptor 1 , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Interferon-gamma/immunology , Interleukin-10/immunology , Monocytes/immunology , Receptors, Chemokine/immunology , Time Factors
16.
Infect Immun ; 78(3): 1193-201, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20008539

ABSTRACT

Hemolytic-uremic syndrome (HUS) is generally caused by Shiga toxin (Stx)-producing Escherichia coli. Endothelial dysfunction mediated by Stx is a central aspect in HUS development. However, inflammatory mediators such as bacterial lipopolysaccharide (LPS) and polymorphonuclear neutrophils (PMN) contribute to HUS pathophysiology by potentiating Stx effects. Acute renal failure is the main feature of HUS, but in severe cases, patients can develop neurological complications, which are usually associated with death. Although the mechanisms of neurological damage remain uncertain, alterations of the blood-brain barrier associated with brain endothelial injury is clear. Astrocytes (ASTs) are the most abundant inflammatory cells of the brain that modulate the normal function of brain endothelium and neurons. The aim of this study was to evaluate the effects of Stx type 1 (Stx1) alone or in combination with LPS in ASTs. Although Stx1 induced a weak inflammatory response, pretreatment with LPS sensitized ASTs to Stx1-mediated effects. Moreover, LPS increased the level of expression of the Stx receptor and its internalization. An early inflammatory response, characterized by the release of tumor necrosis factor alpha (TNF-alpha) and nitric oxide and PMN-chemoattractant activity, was induced by Stx1 in LPS-sensitized ASTs, whereas activation, evidenced by higher levels of glial fibrillary acid protein and cell death, was induced later. Furthermore, increased adhesion and PMN-mediated cytotoxicity were observed after Stx1 treatment in LPS-sensitized ASTs. These effects were dependent on NF-kappaB activation or AST-derived TNF-alpha. Our results suggest that TNF-alpha is a pivotal effector molecule that amplifies Stx1 effects on LPS-sensitized ASTs, contributing to brain inflammation and leading to endothelial and neuronal injury.


Subject(s)
Astrocytes/drug effects , Astrocytes/immunology , Lipopolysaccharides/immunology , Shiga Toxin 1/immunology , Shiga Toxin 1/toxicity , Tumor Necrosis Factor-alpha/immunology , Animals , Cells, Cultured , Escherichia coli/pathogenicity , Inflammation Mediators/metabolism , Nitric Oxide/metabolism , Rats
17.
Cancer Immunol Immunother ; 57(5): 701-18, 2008 May.
Article in English | MEDLINE | ID: mdl-17962945

ABSTRACT

Although animals can be immunized against the growth of some tumor implants, most of the attempts to use immunotherapy to cause the regression of animal and human tumors once they have become established have been disappointing even when strongly immunogenic tumors were used as target. In this paper, we demonstrate that the failure to achieve an efficient immunological treatment against an established strongly immunogenic murine fibrosarcoma was paralleled with the emergence of a state of immunological unresponsiveness (immunological eclipse) against tumor antigens observed when the tumor surpassed the critical size of 500 mm(3). In turn, the onset of the immunological eclipse was coincidental with the onset of a systemic inflammatory condition characterized by a high number of circulating and splenic polymorphonucleated neutrophils (PMN) displaying activation and Gr1(+)Mac1(+) phenotype and an increasing serum concentration of the pro-inflammatory cytokines TNF-alpha, IL-1beta and IL-6 cytokines and C-reactive protein (CRP) and serum A amyloid (SAA) phase acute proteins. Treatment of tumor-bearing mice with a single low dose (0.75 mg/kg) of the synthetic corticoid dexamethasone (DX) significantly reduced all the systemic inflammatory parameters and simultaneously reversed the immunological eclipse, as evidenced by the restoration of specific T-cell-dependent concomitant immunity, ability of spleen cells to transfer anti-tumor activity and recovery of T-cell signal transduction molecules. Two other anti-inflammatory treatments by using indomethacin or dimeric TNF-alpha receptor, also partially reversed the immunological eclipse although the effect was not as striking as that observed with DX. The reversion of the immunological eclipse was not enough on its own to inhibit the primary growing tumor. However, when we used the two-step strategy of inoculating DX to reverse the eclipse and then dendritic cells loaded with tumor antigens (DC) as an immunization booster, a significant inhibition of the growth of both established tumors and remnant tumor cells after excision of large established tumors was observed, despite the fact that the vaccination alone (DC) had no effect or even enhanced tumor growth in certain circumstances. The two-step strategy of tumor immunotherapy that we present is based on the rationale that it is necessary to eliminate or ameliorate the immunological eclipse as a precondition to allow an otherwise ineffective anti-tumor immunological therapy to have a chance to be successful.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Dendritic Cells/transplantation , Immune Tolerance/drug effects , Immunotherapy/methods , Inflammation/drug therapy , Neoplasms, Experimental/therapy , Animals , Blotting, Western , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Dexamethasone/therapeutic use , Female , Flow Cytometry , Male , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/immunology
18.
J Immunol ; 179(1): 673-81, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17579090

ABSTRACT

The interaction between immune complexes (IC) and the receptors for the Fc portion of IgG (FcgammaRs) triggers regulatory and effector functions in the immune system. In this study, we investigated the effects of IC on differentiation, maturation, and functions of human monocyte-derived dendritic cells (DC). When IC were added on day 0, DC generated on day 6 (IC-DC) showed lower levels of CD1a and increased expression of CD14, MHC class II, and the macrophage marker CD68, as compared with normally differentiated DC. The use of specific blocking FcgammaR mAbs indicated that the effect of IC was exerted mainly through their interaction with FcgammaRI and to a lesser extend with FcgammaRII. Immature IC-DC also expressed higher levels of CD83, CD86, and CD40 and the expression of these maturation markers was not further regulated by LPS. The apparent lack of maturation following TLR stimulation was associated with a decreased production of IL-12, normal secretion of IL-10 and CCL22, and increased production of CXCL8 and CCL2. IC-DC displayed low endocytic activity and a reduced ability to induce allogeneic T cell proliferation both at basal and LPS-stimulated conditions. Altogether, these data reveal that IC strongly affect DC differentiation and maturation. Skewing of DC function from Ag presentation to a proinflammatory phenotype by IC resembles the state of activation observed in DC obtained from patients with chronic inflammatory autoimmune disorders, such as systemic lupus erythematosus disease and arthritis. Therefore, the altered maturation of DC induced by IC may be involved in the pathogenesis of autoimmune diseases.


Subject(s)
Antigen-Antibody Complex/physiology , Cell Differentiation/immunology , Cell Lineage/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Growth Inhibitors/physiology , Monocytes/cytology , Monocytes/immunology , Animals , Antigen-Antibody Complex/blood , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/pathology , Dose-Response Relationship, Immunologic , Growth Inhibitors/blood , Humans , Immunoglobulin G/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Monocytes/metabolism , Monocytes/pathology , Rabbits , Receptors, IgG/antagonists & inhibitors , Receptors, IgG/biosynthesis , Receptors, IgG/physiology , T-Lymphocytes/immunology , Time Factors
19.
Pediatr Res ; 61(1): 123-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17211153

ABSTRACT

Hemolytic Uremic Syndrome (HUS) is the main cause of acute renal failure in children. The high percentage of patients who develop long-term sequelae constitutes an important medical concern. The identification of parameters that correlate with the degree of renal failure may be useful to plan the best treatment soon after hospitalization. Here, we investigated the functional state of neutrophils (PMN) from HUS patients on admission, before dialysis and/or transfusion, in relation to the severity of renal impairment reached during the acute period (AP). We found that all PMN activation parameters measured in severe cases of HUS (HUS AP3) were statistically lower comparing to children with mild cases of HUS (HUS AP1). As HUS PMN phenotype and dysfunction is compatible with that of cells undergoing cell death, we also studied spontaneous apoptosis. Not only were HUS PMN not apoptotic, but HUS AP3 PMN showed an increased survival. Almost all phenotypic and functional parameters measured on PMN correlated with severity. Our results revealed a marked deactivation of PMN in severe cases of HUS, and suggest that studying the functional state of PMN could be of prognostic value.


Subject(s)
Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/physiopathology , Neutrophils/physiology , Child, Preschool , Female , Humans , Infant , Male , Prognosis
20.
J Immunol ; 178(1): 436-45, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17182582

ABSTRACT

Several environmental factors can differentially regulate monocyte and macrophage response patterns, resulting in the display of distinct functional phenotypes. Galectin-1, an endogenous lectin found at peripheral lymphoid organs and inflammatory sites, has shown immunoregulatory activity in vivo in experimental models of autoimmunity and cancer. Whereas compelling evidence has been accumulated regarding the effects of galectin-1 on T cell fate, limited information is available on how galectin-1 may impact other immune cell types. In the present study, we report a novel role for galectin-1 in the regulation of monocyte and macrophage physiology. Treatment with galectin-1 in vitro differentially regulates constitutive and inducible FcgammaRI expression on human monocytes and FcgammaRI-dependent phagocytosis. In addition, galectin-1 inhibits IFN-gamma-induced MHC class II (MHC-II) expression and MHC-II-dependent Ag presentation in a dose-dependent manner. These regulatory effects were also evident in mouse macrophages recruited in response to inflammatory stimuli following treatment with recombinant galectin-1 and further confirmed in galectin-1-deficient mice. Investigation of the mechanisms involved in these functions showed that galectin-1 does not affect survival of human monocytes, but rather influences FcgammaRI- and MHC-II-dependent functions through active mechanisms involving modulation of an ERK1/2-dependent pathway. Our results provide evidence of a novel unrecognized role for galectin-1 in the control of monocyte/macrophage physiology with potential implications at the crossroad of innate and adaptive immunity.


Subject(s)
Galectin 1/physiology , Macrophages/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/immunology , Receptors, IgG/metabolism , Animals , Antigen Presentation , Apoptosis , Cell Survival , Galectin 1/pharmacology , Histocompatibility Antigens Class II/analysis , Histocompatibility Antigens Class II/metabolism , Humans , Immunity, Innate , Macrophage Activation , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Monocytes/drug effects , Phagocytosis , Receptors, IgG/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...