Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(1): e2210490120, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36574651

ABSTRACT

γδ T cells are involved in the control of Staphylococcus aureus infection, but their importance in protection compared to other T cells is unclear. We used a mouse model of systemic S. aureus infection associated with high bacterial load and persistence in the kidney. Infection caused fulminant accumulation of γδ T cells in the kidney. Renal γδ T cells acquired tissue residency and were maintained in high numbers during chronic infection. At day 7, up to 50% of renal γδ T cells produced IL-17A in situ and a large fraction of renal γδ T cells remained IL-17A+ during chronic infection. Controlled depletion revealed that γδ T cells restricted renal S. aureus replication in the acute infection and provided protection during chronic renal infection and upon reinfection. Our results demonstrate that kidney-resident γδ T cells are nonredundant in limiting local S. aureus growth during chronic infection and provide enhanced protection against reinfection.


Subject(s)
Interleukin-17 , Staphylococcal Infections , Mice , Animals , Staphylococcus aureus , Receptors, Antigen, T-Cell, gamma-delta , Persistent Infection , Reinfection , Kidney , Mice, Inbred C57BL
2.
Int J Mol Sci ; 25(8)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38673763

ABSTRACT

Chronic ethanol exposure often triggers neuroinflammation in the brain's reward system, potentially promoting the drive for ethanol consumption. A main marker of neuroinflammation is the microglia-derived monocyte chemoattractant protein 1 (MCP1) in animal models of alcohol use disorder in which ethanol is forcefully given. However, there are conflicting findings on whether MCP1 is elevated when ethanol is taken voluntarily, which challenges its key role in promoting motivation for ethanol consumption. Here, we studied MCP1 mRNA levels in areas implicated in consumption motivation-specifically, the prefrontal cortex, hippocampus, and striatum-as well as in the cerebellum, a brain area highly sensitive to ethanol, of C57BL/6 mice subjected to intermittent and voluntary ethanol consumption for two months. We found a significant increase in MCP1 mRNA levels in the cerebellum of mice that consumed ethanol compared to controls, whereas no significant changes were observed in the prefrontal cortex, hippocampus, or striatum or in microglia isolated from the hippocampus and striatum. To further characterize cerebellar neuroinflammation, we measured the expression changes in other proinflammatory markers and chemokines, revealing a significant increase in the proinflammatory microRNA miR-155. Notably, other classical proinflammatory markers, such as TNFα, IL6, and IL-1ß, remained unaltered, suggesting mild neuroinflammation. These results suggest that the onset of neuroinflammation in motivation-related areas is not required for high voluntary consumption in C57BL/6 mice. In addition, cerebellar susceptibility to neuroinflammation may be a trigger to the cerebellar degeneration that occurs after chronic ethanol consumption in humans.


Subject(s)
Alcohol Drinking , Cerebellum , Chemokine CCL2 , Corpus Striatum , Ethanol , Hippocampus , Mice, Inbred C57BL , Prefrontal Cortex , Animals , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/pathology , Mice , Hippocampus/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Cerebellum/metabolism , Cerebellum/drug effects , Cerebellum/pathology , Male , Corpus Striatum/metabolism , Corpus Striatum/pathology , Corpus Striatum/drug effects , Ethanol/adverse effects , Alcohol Drinking/adverse effects , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/pathology , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , Inflammation/metabolism , Inflammation/pathology , Inflammation/chemically induced
3.
Int J Mol Sci ; 25(4)2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38396750

ABSTRACT

Acute-on-chronic liver failure (ACLF) is a syndrome marked by sudden liver function decline and multiorgan failure, predominantly acute kidney injury (AKY), in patients with chronic liver disease. Unregulated inflammation is a hallmark of ACLF; however, the key drivers of ACLF are not fully understood. This study explores the therapeutic properties of human mesenchymal stem cell (MSC) secretome, particularly focusing on its enhanced anti-inflammatory and pro-regenerative properties after the in vitro preconditioning of the cells. We evaluated the efficacy of the systemic administration of MSC secretome in preventing liver failure and AKI in a rat ACLF model where chronic liver disease was induced using by the administration of porcine serum, followed by D-galN/LPS administration to induce acute failure. After ACLF induction, animals were treated with saline (ACLF group) or MSC-derived secretome (ACLF-secretome group). The study revealed that MSC-secretome administration strongly reduced liver histological damage in the ACLF group, which was correlated with higher hepatocyte proliferation, increased hepatic and systemic anti-inflammatory molecule levels, and reduced neutrophil and macrophage infiltration. Additionally, renal examination revealed that MSC-secretome treatment mitigated tubular injuries, reduced apoptosis, and downregulated injury markers. These improvements were linked to increased survival rates in the ACLF-secretome group, endorsing MSC secretomes as a promising therapy for multiorgan failure in ACLF.


Subject(s)
Acute-On-Chronic Liver Failure , Humans , Rats , Animals , Swine , Acute-On-Chronic Liver Failure/therapy , Secretome , Stem Cells , Anti-Inflammatory Agents
4.
BMC Med ; 20(1): 216, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35676738

ABSTRACT

BACKGROUND: Chile was severely affected by COVID19 outbreaks but was also one of the first countries to start a nationwide program to vaccinate against the disease. Furthermore, Chile became one of the fastest countries to inoculate a high percentage of the target population and implemented homologous and heterologous booster schemes in late 2021 to prevent potential immunological waning. The aim of this study is to compare the immunogenicity and time course of the humoral response elicited by the CoronaVac vaccine in combination with homologous versus heterologous boosters. METHODS: We compared the immunogenicity of two doses of CoronaVac and BNT162b2 vaccines and one homologous or heterologous booster through an ELISA assay directed against the ancestral spike protein of SARS-CoV-2. Sera were collected from individuals during the vaccination schedule and throughout the implementation of homologous and heterologous booster programs in Chile. RESULTS: Our findings demonstrate that a two-dose vaccination scheme with CoronaVac induces lower levels of anti-SARS-CoV-2 spike antibodies than BNT162b2 in a broad age range (median age 42 years; interquartile range (IQR) 27-61). Furthermore, antibody production declines with time in individuals vaccinated with CoronaVac and less noticeably, with BNT162b2. Analysis of booster schemes revealed that individuals vaccinated with two doses of CoronaVac generate immunological memory against the SARS-CoV-2 ancestral strain, which can be re-activated with homologous or heterologous (BNT162b2 and ChAdOx1) boosters. Nevertheless, the magnitude of the antibody response with the heterologous booster regime was considerably higher (induction fold BNT162b2: 11.2x; ChAdoX1; 12.4x; CoronaVac: 6.0x) than the responses induced by the homologous scheme. Both homologous and heterologous boosters induced persistent humoral responses (median 122 days, IQR (108-133)), although heterologous boosters remained superior in activating a humoral response after 100 days. CONCLUSIONS: Two doses of CoronaVac induces antibody titers against the SARS-CoV-2 ancestral strain which are lower in magnitude than those induced by the BNT162b2 vaccine. However, the response induced by CoronaVac can be greatly potentiated with a heterologous booster scheme with BNT162b2 or ChAdOx1 vaccines. Furthermore, the heterologous and homologous booster regimes induce a durable antibody response which does not show signs of decay 3 months after the booster dose.


Subject(s)
COVID-19 , SARS-CoV-2 , Adult , Antibodies, Viral , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines , Chile/epidemiology , Humans
5.
Mol Biol Rep ; 49(6): 4193-4204, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35211864

ABSTRACT

BACKGROUND: Several studies have demonstrated the contribution of innate immune cells, including macrophages, in promoting systemic lupus erythematosus (SLE). Macrophages, one of the most abundant cell populations in the peritoneal cavity, are considered multifunctional cells with phenotypic plasticity. However, the functional properties of peritoneal macrophages in steady-state and during the progression of SLE remain poorly defined. METHODS AND RESULTS: Using the [NZB × NZW]F1 (BWF1) murine model of SLE, we analyzed the phenotype and function of peritoneal macrophages during the disease's onset. We found a higher frequency of peritoneal macrophages and B1a cells in BWF1-diseased mice than age-matched controls. Additionally, macrophages from diseased animals expressed lower levels of CD206, MHC-II, and Sirpα. RNAseq analysis identified 286 differentially expressed genes in peritoneal macrophages from diseased-BWF1 mice compared to control mice. Functional experiments demonstrate that peritoneal macrophages from diseased-BWF1 mice secrete higher levels of pro-inflammatory cytokines when activated with TLR7 and TLR9 agonists, and they were less efficient in suppressing the activation and proliferation of peritoneal LPS-activated B cells. These data demonstrate that peritoneal macrophages from BWF1-diseased mice present phenotypic and functional alterations shifting to a more pro-inflammatory state. CONCLUSIONS: The increase of macrophages with an altered phenotype and function together with the accumulation of B1a cells in the peritoneal cavity of diseased-BWF1 mice may promote the progression of the disease. Advancing awareness of the role and phenotype of peritoneal macrophages in SLE may contribute to a better understanding of these types of diseases and the development of novel therapies.


Subject(s)
Lupus Erythematosus, Systemic , Macrophages, Peritoneal , Animals , B-Lymphocytes , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred NZB , Phenotype
6.
Int J Mol Sci ; 21(14)2020 Jul 13.
Article in English | MEDLINE | ID: mdl-32668623

ABSTRACT

The P2X7 receptor is a ligand-gated, cation-selective channel whose main physiological ligand is ATP. P2X7 receptor activation may also be triggered by ARTC2.2-dependent ADP ribosylation in the presence of extracellular NAD. Upon activation, this receptor induces several responses, including the influx of calcium and sodium ions, phosphatidylserine externalization, the formation of a non-selective membrane pore, and ultimately cell death. P2X7 receptor activation depends on the availability of extracellular nucleotides, whose concentrations are regulated by the action of extracellular nucleotidases such as CD39 and CD38. The P2X7 receptor has been extensively studied in the context of the immune response, and it has been reported to be involved in inflammasome activation, cytokine production, and the migration of different innate immune cells in response to ATP. In adaptive immune responses, the P2X7 receptor has been linked to T cell activation, differentiation, and apoptosis induction. In this review, we will discuss the evidence of the role of the P2X7 receptor on T cell differentiation and in the control of T cell responses in inflammatory conditions.


Subject(s)
Receptors, Purinergic P2X7/physiology , T-Lymphocyte Subsets/immunology , ADP-ribosyl Cyclase 1/physiology , Adenosine Triphosphate/physiology , Animals , Antigens, CD/physiology , Apoptosis/physiology , Apyrase/physiology , Cell Differentiation/physiology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Inflammasomes/metabolism , Ion Channel Gating/physiology , Lymphocyte Activation/physiology , Mice , Nucleotides/metabolism , Phosphatidylserines/metabolism , Rats , Receptors, Purinergic P2X7/chemistry , Receptors, Purinergic P2X7/drug effects , Receptors, Purinergic P2X7/genetics , Signal Transduction/physiology , Structure-Activity Relationship , T-Lymphocyte Subsets/metabolism
7.
J Neuroinflammation ; 16(1): 258, 2019 Dec 06.
Article in English | MEDLINE | ID: mdl-31810491

ABSTRACT

BACKGROUND: Neuroinflammation constitutes a pathogenic process leading to neurodegeneration in several disorders, including Alzheimer's disease, Parkinson's disease (PD) and sepsis. Despite microglial cells being the central players in neuroinflammation, astrocytes play a key regulatory role in this process. Our previous results indicated that pharmacologic-antagonism or genetic deficiency of dopamine receptor D3 (DRD3) attenuated neuroinflammation and neurodegeneration in two mouse models of PD. Here, we studied how DRD3-signalling affects the dynamic of activation of microglia and astrocyte in the context of systemic inflammation. METHODS: Neuroinflammation was induced by intraperitoneal administration of LPS. The effect of genetic DRD3-deficiency or pharmacologic DRD3-antagonism in the functional phenotype of astrocytes and microglia was determined by immunohistochemistry and flow cytometry at different time-points. RESULTS: Our results show that DRD3 was expressed in astrocytes, but not in microglial cells. DRD3 deficiency resulted in unresponsiveness of astrocytes and in attenuated microglial activation upon systemic inflammation. Furthermore, similar alterations in the functional phenotypes of glial cells were observed by DRD3 antagonism and genetic deficiency of DRD3 upon LPS challenge. Mechanistic analyses show that DRD3 deficiency resulted in exacerbated expression of the anti-inflammatory protein Fizz1 in glial cells both in vitro and in vivo. CONCLUSIONS: These results suggest that DRD3 signalling regulates the dynamic of the acquisition of pro-inflammatory and anti-inflammatory features by astrocytes and microglia, finally favouring microglial activation and promoting neuroinflammation.


Subject(s)
Astrocytes/metabolism , Inflammation Mediators/metabolism , Microglia/metabolism , Receptors, Dopamine D3/metabolism , Signal Transduction/physiology , Animals , Astrocytes/drug effects , Cells, Cultured , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , Receptors, Dopamine D3/antagonists & inhibitors , Receptors, Dopamine D3/genetics , Signal Transduction/drug effects
8.
J Autoimmun ; 75: 105-117, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27528513

ABSTRACT

As it has been established that demethylation of lysine 27 of histone H3 by the lysine-specific demethylase JMJD3 increases immune responses and thus elicits inflammation, we hypothesize that inhibition of JMJD3 may attenuate autoimmune disorders. We found that in vivo administration of GSK-J4, a selective inhibitor of JMJD3 and UTX, ameliorates the severity of experimental autoimmune encephalomyelitis (EAE). In vitro experiments revealed that the anti-inflammatory effect of GSK-J4 was exerted through an effect on dendritic cells (DCs), promoting a tolerogenic profile characterized by reduced expression of costimulatory molecules CD80/CD86, an increased expression of tolerogenic molecules CD103 and TGF-ß1, and reduced secretion of proinflammatory cytokines IL-6, IFN-γ, and TNF. Adoptive transfer of GSK-J4-treated DCs into EAE mice reduced the clinical manifestation of the disease and decreased the extent of inflammatory CD4+ T cells infiltrating the central nervous system. Notably, Treg generation, stability, and suppressive activity were all exacerbated by GSK-J4-treated DCs without affecting Th1 and Th17 cell production. Our data show that GSK-J4-mediated modulation of inflammation is achieved by a direct effect on DCs and that systemic treatment with GSK-J4 or adoptive transfer of GSK-J4-treated DCs ex vivo may be promising approaches for the treatment of inflammatory and autoimmune disorders.


Subject(s)
Benzazepines/pharmacology , Dendritic Cells/drug effects , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Pyrimidines/pharmacology , Adoptive Transfer , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression/drug effects , Immune Tolerance/genetics , Immune Tolerance/immunology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Jumonji Domain-Containing Histone Demethylases/immunology , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism
9.
Immunology ; 146(4): 582-94, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26331349

ABSTRACT

The CD73 ectonucleotidase catalyses the hydrolysis of AMP to adenosine, an immunosuppressive molecule. Recent evidence has demonstrated that this ectonucleotidase is up-regulated in T helper type 17 cells when generated in the presence of transforming growth factor-ß (TGF-ß), and hence CD73 expression is related to the acquisition of immunosuppressive potential by these cells. TGF-ß is also able to induce CD73 expression in CD8(+) T cells but the function of this ectonucleotidase in CD8(+) T cells is still unknown. Here, we show that Tc17 cells present high levels of the CD73 ectonucleotidase and produce adenosine; however, they do not suppress the proliferation of CD4(+) T cells. Interestingly, we report that adenosine signalling through A2A receptor favours interleukin-17 production and the expression of stem cell-associated transcription factors such as tcf-7 and lef-1 but restrains the acquisition of Tc1-related effector molecules such as interferon-γ and Granzyme B by Tc17 cells. Within the tumour microenvironment, CD73 is highly expressed in CD62L(+) CD127(+) CD8(+) T cells (memory T cells) and is down-regulated in GZMB(+) KLRG1(+) CD8(+) T cells (terminally differentiated T cells), demonstrating that CD73 is expressed in memory/naive cells and is down-regulated during differentiation. These data reveal a novel function of CD73 ectonucleotidase in arresting CD8(+) T-cell differentiation and support the idea that CD73-driven adenosine production by Tc17 cells may promote stem cell-like properties in Tc17 cells.


Subject(s)
5'-Nucleotidase/metabolism , Adenosine/biosynthesis , CD8-Positive T-Lymphocytes/metabolism , Stem Cells/metabolism , T-Lymphocyte Subsets/metabolism , Adenosine Monophosphate/metabolism , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cytokines/biosynthesis , Down-Regulation , Immunologic Memory , Immunophenotyping , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Transgenic , Phenotype , Stem Cells/cytology , Stem Cells/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology
10.
Eur J Immunol ; 43(5): 1264-73, 2013 May.
Article in English | MEDLINE | ID: mdl-23436642

ABSTRACT

Plasmacytoid pre-dendritic cells (pDCs) are specialized in responding to nucleic acids, and link innate with adaptive immunity. Although the response of pDCs to viruses is well established, whether pDCs can respond to extracellular bacteria remains controversial. Here, we demonstrate that extracellular bacteria such as Neisseria meningitidis, Haemophilus influenzae, and Staphylococcus aureus activate pDCs to produce IFN-α, TNF-α, IL-6, and to upregulate CD86 expression. We observed that pDCs were present within tonsillar crypts and oro-nasopharyngeal epithelium, where they may contact extracellular bacteria, in situ. Tonsil epithelium-conditioned supernatants inhibited IFN-α, TNF-α, and IL-6 triggered by the direct contact of N. meningitidis or S. aureus with pDCs. However, pDC priming of naive T cells was not affected, suggesting that tonsil epithelium micro-environment limits local inflammation while preserving adaptive immunity in response to extracellular bacteria. Our results reveal an important and novel function of pDCs in the initiation of the mucosal innate and adaptive immunity to extracellular bacteria.


Subject(s)
Adenoids/cytology , Dendritic Cells/cytology , Epithelial Cells/cytology , Immunity, Mucosal , Respiratory Mucosa/cytology , Adaptive Immunity , Adenoids/immunology , Adenoids/microbiology , B7-2 Antigen/genetics , B7-2 Antigen/immunology , Cell Communication , Coculture Techniques , Culture Media, Conditioned/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/microbiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Haemophilus influenzae/growth & development , Humans , Immunity, Innate , Interferon-alpha/biosynthesis , Interferon-alpha/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Lymphocyte Activation , Neisseria meningitidis/growth & development , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Staphylococcus aureus/growth & development , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology , Up-Regulation
11.
Immunology ; 139(1): 61-71, 2013 May.
Article in English | MEDLINE | ID: mdl-23278668

ABSTRACT

T helper type 17 (Th17) lymphocytes are found in high frequency in tumour-burdened animals and cancer patients. These lymphocytes, characterized by the production of interleukin-17 and other pro-inflammatory cytokines, have a well-defined role in the development of inflammatory and autoimmune pathologies; however, their function in tumour immunity is less clear. We explored possible opposing anti-tumour and tumour-promoting functions of Th17 cells by evaluating tumour growth and the ability to promote tumour infiltration of myeloid-derived suppressor cells (MDSC), regulatory T cells and CD4(+)  interferon-γ(+) cells in a retinoic acid-like orphan receptor γt (RORγt) -deficient mouse model. A reduced percentage of Th17 cells in the tumour microenvironment in RORγt-deficient mice led to enhanced tumour growth, that could be reverted by adoptive transfer of Th17 cells. Differences in tumour growth were not associated with changes in the accumulation or suppressive function of MDSC and regulatory T cells but were related to a decrease in the proportion of CD4(+) T cells in the tumour. Our results suggest that Th17 cells do not affect the recruitment of immunosuppressive populations but favour the recruitment of effector Th1 cells to the tumour, thereby promoting anti-tumour responses.


Subject(s)
Immune Tolerance , Neoplasms/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Cell Line, Tumor , Mice , Mice, Mutant Strains , Neoplasms/genetics , Neoplasms/pathology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Th1 Cells/pathology , Th17 Cells/pathology
12.
Clin Dev Immunol ; 2013: 679804, 2013.
Article in English | MEDLINE | ID: mdl-23737814

ABSTRACT

Regulatory T cells are a specific subset of lymphocytes that suppress immune responses and play a crucial role in the maintenance of self-tolerance. They can be generated in the thymus as well as in the periphery through differentiation of naïve CD4(+) T cells. The forkhead box P3 transcription factor (Foxp3) is a crucial molecule regulating the generation and function of Tregs. Here we show that the foxp3 gene promoter becomes hyperacetylated in in vitro differentiated Tregs compared to naïve CD4(+) T cells. We also show that the histone deacetylase inhibitor TSA stimulated the in vitro differentiation of naïve CD4(+) T cells into Tregs and that this induction was accompanied by a global increase in histone H3 acetylation. Importantly, we also demonstrated that Tregs generated in the presence of TSA have phenotypical and functional differences from the Tregs generated in the absence of TSA. Thus, TSA-generated Tregs showed increased suppressive activities, which could potentially be explained by a mechanism involving the ectonucleotidases CD39 and CD73. Our data show that TSA could potentially be used to enhance the differentiation and suppressive function of CD4(+)Foxp3(+) Treg cells.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Immune Tolerance , T-Lymphocytes, Regulatory/drug effects , 5'-Nucleotidase/genetics , 5'-Nucleotidase/immunology , Acetylation , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Apyrase/genetics , Apyrase/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cytokines/biosynthesis , Cytokines/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins , Histones/genetics , Histones/immunology , Histones/metabolism , Mice , Mice, Transgenic , Promoter Regions, Genetic , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
13.
Clin Dev Immunol ; 2013: 210506, 2013.
Article in English | MEDLINE | ID: mdl-23762087

ABSTRACT

One of the greatest advances in medicine during the past century is the introduction of organ transplantation. This therapeutic strategy designed to treat organ failure and organ dysfunction allows to prolong the survival of many patients that are faced with no other treatment option. Today, organ transplantation between genetically dissimilar individuals (allogeneic grafting) is a procedure widely used as a therapeutic alternative in cases of organ failure, hematological disease treatment, and some malignancies. Despite the potential of organ transplantation, the administration of immunosuppressive drugs required for allograft acceptance induces severe immunosuppression in transplanted patients, which leads to serious side effects such as infection with opportunistic pathogens and the occurrence of neoplasias, in addition to the known intrinsic toxicity of these drugs. To solve this setback in allotransplantation, researchers have focused on manipulating the immune response in order to create a state of tolerance rather than unspecific immunosuppression. Here, we describe the different treatments and some of the novel immunotherapeutic strategies undertaken to induce transplantation tolerance.


Subject(s)
Graft Rejection/prevention & control , Immunologic Factors/therapeutic use , Organ Transplantation , Transplantation Tolerance/drug effects , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival/drug effects , Graft Survival/immunology , Humans , Immunosuppression Therapy , Immunosuppressive Agents/adverse effects , Macrophages/immunology , Macrophages/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transplantation, Homologous
14.
Rev Med Chil ; 141(5): 574-81, 2013 May.
Article in Spanish | MEDLINE | ID: mdl-24089271

ABSTRACT

BACKGROUND: An increased inflammatory innate response may play a role in pathogenesis of respiratory syncytial virus (RSV) infection. AIM: To quantify pro-inflammatory cytokines (IL-6-IL-8, ÍL-2-P and TNF-a) in nasopharyngeal aspirate (NPA) and plasma, and plasma cortisol in previously healthy infants with RSV bronchiolitis. PATIENTS AND METHODS: We studied 49 infants aged less than one year of age with RSV bronchiolitis and 25 healthy controls. Severity was defined using a previously described modified score. We quantified interleukins in NPA and plasma by flow cytometry and plasma cortisol by radioimmunoanalysis. RESULTS: Among patients with RSV bronchiolitis, 25 were classified as severe and 24 as moderate or mild. Significantly higher levels of IL-6 and IL-8 in NPA and plasma and IL-lfi in NPA were found in children classified as severe, when compared to those with moderate or mild disease and controls. There was a positive correlation between IL-6 and cortisol in plasma (r = 0,55; p < 0,0001) and both were correlated with the severity of the disease. CONCLUSIONS: RSV bronchiolitis severity was associated with higher levéis of inflammatory interleukins and plasma cortisol.


Subject(s)
Bronchiolitis/blood , Hydrocortisone/blood , Interleukins/blood , Respiratory Syncytial Virus Infections/blood , Tumor Necrosis Factor-alpha/blood , Bronchiolitis/immunology , Bronchiolitis/virology , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infant , Male , Nasopharynx/virology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Severity of Illness Index
15.
Am J Reprod Immunol ; 89(4): e13687, 2023 04.
Article in English | MEDLINE | ID: mdl-36757025

ABSTRACT

BACKGROUND: Maternal obesity (MO) is associated with a higher risk of immune-mediated diseases in the offspring and higher leptin levels in cord blood (CB). This study evaluates the number and function of lymphocyte subtypes in CB related to MO and its relationship with leptin concentration and leptin receptor expression. METHODS: Pregnant women with (n = 32) or without obesity (n = 41) were enrolled at delivery. Cord blood mononuclear cells were separated with Ficoll-Hypaque. B and CD4+, regulatory and effector T cells were quantified by Flow Cytometry. Cord blood leptin concentration was measured by ELISA, and the leptin receptor (sLepR) on Treg cells was determined by Flow Cytometry. RESULTS: MO was associated with higher numbers of CD4+, Treg and effector T cells in the CB of their offspring, without differences in the suppressive function of Tregs. Female offspring had a higher number of these cells and a higher cord leptin concentration. Tregs expressed higher levels of sLepR than effector T cells, without differences between groups. CONCLUSIONS: MO is associated with changes in the newborn's immune profile, more evident in female newborns with higher leptin concentrations. More studies are needed to identify the mechanisms by which the high levels of cord leptin in the newborn of women with obesity could affect the offspring's immune system.


Subject(s)
Obesity, Maternal , T-Lymphocytes, Regulatory , Female , Humans , Infant, Newborn , Pregnancy , Leptin , Receptors, Leptin/metabolism , Obesity/metabolism , Fetal Blood
16.
Front Cell Dev Biol ; 11: 1089728, 2023.
Article in English | MEDLINE | ID: mdl-37025177

ABSTRACT

The initiation of adaptive immunity relies on the performance of dendritic cells (DCs), which are specialized leukocytes with professional antigen presenting capabilities. As such, the molecular mechanisms safeguarding DC homeostasis are matter of intense research. Sensors of the unfolded protein response (UPR) of the endoplasmic reticulum, a three-pronged signaling pathway that maintains the fidelity of the cellular proteome, have emerged as regulators of DC biology. The archetypical example is the IRE1/XBP1s axis, which supports DC development and survival of the conventional type 1 DC (cDC1) subtype. However, the role of additional UPR sensors in DC biology, such as the ATF6α branch, has not been clearly elucidated. Even though Xbp1 is transcriptionally induced by ATF6α under ER stress, it is unclear if cDCs also co-opt the ATF6α branch in tissues. Here, we examine the role of ATF6α in cDC homeostasis in vivo and upon innate stimulation in vitro. In steady state, animals lacking ATF6α in CD11c+ cells (Itgax Cre x Atf6 fl/fl mice) display normal cDC frequencies in spleen, intestine, liver, and lung. Also, ATF6α deficient cDCs express normal levels of Xbp1 mRNA and additional UPR components. However, a reduction of lung monocytes is observed in Itgax Cre x Atf6 fl/fl conditional deficient animals suggesting that ATF6α may play a role in the biology of monocyte subsets. Notably, in settings of DC activation, ATF6α contributes to the production of IL-12 and IL-6 to inflammatory stimuli. Thus, although ATF6α may be dispensable for tissue cDC homeostasis in steady state, the transcription factor plays a role in the acquisition of selective immunogenic features by activated DCs.

17.
Front Immunol ; 14: 1209588, 2023.
Article in English | MEDLINE | ID: mdl-37346037

ABSTRACT

In cancer, activation of the IRE1/XBP1s axis of the unfolded protein response (UPR) promotes immunosuppression and tumor growth, by acting in cancer cells and tumor infiltrating immune cells. However, the role of IRE1/XBP1s in dendritic cells (DCs) in tumors, particularly in conventional type 1 DCs (cDC1s) which are cellular targets in immunotherapy, has not been fully elucidated. Here, we studied the role of IRE1/XBP1s in subcutaneous B16/B78 melanoma and MC38 tumors by generating loss-of-function models of IRE1 and/or XBP1s in DCs or in cDC1s. Data show that concomitant deletion of the RNase domain of IRE1 and XBP1s in DCs and cDC1s does not influence the kinetics of B16/B78 and MC38 tumor growth or the effector profile of tumor infiltrating T cells. A modest effect is observed in mice bearing single deletion of XBP1s in DCs, which showed slight acceleration of melanoma tumor growth and dysfunctional T cell responses, however, this effect was not recapitulated in animals lacking XBP1 only in cDC1s. Thus, evidence presented here argues against a general pro-tumorigenic role of the IRE1/XBP1s pathway in tumor associated DC subsets.


Subject(s)
Melanoma, Experimental , Ribonucleases , Mice , Animals , Ribonucleases/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Adaptive Immunity , Ribonuclease, Pancreatic/metabolism , Melanoma, Experimental/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Dendritic Cells
18.
Gastroenterology ; 141(1): 176-85, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21596042

ABSTRACT

BACKGROUND & AIMS: Gut-associated dendritic cells (DC) metabolize vitamin A into all-trans retinoic acid (RA), which is required to induce lymphocytes to localize to the gastrointestinal tract and promotes the differentiation of Foxp3+ regulatory T cells and IgA antibody-secreting cells. We investigated whether RA functions in a positive-feedback loop in DC to induce its own synthesis. METHODS: We measured levels of retinoids in intestinal tissues from mice and assessed the role of RA in the functional specialization of gut-associated DC in cell cultures and mice. We used pharmacologic antagonists to determine the signaling pathways involved in regulation of DC and used MyD88-/- mice to determine the contribution of Toll-like receptor signaling in RA-mediated effects on DC. RESULTS: The concentration of retinoids decreased in a proximal-to-distal gradient along the intestine, which correlated with the activity of gut-specific DC. Importantly, RA regulated the ability of gut-associated DC to produce RA, induce T cells to localize to the gastrointestinal tract, and generate regulatory T cells and IgA-secreting cells. RA was sufficient to induce its own production by extraintestinal DC in vitro and in vivo. RA-mediated regulation of DC required signaling through the mitogen-activated protein kinase signaling pathway and unexpectedly required MyD88, which is conventionally associated with Toll-like receptor, interleukin-1, and interleukin-18 signaling. CONCLUSIONS: RA is necessary and sufficient to induce DC to regulate T-cell localization to the gastrointestinal tract and IgA secretion. Our findings also indicate crosstalk between the RA receptor and MyD88-dependent Toll-like receptor signaling pathways.


Subject(s)
Dendritic Cells/metabolism , Intestinal Mucosa/metabolism , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , Tretinoin/metabolism , Analysis of Variance , Animals , Cells, Cultured , Chemotaxis, Leukocyte , Coculture Techniques , Dendritic Cells/drug effects , Dendritic Cells/immunology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological , Humans , Immunoglobulin A, Secretory/metabolism , Intestines/drug effects , Intestines/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Time Factors , Toll-Like Receptors/metabolism , Tretinoin/administration & dosage
19.
J Immunol ; 184(2): 775-86, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20018631

ABSTRACT

Autoantibodies are of central importance in the pathogenesis of Ab-mediated autoimmune disorders. The murine lupus susceptibility locus Nba2 on chromosome 1 and the syntenic human locus are associated with a loss of immune tolerance that leads to antinuclear Ab production. To identify gene intervals within Nba2 that control the development of autoantibody-producing B cells and to determine the cellular components through which Nba2 genes accomplish this, we generated congenic mice expressing various Nba2 intervals where genes for the FcgammaR, SLAM, and IFN-inducible families are encoded. Analysis of congenic strains demonstrated that the FcgammaR and SLAM intervals independently controlled the severity of autoantibody production and renal disease, yet are both required for lupus susceptibility. Deregulated homeostasis of terminally differentiated B cells was found to be controlled by the FcgammaR interval where FcgammaRIIb-mediated apoptosis of germinal center B cells and plasma cells was impaired. Increased numbers of activated plasmacytoid dendritic cells that were distinctly CD19+ and promoted plasma cell differentiation via the proinflammatory cytokines IL-10 and IFNalpha were linked to the SLAM interval. These findings suggest that SLAM and FcgammaR intervals act cooperatively to influence the clinical course of disease through supporting the differentiation and survival of autoantibody-producing cells.


Subject(s)
Antigens, CD/genetics , Lupus Erythematosus, Systemic/genetics , Receptors, Cell Surface/genetics , Receptors, IgG/genetics , Animals , Apoptosis , Autoantibodies/biosynthesis , B-Lymphocytes/pathology , Cell Differentiation , Cytokines/physiology , Disease Progression , Genetic Predisposition to Disease/genetics , Kidney Diseases , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Congenic , Plasma Cells/pathology , Signaling Lymphocytic Activation Molecule Family Member 1
20.
Front Immunol ; 12: 766698, 2021.
Article in English | MEDLINE | ID: mdl-34790201

ABSTRACT

The thymus is home to a significant number of resident B cells which possess several unique characteristics regarding their origin, phenotype and function. Evidence shows that they originate both from precursors that mature intrathymically and as the entry of recirculating mature B cells. Under steady-state conditions they exhibit hallmark signatures of activated B cells, undergo immunoglobulin class-switch, and express the Aire transcription factor. These features are imprinted within the thymus and enable B cells to act as specialized antigen-presenting cells in the thymic medulla that contribute negative selection of self-reactive T cells. Though, most studies have focused on B cells located in the medulla, a second contingent of B cells is also present in non-epithelial perivascular spaces of the thymus. This latter group of B cells, which includes memory B cells and plasma cells, is not readily detected in the thymus of infants or young mice but gradually accumulates during normal aging. Remarkably, in many autoimmune diseases the thymus suffers severe structural atrophy and infiltration of B cells in the perivascular spaces, which organize into follicles similar to those typically found in secondary lymphoid organs. This review provides an overview of the pathways involved in thymic B cell origin and presents an integrated view of both thymic medullary and perivascular B cells and their respective physiological and pathological roles in central tolerance and autoimmune diseases.


Subject(s)
Autoimmunity/immunology , B-Lymphocytes/immunology , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Thymus Gland/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Movement/immunology , Humans , Plasma Cells/immunology , Plasma Cells/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism , AIRE Protein
SELECTION OF CITATIONS
SEARCH DETAIL