Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 5208, 2022 09 05.
Article in English | MEDLINE | ID: mdl-36064857

ABSTRACT

Adipose tissue macrophage (ATM) inflammation is involved with meta-inflammation and pathology of metabolic complications. Here we report that in adipocytes, elevated lactate production, previously regarded as the waste product of glycolysis, serves as a danger signal to promote ATM polarization to an inflammatory state in the context of obesity. Adipocyte-selective deletion of lactate dehydrogenase A (Ldha), the enzyme converting pyruvate to lactate, protects mice from obesity-associated glucose intolerance and insulin resistance, accompanied by a lower percentage of inflammatory ATM and reduced production of pro-inflammatory cytokines such as interleukin 1ß (IL-1ß). Mechanistically, lactate, at its physiological concentration, fosters the activation of inflammatory macrophages by directly binding to the catalytic domain of prolyl hydroxylase domain-containing 2 (PHD2) in a competitive manner with α-ketoglutarate and stabilizes hypoxia inducible factor (HIF-1α). Lactate-induced IL-1ß was abolished in PHD2-deficient macrophages. Human adipose lactate level is positively linked with local inflammatory features and insulin resistance index independent of the body mass index (BMI). Our study shows a critical function of adipocyte-derived lactate in promoting the pro-inflammatory microenvironment in adipose and identifies PHD2 as a direct sensor of lactate, which functions to connect chronic inflammation and energy metabolism.


Subject(s)
Adipocytes , Hypoxia-Inducible Factor-Proline Dioxygenases , Inflammation , Lactate Dehydrogenase 5 , Lactic Acid , Macrophages , Adipocytes/immunology , Adipose Tissue/immunology , Animals , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Insulin Resistance/genetics , Insulin Resistance/immunology , Insulin Resistance/physiology , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/immunology , Lactate Dehydrogenase 5/genetics , Lactate Dehydrogenase 5/immunology , Lactic Acid/immunology , Macrophages/immunology , Mice , Obesity/genetics , Obesity/immunology , Obesity/pathology , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/immunology , Prolyl Hydroxylases
2.
J Clin Invest ; 132(2)2022 01 18.
Article in English | MEDLINE | ID: mdl-35040434

ABSTRACT

As cancers progress, they produce a local environment that acts to redirect, paralyze, exhaust, or otherwise evade immune detection and destruction. The tumor microenvironment (TME) has long been characterized as a metabolic desert, depleted of essential nutrients such as glucose, oxygen, and amino acids, that starves infiltrating immune cells and renders them dysfunctional. While not incorrect, this perspective is only half the picture. The TME is not a metabolic vacuum, only consuming essential nutrients and never producing by-products. Rather, the by-products of depleted nutrients, "toxic" metabolites in the TME such as lactic acid, kynurenine, ROS, and adenosine, play an important role in shaping immune cell function and cannot be overlooked in cancer immunotherapy. Moreover, while the metabolic landscape is distinct, it is not unique, as these toxic metabolites are encountered in non-tumor tissues, where they evolutionarily shape immune cells and their response. In this Review, we discuss how depletion of essential nutrients and production of toxic metabolites shape the immune response within the TME and how toxic metabolites can be targeted to improve current cancer immunotherapies.


Subject(s)
Neoplasms/immunology , Tumor Microenvironment/immunology , Adenosine/immunology , Adenosine/metabolism , Animals , Humans , Immunotherapy , Kynurenine/immunology , Kynurenine/metabolism , Lactic Acid/immunology , Lactic Acid/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism
3.
Front Immunol ; 12: 729209, 2021.
Article in English | MEDLINE | ID: mdl-34899690

ABSTRACT

Elevated blood lactate levels are frequently found in critically ill patients and thought to result from tissue hypoperfusion and cellular oxygen shortage. Considering the close relationship between immune cell function and intracellular metabolism, lactate is more than a glycolytic waste molecule but able to regulate the immune response. Our aim was to elucidate the temporal and mechanistic effect of extracellular lactate on monocytes. To this end, primary human monocytes and the human monocytic cell line MonoMac6 were stimulated with various toll-like-receptor agonists after priming with Na-L-lactate under constant pH conditions. As readout, cytokine production was measured, real-time assessment of intracellular energy pathways was performed, and intracellular metabolite concentrations were determined. Irrespective of the immunogenic stimulus, short-term Na-lactate-priming strongly reduced cytokine production capacity. Lactate and hexoses accumulated intracellularly and, together with a decreased glycolytic flux, indicate a lactate-triggered impairment of glycolysis. To counteract intracellular hyperglycemia, glucose is shunted into the branching polyol pathway, leading to sorbitol accumulation. In contrast, long-term priming with Na-L-lactate induced cellular adaption and abolished the suppressive effect. This lactate tolerance is characterized by a decreased cellular respiration due to a reduced complex-I activity. Our results indicate that exogenous lactate shapes monocyte function by altering the intracellular energy metabolism and acts as a metabolic checkpoint of monocyte activation.


Subject(s)
Lactic Acid/immunology , Lactic Acid/metabolism , Monocytes/immunology , Monocytes/metabolism , Cell Line , Extracellular Fluid/immunology , Extracellular Fluid/metabolism , Humans
4.
Front Immunol ; 12: 691134, 2021.
Article in English | MEDLINE | ID: mdl-34394085

ABSTRACT

For decades, lactate has been considered an innocuous bystander metabolite of cellular metabolism. However, emerging studies show that lactate acts as a complex immunomodulatory molecule that controls innate and adaptive immune cells' effector functions. Thus, recent advances point to lactate as an essential and novel signaling molecule that shapes innate and adaptive immune responses in the intestine and systemic sites. Here, we review these recent advances in the context of the pleiotropic effects of lactate in regulating diverse functions of immune cells in the tissue microenvironment and under pathological conditions.


Subject(s)
Dendritic Cells/immunology , Lactic Acid/immunology , Macrophages/immunology , Animals , Autoimmunity , Cell Cycle Proteins/immunology , Humans , Immunomodulation , Infections/immunology , Inflammatory Bowel Diseases/immunology , Monocarboxylic Acid Transporters/immunology , Neoplasms/immunology , Receptors, G-Protein-Coupled/immunology
5.
Front Immunol ; 12: 688910, 2021.
Article in English | MEDLINE | ID: mdl-34177945

ABSTRACT

Lactate is an end product of glycolysis. As a critical energy source for mitochondrial respiration, lactate also acts as a precursor of gluconeogenesis and a signaling molecule. We briefly summarize emerging concepts regarding lactate metabolism, such as the lactate shuttle, lactate homeostasis, and lactate-microenvironment interaction. Accumulating evidence indicates that lactate-mediated reprogramming of immune cells and enhancement of cellular plasticity contribute to establishing disease-specific immunity status. However, the mechanisms by which changes in lactate states influence the establishment of diverse functional adaptive states are largely uncharacterized. Posttranslational histone modifications create a code that functions as a key sensor of metabolism and are responsible for transducing metabolic changes into stable gene expression patterns. In this review, we describe the recent advances in a novel lactate-induced histone modification, histone lysine lactylation. These observations support the idea that epigenetic reprogramming-linked lactate input is related to disease state outputs, such as cancer progression and drug resistance.


Subject(s)
Lactic Acid/metabolism , Acetyl Coenzyme A/metabolism , Animals , Epigenesis, Genetic , Histones/metabolism , Humans , Lactic Acid/immunology , Tumor Microenvironment
6.
Med Hypotheses ; 148: 110520, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33561624

ABSTRACT

Following the decline in Physical Activity (PA) due to COVID-19 restrictions in the form of government mandated lockdowns and closures of public spaces, the modulatory effect of physical exercise on immunity is being heavily revisited. In an attempt to comprehend the wide discrepancy in patient response to COVID-19 and the factors that potentially modulate it, we summarize the findings relating PA to inflammation and immunity. A distinction is drawn between moderate intensity and high intensity physical exercise based on the high lactate production observed in the latter. We hypothesize that, the lactate production associated with high intensity anaerobic exercise is implicated in the modulation of several components of the innate and adaptive immunity. In this review, we also summarize these immunomodulatory effects of lactate. These include increasing serum IL-6 levels, the main mediator of cytokine storms, as well as affecting NK cells, Macrophages, Dendritic cells and cytotoxic T-lymphocytes. The implications of high lactate levels in athletic performance are highlighted where athletes should undergo endurance training to increase VO2 max and minimize lactate production. Tumor models of hypoxia were also reported where lactate levels are elevated leading to increased invasiveness and angiogenesis. Accordingly, the novel lactate blocking strategy employed in cancer treatment is evaluated for its potential benefit in COVID-19 in addition to the readily available beta-blockers as an antagonist to lactate. Finally, we suggest the diagnostic/prognostic purpose of the elevated lactate levels that can be determined through sweat lactate testing. It is the detrimental effect of lactate on immunity and its presence in sweat that qualify it to be used as a potential non-invasive marker of poor COVID-19 outcome.


Subject(s)
COVID-19 Drug Treatment , Lactic Acid/antagonists & inhibitors , Anaerobiosis/immunology , COVID-19/immunology , COVID-19/physiopathology , Exercise/physiology , Humans , Inflammation/immunology , Interleukin-6/blood , Lactic Acid/immunology , Lactic Acid/metabolism , Models, Immunological , Pandemics , SARS-CoV-2
7.
J Microbiol Immunol Infect ; 54(3): 404-410, 2021 Jun.
Article in English | MEDLINE | ID: mdl-31727535

ABSTRACT

BACKGROUND: This study aims to explore the mechanism of immunosuppression in septic Acute Renal Injury (AKI) and the role of programmed death-1 (PD-1/PD-L1) pathway in septic AKI. METHODS: This study established a septic AKI model by Cecal ligation and puncture (CLP) in C57/B6 mice, ELISA was used to test the level of lactate and creatinine in serum, blood was collected for flow cytometry and kidney samples for Western blot analyses. This study further analyzed the expression of PD-L1 in kidney and the expression of PD-1 in CD4+, CD8+ T cell, and the number of CD3+ T cells to identify apoptosis in T cells in the blood. RESULTS: The CLP sepsis model induced AKI in C57/B6 mice; The expression of PD-1 and PD-L1 were increased in septic AKI mice; PD-1/PD-L1 induced apoptosis in T cells: the number of lymphocytes decreased by 64%, while the number of CD3+ T cells decreased by 27% compared with the sham group; Results also indicated that lactate up-regulates expression of PD-L1 in the kidney. CONCLUSIONS: Lactate activated PD-1/PD-L1 pathway can induce immunosuppression by inducing apoptosis in lymphocytes in septic AKI. Moreover, blocking the receptor of lactate or PD-1/PD-L1 might be a new therapy for septic AKI.


Subject(s)
Acute Kidney Injury/genetics , Acute Kidney Injury/immunology , B7-H1 Antigen/genetics , Immune Tolerance/immunology , Kidney/pathology , Lactic Acid/immunology , Animals , Apoptosis/immunology , Disease Models, Animal , Epithelial Cells , Lactic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Sepsis/complications , Sepsis/microbiology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
8.
Cancer Lett ; 500: 75-86, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33347908

ABSTRACT

The tumour microenvironment is of critical importance in cancer development and progression and includes the surrounding stromal and immune cells, extracellular matrix, and the milieu of metabolites and signalling molecules in the intercellular space. To support sustained mitotic activity cancer cells must reconfigure their metabolic phenotype. Lactate is the major by-product of such metabolic alterations and consequently, accumulates in the tumour. Lactate actively contributes to immune evasion, a hallmark of cancer, by directly inhibiting immune cell cytotoxicity and proliferation. Furthermore, lactate can recruit and induce immunosuppressive cell types, such as regulatory T cells, tumour-associated macrophages, and myeloid-derived suppressor cells which further suppress anti-tumour immune responses. Given its roles in oncogenesis, measuring intratumoural and systemic lactate levels has shown promise as a both predictive and prognostic biomarker in several cancer types. The efficacies of many anti-cancer therapies are limited by an immunosuppressive TME in which lactate is a major contributor, therefore, targeting lactate metabolism is a priority. Developing inhibitors of key proteins in lactate metabolism such as GLUT1, hexokinase, LDH, MCT and HIF have shown promise in preclinical studies, however there is a corresponding lack of success in human trials so far. This may be explained by a weakness of preclinical models that fail to reproduce the complexities of metabolic interactions in natura. The future of these therapies may be as an adjunct to more conventional treatments.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Glycolysis/genetics , Neoplasms/drug therapy , Glucose Transporter Type 1/antagonists & inhibitors , Glycolysis/drug effects , Hexokinase/antagonists & inhibitors , Humans , Immune Tolerance/genetics , Immune Tolerance/immunology , Immunity/drug effects , Immunity/immunology , Immunosuppression Therapy , L-Lactate Dehydrogenase/antagonists & inhibitors , Lactic Acid/immunology , Lactic Acid/metabolism , Mitosis/drug effects , Mitosis/immunology , Monocarboxylic Acid Transporters/antagonists & inhibitors , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/drug effects
9.
J Biol Chem ; 295(29): 10032-10044, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32503842

ABSTRACT

Fungal pathogen Candida albicans has a complex cell wall consisting of an outer layer of mannans and an inner layer of ß-glucans and chitin. The fungal cell wall is the primary target for antifungals and is recognized by host immune cells. Environmental conditions such as carbon sources, pH, temperature, and oxygen tension can modulate the fungal cell wall architecture. Cellular signaling pathways, including the mitogen-activated protein kinase (MAPK) pathways, are responsible for sensing environmental cues and mediating cell wall alterations. Although iron has recently been shown to affect ß-1,3-glucan exposure on the cell wall, we report here that iron changes the composition of all major C. albicans cell wall components. Specifically, high iron decreased the levels of mannans (including phosphomannans) and chitin; and increased ß-1,3-glucan levels. These changes increased the resistance of C. albicans to cell wall-perturbing antifungals. Moreover, high iron cells exhibited adequate mitochondrial functioning; leading to a reduction in accumulation of lactate that signals through the transcription factor Crz1 to induce ß-1,3-glucan masking in C. albicans We show here that iron-induced changes in ß-1,3-glucan exposure are lactate-dependent; and high iron causes ß-1,3-glucan exposure by preventing lactate-induced, Crz1-mediated inhibition of activation of the fungal MAPK Cek1. Furthermore, despite exhibiting enhanced antifungal resistance, high iron C. albicans cells had reduced survival upon phagocytosis by macrophages. Our results underscore the role of iron as an environmental signal in multiple signaling pathways that alter cell wall architecture in C. albicans, thereby affecting its survival upon exposure to antifungals and host immune response.


Subject(s)
Antifungal Agents/pharmacology , Candida albicans , Candidiasis , Cell Wall , Iron , Lactic Acid , Macrophages , Phagocytosis , Animals , Candida albicans/immunology , Candida albicans/metabolism , Candidiasis/drug therapy , Candidiasis/immunology , Candidiasis/metabolism , Cell Wall/immunology , Cell Wall/metabolism , Female , Iron/immunology , Iron/metabolism , Lactic Acid/immunology , Lactic Acid/metabolism , Macrophages/immunology , Macrophages/microbiology , Mice
10.
Mol Cell ; 78(5): 814-823, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32333837

ABSTRACT

Metabolites have functions in the immune system independent of their conventional roles as sources or intermediates in biosynthesis and bioenergetics. We are still in the pioneering phase of gathering information about the functions of specific metabolites in immunoregulation. In this review, we cover succinate, itaconate, α-ketoglutarate, and lactate as examples. Each of these metabolites has a different story of how their immunoregulatory functions were discovered and how their roles in the complex process of inflammation were revealed. Parallels and interactions are emerging between metabolites and cytokines, well-known immunoregulators. We depict molecular mechanisms by which metabolites prime cellular and often physiological changes focusing on intra- and extra-cellular activities and signaling pathways. Possible therapeutic opportunities for immune and inflammatory diseases are emerging.


Subject(s)
Carboxylic Acids/immunology , Carboxylic Acids/metabolism , Immunity/immunology , Animals , Citric Acid Cycle , Cytokines/metabolism , Energy Metabolism , Humans , Immunity/physiology , Inflammation/metabolism , Ketoglutaric Acids/immunology , Ketoglutaric Acids/metabolism , Lactic Acid/immunology , Lactic Acid/metabolism , Signal Transduction , Succinates/immunology , Succinates/metabolism , Succinic Acid/immunology , Succinic Acid/metabolism
12.
Front Immunol ; 10: 1878, 2019.
Article in English | MEDLINE | ID: mdl-31440253

ABSTRACT

Plasmacytoid dendritic cells are the most efficient producers of type I interferons, viz. IFNα, in the body and thus have the ability to influence anti-tumor immune responses. But repression of effective intra-tumoral pDC activation is a key immuno-evasion strategy exhibited in tumors-tumor-recruited pDCs are rendered "tolerogenic," characterized by deficiency in IFNα induction and ability to expand regulatory T cells in situ. But the tumor-derived factors that drive this functional reprogramming of intra-tumoral pDCs are not established. In this study we aimed at exploring if intra-tumoral abundance of the oncometabolite lactate influences intra-tumoral pDC function. We found that lactate attenuates IFNα induction by pDCs mediated by intracellular Ca2+ mobilization triggered by cell surface GPR81 receptor as well as directly by cytosolic import of lactate in pDCs through the cell surface monocarboxylate transporters, affecting cellular metabolism needed for effective pDC activation. We also found that lactate enhances tryptophan metabolism and kynurenine production by pDCs which contribute to induction of FoxP3+ CD4+ regulatory T cells, the major immunosuppressive immune cell subset in tumor microenvironment. We validated these mechanisms of lactate-driven pDC reprogramming by looking into tumor recruited pDCs isolated from patients with breast cancers as well as in a preclinical model of breast cancer in mice. Thus, we discovered a hitherto unknown link between intra-tumoral abundance of an oncometabolite resulting from metabolic adaptation in cancer cells and the pro-tumor tolerogenic function of tumor-recruited pDCs, revealing new therapeutic targets for potentiating anti-cancer immune responses.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Dendritic Cells/immunology , Lactic Acid/immunology , Tumor Escape/physiology , Animals , Cellular Reprogramming/immunology , Dendritic Cells/metabolism , Female , Humans , Lactic Acid/metabolism , Mice , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology
13.
Cell Immunol ; 341: 103918, 2019 07.
Article in English | MEDLINE | ID: mdl-31030957

ABSTRACT

Mast cells have functional plasticity affected by their tissue microenvironment, which greatly impacts their inflammatory responses. Because lactic acid (LA) is abundant in inflamed tissues and tumors, we investigated how it affects mast cell function. Using IgE-mediated activation as a model system, we found that LA suppressed inflammatory cytokine production and degranulation in mouse peritoneal mast cells, data that were confirmed with human skin mast cells. In mouse peritoneal mast cells, LA-mediated cytokine suppression was dependent on pH- and monocarboxylic transporter-1 expression. Additionally, LA reduced IgE-induced Syk, Btk, and ERK phosphorylation, key signals eliciting inflammation. In vivo, LA injection reduced IgE-mediated hypothermia in mice undergoing passive systemic anaphylaxis. Our data suggest that LA may serve as a feedback inhibitor that limits mast cell-mediated inflammation.


Subject(s)
Anaphylaxis/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Feedback, Physiological , Immunoglobulin E/genetics , Lactic Acid/pharmacology , Mast Cells/drug effects , Agammaglobulinaemia Tyrosine Kinase/genetics , Agammaglobulinaemia Tyrosine Kinase/immunology , Anaphylaxis/chemically induced , Anaphylaxis/immunology , Anaphylaxis/pathology , Animals , Dinitrophenols/administration & dosage , Dinitrophenols/antagonists & inhibitors , Female , Gene Expression Regulation , Ketoprofen/pharmacology , Lactic Acid/immunology , Lactic Acid/metabolism , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/immunology , Peritoneal Cavity/pathology , Phosphorylation/drug effects , Primary Cell Culture , Serum Albumin/administration & dosage , Serum Albumin/antagonists & inhibitors , Signal Transduction , Skin/drug effects , Skin/immunology , Skin/pathology , Syk Kinase/genetics , Syk Kinase/immunology , Symporters/genetics , Symporters/immunology
14.
Int J Oncol ; 54(2): 585-599, 2019 02.
Article in English | MEDLINE | ID: mdl-30535454

ABSTRACT

Nasopharyngeal carcinoma (NPC) is the most common malignant tumor type in Southern China and South­East Asia. Cluster of differentiation (CD)38 is highly expressed in the human immune system and participates in the activation of T, natural killer and plasma cells mediated by CD2 and CD3 through synergistic action. CD38 is a type II transmembrane glycoprotein, which was observed to mediate diverse activities, including signal transduction, cell adhesion and cyclic ADP­ribose synthesis. However, the significance of CD38 in NPC biological behavior and cellular energy metabolism has not been examined. In order to elucidate the effect of CD38 on the biological behavior of NPC cells, stable CD38­overexpressed NPC cell lines were established. It was demonstrated that CD38 promoted NPC cell proliferation with Cell Counting Kit­8 and colony formation assays. It was also indicated that CD38 inhibited cell senescence, and promoted cell metastasis. Furthermore, it was determined that CD38 promoted the conversion of cells to the S phase and decreased the content of reactive oxygen species and Ca2+. Additionally, cell metabolism assays demonstrated that CD38 increased the concentration of ATP, lactic acid, cyclic adenosine monophosphate and human ADP/acrp30 concentration in NPC cells. To investigate the possible mechanism, bioinformatics analysis and mass spectrometry technology was used to determine the most notably changing molecule and signaling pathways, and it was determined and verified that CD38 regulated the metabolic­associated signaling pathways associated with tumor protein 53, hypoxia inducible factor­1α and sirtuin 1. The present results indicated that CD38 may serve a carcinogenic role in NPC by regulating metabolic­associated signaling pathways.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Nasopharyngeal Carcinoma/genetics , Tumor Suppressor Protein p53/genetics , ADP-ribosyl Cyclase 1/immunology , ADP-ribosyl Cyclase 1/metabolism , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Energy Metabolism/genetics , Energy Metabolism/immunology , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lactic Acid/immunology , Lactic Acid/metabolism , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Reactive Oxygen Species/metabolism , Signal Transduction , Sirtuin 1/genetics , Sirtuin 1/immunology , Sirtuin 1/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism
15.
Vet Microbiol ; 212: 67-74, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29173590

ABSTRACT

Campylobacter jejuni (C. jejuni) is a leading bacterial cause of food-borne illness in humans. Contaminated chicken meat is an important source of infection for humans. Chickens are not clinically affected by colonization, and immune responses following natural infection have limited effects on bacterial load in the gut. Induction of intestinal immune responses may possibly lead to a breakdown of the commensal relationship of chickens with Campylobacter. We have recently shown that soluble and poly D, L-lactic-co-glycolic acid (PLGA)-encapsulated CpG oligodeoxynucleotide (ODN) as well as C. jejuni lysate, are effective in reducing the intestinal burden of C. jejuni in chickens; however, the mechanisms behind this protection have yet to be determined. The present study was undertaken to investigate the mechanisms of host responses conferred by these treatments. Chickens were treated orally with soluble CpG ODN, or PLGA-encapsulated CpG ODN, or C. jejuni lysate, and expression of cytokines and antimicrobial peptides was evaluated in cecal tonsils and ileum using quantitative RT-PCR. Oral administration of soluble CpG ODN upregulated the expression of interferon (IFN)-γ, interleukin (IL)-1ß, CXCLi2, transforming growth factor (TGF)-ß4/1, IL-10 and IL-13, while treatment with PLGA-encapsulated CpG ODN upregulated the expression of IL-1ß, CXCLi2, TGF-ß4/1, IL-13, avian ß-defensin (AvBD) 1, AvBD2 and cathelicidin 3 (CATHL-3). C. jejuni lysate upregulated the expression of IFN-γ, IL-1ß, TGF-ß4/1, IL-13, AvBD1, and CATHL-3. In conclusion, induction of cytokine and antimicrobial peptides expression in intestinal microenvironments may provide a means of reducing C. jejuni colonization in broiler chickens, a key step in reducing the incidence of campylobacteriosis in humans.


Subject(s)
Campylobacter Infections/veterinary , Campylobacter jejuni/immunology , Chickens/genetics , Immunity, Innate , Lactic Acid/immunology , Oligodeoxyribonucleotides/immunology , Administration, Oral , Animals , Campylobacter Infections/microbiology , Chickens/immunology , Chickens/microbiology , Cytokines/genetics , Cytokines/immunology , Gene Expression Profiling , Ileum/immunology , Palatine Tonsil/immunology , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer
16.
Int J Nanomedicine ; 12: 6169-6184, 2017.
Article in English | MEDLINE | ID: mdl-28883727

ABSTRACT

Visceral leishmaniasis (VL) persists as a major public health problem, and since the existing chemotherapy is far from satisfactory, development of an effective vaccine emerges as the most appropriate strategy for confronting VL. The development of an effective vaccine relies on the selection of the appropriate antigen and also the right adjuvant and/or delivery vehicle. In the present study, the protective efficacy of poly(D,L-lactide-co-glycolide) (PLGA) nanoparticles (NPs), which were surface-modified with a TNFα-mimicking eight-amino-acid peptide (p8) and further functionalized by encapsulating soluble Leishmania infantum antigens (sLiAg) and monophosphoryl lipid A (MPLA), a TLR4 ligand, was evaluated against challenge with L. infantum parasites in BALB/c mice. Vaccination with these multifunctionalized PLGA nanoformulations conferred significant protection against parasite infection in vaccinated mice. In particular, vaccination with PLGA-sLiAg-MPLA or p8-PLGA-sLiAg NPs resulted in almost complete elimination of the parasite in the spleen for up to 4 months post-challenge. Parasite burden reduction was accompanied by antigen-specific humoral and cellular immune responses. Specifically, injection with PLGA-sLiAg-MPLA raised exclusively anti-sLiAg IgG1 antibodies post-vaccination, while in p8-PLGA-sLiAg-vaccinated mice, no antibody production was detected. However, 4 months post-challenge, in mice vaccinated with all the multifunctionalized NPs, antibody class switching towards IgG2a subtype was observed. The study of cellular immune responses revealed the increased proliferation capacity of spleen cells against sLiAg, consisting of IFNγ-producing CD4+ and CD8+ T cells. Importantly, the activation of CD8+ T cells was exclusively attributed to vaccination with PLGA NPs surface-modified with the p8 peptide. Moreover, characterization of cytokine production in vaccinated-infected mice revealed that protection was accompanied by significant increase of IFNγ and lower levels of IL-4 and IL-10 in protected mice when compared to control infected group. Conclusively, the above nanoformulations hold promise for future vaccination strategies against VL.


Subject(s)
Leishmaniasis Vaccines/chemistry , Leishmaniasis Vaccines/pharmacology , Leishmaniasis, Visceral/prevention & control , Nanoparticles/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Protozoan/chemistry , CD8-Positive T-Lymphocytes/immunology , Female , Immunity, Cellular , Interleukin-10/metabolism , Interleukin-4/metabolism , Lactic Acid/chemistry , Lactic Acid/immunology , Leishmania infantum/chemistry , Leishmaniasis, Visceral/immunology , Lipid A/analogs & derivatives , Lipid A/chemistry , Lipid A/immunology , Mice, Inbred BALB C , Nanoparticles/chemistry , Peptides/chemistry , Peptides/immunology , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Tumor Necrosis Factor-alpha/chemistry
17.
J Cancer Res Ther ; 13(3): 406-411, 2017.
Article in English | MEDLINE | ID: mdl-28862200

ABSTRACT

Prostate cancer, one of the most common male malignancies with an increasing incidence in the recent years, requires the development of new methods of treatment. One of the most debated subjects is the tumor-associated macrophages (TAM). Although, the pathophysiological mechanisms are still a subject of intense research, TAM acts as procarcinogenic factors. It was also demonstrated that hypoxia-inducible factor 1 (HIF1) induces the expression of TAM genes involved in prostate carcinogenesis. Furthermore, it should be noted that the stromal extracellular lactate, the result of tumoral glycolysis process is one of the HIF1 activators. In addition, lactate inhibits the differentiation of monocytes and dendritic cells and also induces the inactivation of the cytotoxic T-lymphocytes. Through an analysis of recent studies, we conclude that lactate is a vital component of several ways of modulating the immune response at the stromal prostatic adenocarcinoma including TAM activation and cytotoxic T lymphocytes immunosuppression. Our review focuses on the impact of lactate on prostatic adenocarcinoma progression in terms of its immunology, and how this influences the therapy of this condition and the clinical outcome.


Subject(s)
Adenocarcinoma/metabolism , Carcinogenesis/immunology , Lactic Acid/metabolism , Prostatic Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Cell Differentiation/genetics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lactic Acid/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology
18.
Cell Metab ; 25(6): 1282-1293.e7, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28416194

ABSTRACT

Immune cells function in diverse metabolic environments. Tissues with low glucose and high lactate concentrations, such as the intestinal tract or ischemic tissues, frequently require immune responses to be more pro-tolerant, avoiding unwanted reactions against self-antigens or commensal bacteria. T-regulatory cells (Tregs) maintain peripheral tolerance, but how Tregs function in low-glucose, lactate-rich environments is unknown. We report that the Treg transcription factor Foxp3 reprograms T cell metabolism by suppressing Myc and glycolysis, enhancing oxidative phosphorylation, and increasing nicotinamide adenine dinucleotide oxidation. These adaptations allow Tregs a metabolic advantage in low-glucose, lactate-rich environments; they resist lactate-mediated suppression of T cell function and proliferation. This metabolic phenotype may explain how Tregs promote peripheral immune tolerance during tissue injury but also how cancer cells evade immune destruction in the tumor microenvironment. Understanding Treg metabolism may therefore lead to novel approaches for selective immune modulation in cancer and autoimmune diseases.


Subject(s)
Cellular Microenvironment/immunology , Cellular Reprogramming/immunology , Forkhead Transcription Factors/immunology , Glucose/immunology , Lactic Acid/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line , Cellular Microenvironment/genetics , Cellular Reprogramming/genetics , Forkhead Transcription Factors/genetics , Glucose/genetics , Glycolysis/genetics , Glycolysis/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Oxidative Phosphorylation , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology
19.
Parasite ; 24: 5, 2017.
Article in English | MEDLINE | ID: mdl-28145222

ABSTRACT

Triphasic sustained release of tachyzoite chimeric protein, rSAG1/2, from poly(lactide-co-glycolide) (PLG)-encapsulated rSAG1/2 (PLG-rSAG1/2) microparticles (MPs) is a promising characteristic for developing a single-dose vaccine against Toxoplasma gondii in domestic animals. In the present study, we aimed to evaluate whether single immunization with PLG-rSAG1/2 MPs in BALB/c mice would achieve effective immunity and protection against T. gondii. Peritoneal immunization of mice with a single dose of PLG-rSAG1/2 MPs enhanced serum IgG titers and lymphocyte proliferation in a triphasic model over a long 12-week period. In addition, 12 weeks after immunization, significant production of IFN-γ was also monitored in mice vaccinated with one dose of PLG-rSAG1/2 MPs. More importantly, the immunity induced by one dose of PLG-rSAG1/2 MPs protected 70% of mice (14/20) against a lethal subcutaneous challenge of 1 × 104 live tachyzoites of T. gondii (RH strain). In conclusion, a single dose of PLG-rSAG1/2 MPs capable of sustaining triphasic release of rSAG1/2 protein induces long-lasting triphasic immunity against T. gondii in mice. Our data indicate the feasibility of PLG-rSAG1/2 MPs to be developed as a single-dose vaccine against T. gondii for potential use in domestic animals.


Subject(s)
Antigens, Protozoan/immunology , Protozoan Proteins/immunology , Protozoan Vaccines , Toxoplasma/immunology , Toxoplasmosis, Animal/prevention & control , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/administration & dosage , Cell-Derived Microparticles/chemistry , Cell-Derived Microparticles/immunology , Epitopes , Female , Immunoglobulin G/blood , Infusions, Parenteral , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lactic Acid/immunology , Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Protozoan Proteins/administration & dosage , Protozoan Vaccines/administration & dosage , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Spleen/cytology , Spleen/immunology , Toxoplasmosis, Animal/immunology
20.
Nanomedicine (Lond) ; 12(5): 491-510, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28181470

ABSTRACT

AIM: Dendritic cells rapidly capture nanoparticles and induce a potent cellular immune response. It is yet unknown whether the immunological response induced by slow release of encapsulated versus soluble antigen and adjuvant is superior. MATERIALS & METHODS: The kinetics of poly(lactic-co-glycolic acid) PLGA nanoparticles antigen release was studied by the DQ-bovine serum albumin (BSA) self-quenching antigen model. The immunological response induced was evaluated by means of dendritic cell activation/maturation markers, cytokine production and their ability to drive antigen-specific T-cell proliferation. RESULTS & CONCLUSION: PLGA-encapsulated antigen and adjuvant showed an enhanced T-cell response when compared with soluble vaccine components by increasing antigenicity and adjuvanticity. Although the kinetic profile followed the same pattern, encapsulation increased strength and duration of the response.


Subject(s)
Dendritic Cells/immunology , Immunity, Cellular/drug effects , Immunogenicity, Vaccine/immunology , Nanoparticles/administration & dosage , T-Lymphocytes/immunology , Animals , Antigens/chemistry , Antigens/immunology , Cattle , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Humans , Immunogenicity, Vaccine/drug effects , Lactic Acid/administration & dosage , Lactic Acid/chemistry , Lactic Acid/immunology , Nanoparticles/chemistry , Polyglycolic Acid/administration & dosage , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/immunology , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...