Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Nat Commun ; 13(1): 5208, 2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-36064857

RESUMO

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.


Assuntos
Adipócitos , Prolina Dioxigenases do Fator Induzível por Hipóxia , Inflamação , Lactato Desidrogenase 5 , Ácido Láctico , Macrófagos , Adipócitos/imunologia , Tecido Adiposo/imunologia , Animais , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Resistência à Insulina/genética , Resistência à Insulina/imunologia , Resistência à Insulina/fisiologia , L-Lactato Desidrogenase/genética , L-Lactato Desidrogenase/imunologia , Lactato Desidrogenase 5/genética , Lactato Desidrogenase 5/imunologia , Ácido Láctico/imunologia , Macrófagos/imunologia , Camundongos , Obesidade/genética , Obesidade/imunologia , Obesidade/patologia , Pró-Colágeno-Prolina Dioxigenase/genética , Pró-Colágeno-Prolina Dioxigenase/imunologia , Prolil Hidroxilases
2.
J Clin Invest ; 132(2)2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35040434

RESUMO

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.


Assuntos
Neoplasias/imunologia , Microambiente Tumoral/imunologia , Adenosina/imunologia , Adenosina/metabolismo , Animais , Humanos , Imunoterapia , Cinurenina/imunologia , Cinurenina/metabolismo , Ácido Láctico/imunologia , Ácido Láctico/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo
3.
Front Immunol ; 12: 691134, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34394085

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Ácido Láctico/imunologia , Macrófagos/imunologia , Animais , Autoimunidade , Proteínas de Ciclo Celular/imunologia , Humanos , Imunomodulação , Infecções/imunologia , Doenças Inflamatórias Intestinais/imunologia , Transportadores de Ácidos Monocarboxílicos/imunologia , Neoplasias/imunologia , Receptores Acoplados a Proteínas G/imunologia
4.
Front Immunol ; 12: 688910, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34177945

RESUMO

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.


Assuntos
Ácido Láctico/metabolismo , Acetilcoenzima A/metabolismo , Animais , Epigênese Genética , Histonas/metabolismo , Humanos , Ácido Láctico/imunologia , Microambiente Tumoral
5.
Med Hypotheses ; 148: 110520, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33561624

RESUMO

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.


Assuntos
Tratamento Farmacológico da COVID-19 , Ácido Láctico/antagonistas & inibidores , Anaerobiose/imunologia , COVID-19/imunologia , COVID-19/fisiopatologia , Exercício Físico/fisiologia , Humanos , Inflamação/imunologia , Interleucina-6/sangue , Ácido Láctico/imunologia , Ácido Láctico/metabolismo , Modelos Imunológicos , Pandemias , SARS-CoV-2
6.
Cancer Lett ; 500: 75-86, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33347908

RESUMO

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.


Assuntos
Antineoplásicos/uso terapêutico , Carcinogênese/efeitos dos fármacos , Glicólise/genética , Neoplasias/tratamento farmacológico , Transportador de Glucose Tipo 1/antagonistas & inibidores , Glicólise/efeitos dos fármacos , Hexoquinase/antagonistas & inibidores , Humanos , Tolerância Imunológica/genética , Tolerância Imunológica/imunologia , Imunidade/efeitos dos fármacos , Imunidade/imunologia , Terapia de Imunossupressão , L-Lactato Desidrogenase/antagonistas & inibidores , Ácido Láctico/imunologia , Ácido Láctico/metabolismo , Mitose/efeitos dos fármacos , Mitose/imunologia , Transportadores de Ácidos Monocarboxílicos/antagonistas & inibidores , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Microambiente Tumoral/efeitos dos fármacos
7.
J Microbiol Immunol Infect ; 54(3): 404-410, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31727535

RESUMO

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.


Assuntos
Injúria Renal Aguda/genética , Injúria Renal Aguda/imunologia , Antígeno B7-H1/genética , Tolerância Imunológica/imunologia , Rim/patologia , Ácido Láctico/imunologia , Animais , Apoptose/imunologia , Modelos Animais de Doenças , Células Epiteliais , Ácido Láctico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sepse/complicações , Sepse/microbiologia , Linfócitos T/imunologia , Linfócitos T/patologia
8.
J Biol Chem ; 295(29): 10032-10044, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32503842

RESUMO

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.


Assuntos
Antifúngicos/farmacologia , Candida albicans , Candidíase , Parede Celular , Ferro , Ácido Láctico , Macrófagos , Fagocitose , Animais , Candida albicans/imunologia , Candida albicans/metabolismo , Candidíase/tratamento farmacológico , Candidíase/imunologia , Candidíase/metabolismo , Parede Celular/imunologia , Parede Celular/metabolismo , Feminino , Ferro/imunologia , Ferro/metabolismo , Ácido Láctico/imunologia , Ácido Láctico/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos
10.
Front Immunol ; 10: 1878, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31440253

RESUMO

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.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Células Dendríticas/imunologia , Ácido Láctico/imunologia , Evasão Tumoral/fisiologia , Animais , Reprogramação Celular/imunologia , Células Dendríticas/metabolismo , Feminino , Humanos , Ácido Láctico/metabolismo , Camundongos , Linfócitos T Reguladores/imunologia , Microambiente Tumoral/imunologia
11.
Cell Immunol ; 341: 103918, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31030957

RESUMO

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.


Assuntos
Anafilaxia/prevenção & controle , Anti-Inflamatórios não Esteroides/farmacologia , Retroalimentação Fisiológica , Imunoglobulina E/genética , Ácido Láctico/farmacologia , Mastócitos/efeitos dos fármacos , Tirosina Quinase da Agamaglobulinemia/genética , Tirosina Quinase da Agamaglobulinemia/imunologia , Anafilaxia/induzido quimicamente , Anafilaxia/imunologia , Anafilaxia/patologia , Animais , Dinitrofenóis/administração & dosagem , Dinitrofenóis/antagonistas & inibidores , Feminino , Regulação da Expressão Gênica , Cetoprofeno/farmacologia , Ácido Láctico/imunologia , Ácido Láctico/metabolismo , Mastócitos/imunologia , Mastócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/imunologia , Cavidade Peritoneal/patologia , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Albumina Sérica/administração & dosagem , Albumina Sérica/antagonistas & inibidores , Transdução de Sinais , Pele/efeitos dos fármacos , Pele/imunologia , Pele/patologia , Quinase Syk/genética , Quinase Syk/imunologia , Simportadores/genética , Simportadores/imunologia
12.
Int J Oncol ; 54(2): 585-599, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30535454

RESUMO

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.


Assuntos
ADP-Ribosil Ciclase 1/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Carcinoma Nasofaríngeo/genética , Proteína Supressora de Tumor p53/genética , ADP-Ribosil Ciclase 1/imunologia , ADP-Ribosil Ciclase 1/metabolismo , Trifosfato de Adenosina/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Metabolismo Energético/genética , Metabolismo Energético/imunologia , Regulação Neoplásica da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/imunologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ácido Láctico/imunologia , Ácido Láctico/metabolismo , Carcinoma Nasofaríngeo/imunologia , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/patologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Sirtuína 1/genética , Sirtuína 1/imunologia , Sirtuína 1/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Proteína Supressora de Tumor p53/imunologia , Proteína Supressora de Tumor p53/metabolismo
13.
J Cancer Res Ther ; 13(3): 406-411, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28862200

RESUMO

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.


Assuntos
Adenocarcinoma/metabolismo , Carcinogênese/imunologia , Ácido Láctico/metabolismo , Neoplasias da Próstata/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Diferenciação Celular/genética , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Ácido Láctico/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/patologia
14.
Int J Nanomedicine ; 12: 6169-6184, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28883727

RESUMO

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.


Assuntos
Vacinas contra Leishmaniose/química , Vacinas contra Leishmaniose/farmacologia , Leishmaniose Visceral/prevenção & controle , Nanopartículas/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Animais , Antígenos de Protozoários/química , Linfócitos T CD8-Positivos/imunologia , Feminino , Imunidade Celular , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Ácido Láctico/química , Ácido Láctico/imunologia , Leishmania infantum/química , Leishmaniose Visceral/imunologia , Lipídeo A/análogos & derivados , Lipídeo A/química , Lipídeo A/imunologia , Camundongos Endogâmicos BALB C , Nanopartículas/química , Peptídeos/química , Peptídeos/imunologia , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Fator de Necrose Tumoral alfa/química
15.
Cell Metab ; 25(6): 1282-1293.e7, 2017 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-28416194

RESUMO

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.


Assuntos
Microambiente Celular/imunologia , Reprogramação Celular/imunologia , Fatores de Transcrição Forkhead/imunologia , Glucose/imunologia , Ácido Láctico/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linhagem Celular , Microambiente Celular/genética , Reprogramação Celular/genética , Fatores de Transcrição Forkhead/genética , Glucose/genética , Glicólise/genética , Glicólise/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fosforilação Oxidativa , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/imunologia
16.
Am J Obstet Gynecol ; 216(1): 60.e1-60.e17, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27615440

RESUMO

BACKGROUND: Uterine inflammatory processes trigger prolabor pathways and orchestrate on-time labor onset. Although essential for successful labor, inflammation needs to be regulated to avoid uncontrolled amplification and resolve postpartum. During labor, myometrial smooth muscle cells generate ATP mainly via anaerobic glycolysis, resulting in accumulation of lactate. Aside from its metabolic function, lactate has been shown to activate a G protein-coupled receptor, GPR81, reported to regulate inflammation. We therefore hypothesize that lactate produced during labor may act via GPR81 in the uterus to exert in a feedback manner antiinflammatory effects, to resolve or mitigate inflammation. OBJECTIVE: We sought to investigate the role of lactate produced during labor and its receptor, GPR81, in regulating inflammation in the uterus. STUDY DESIGN: We investigated the expression of GPR81 in the uterus and the pharmacological role of lactate acting via GPR81 during labor, using shRNA-GPR81 and GPR81-/- mice. RESULTS: (1) Uterine lactate levels increased substantially from 2 to 9 mmol/L during labor. (2) Immunohistological analysis revealed expression of GPR81 in the uterus with high expression in myometrium. (3) GPR81 expression increased during gestation, and peaked near labor. (4) In primary myometrial smooth muscle cell and ex vivo uteri from wild-type mice, lactate decreased interleukin-1ß-induced transcription of key proinflammatory Il1b, Il6, Ccl2, and Pghs2; suppressive effects of lactate were not observed in cells and tissues from GPR81-/- mice. (5) Conversely, proinflammatory gene expression was augmented in the uterus at term in GPR81-/- mice and wild-type mice treated intrauterine with lentiviral-encoded shRNA-GPR81; GPR81 silencing also induced proinflammatory gene transcription in the uterus when labor was induced by endotoxin (lipopolysaccharide). (6) Importantly, administration to pregnant mice of a metabolically stable specific GPR81 agonist, 3,5-dihydroxybenzoic acid, decreased endotoxin-induced uterine inflammation, preterm birth, and associated neonatal mortality. CONCLUSION: Collectively, our data uncover a novel link between the anaerobic glycolysis and the control of uterine inflammation wherein the high levels of lactate produced during labor act on uterine GPR81 to down-regulate key proinflammatory genes. This discovery may represent a novel feedback mechanism to regulate inflammation during labor, and conveys a potential rationale for the use of GPR81 agonists to attenuate inflammation and resulting preterm birth.


Assuntos
Inflamação , Trabalho de Parto/imunologia , Ácido Láctico/imunologia , Miométrio/imunologia , Receptores Acoplados a Proteínas G/genética , Animais , Quimiocina CCL2/efeitos dos fármacos , Quimiocina CCL2/genética , Ciclo-Oxigenase 2/efeitos dos fármacos , Ciclo-Oxigenase 2/genética , Feminino , Hidroxibenzoatos/farmacologia , Imuno-Histoquímica , Técnicas In Vitro , Interleucina-1beta/efeitos dos fármacos , Interleucina-1beta/genética , Interleucina-1beta/farmacologia , Interleucina-6/genética , Trabalho de Parto/metabolismo , Ácido Láctico/metabolismo , Ácido Láctico/farmacologia , Camundongos Knockout , Miométrio/metabolismo , Gravidez , RNA Interferente Pequeno , Receptores Acoplados a Proteínas G/imunologia , Resorcinóis/farmacologia , Útero/imunologia , Útero/metabolismo
17.
PLoS One ; 11(4): e0151922, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27093541

RESUMO

Pigs are believed to be one of the important sources of emerging human and swine influenza viruses (SwIV). Influenza virus conserved peptides have the potential to elicit cross-protective immune response, but without the help of potent adjuvant and delivery system they are poorly immunogenic. Biodegradable polylactic-co-glycolic acid (PLGA) nanoparticle (PLGA-NP) based vaccine delivery system enhances cross-presentation of antigens by the professional antigen presenting cells. In this study, Norovirus P particle containing SwIV M2e (extracellular domain of the matrix protein 2) chimera and highly conserved two each of H1N1 peptides of pandemic 2009 and classical human influenza viruses were entrapped in PLGA-NPs. Influenza antibody-free pigs were vaccinated with PLGA-NPs peptides cocktail vaccine twice with or without an adjuvant, Mycobacterium vaccae whole cell lysate, intranasally as mist. Vaccinated pigs were challenged with a virulent heterologous zoonotic SwIV H1N1, and one week later euthanized and the lung samples were analyzed for the specific immune response and viral load. Clinically, pigs vaccinated with PLGA-NP peptides vaccine had no fever and flu symptoms, and the replicating challenged SwIV was undetectable in the bronchoalveolar lavage fluid. Immunologically, PLGA-NP peptides vaccination (without adjuvant) significantly increased the frequency of antigen-specific IFNγ secreting CD4 and CD8 T cells response in the lung lymphocytes, despite not boosting the antibody response both at pre- and post-challenge. In summary, our data indicated that nanoparticle-mediated delivery of conserved H1N1 influenza peptides induced the virus specific T cell response in the lungs and reduced the challenged heterologous virus load in the airways of pigs.


Assuntos
Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/química , Ácido Láctico/química , Nanopartículas/química , Peptídeos/química , Ácido Poliglicólico/química , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Animais , Anticorpos Antivirais/imunologia , Linhagem Celular , Cães , Vírus da Influenza A Subtipo H1N1/química , Vacinas contra Influenza/imunologia , Interferon gama/imunologia , Ácido Láctico/imunologia , Pulmão/imunologia , Pulmão/virologia , Células Madin Darby de Rim Canino , Nanopartículas/administração & dosagem , Infecções por Orthomyxoviridae/imunologia , Peptídeos/imunologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Suínos , Doenças dos Suínos/imunologia , Vacinas de Produtos Inativados/química , Vacinas de Produtos Inativados/imunologia
18.
Trends Biochem Sci ; 41(5): 460-471, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26935843

RESUMO

The integration of biochemistry into immune cell biology has contributed immensely to our understanding of immune cell function and the associated pathologies. So far, most studies have focused on the regulation of metabolic pathways during an immune response and their contribution to its success. More recently, novel signalling functions of metabolic intermediates are being discovered that might play important roles in the regulation of immunity. Here we describe the three long-known small metabolites lactate, acetyl-CoA, and succinate in the context of immunometabolic signalling. Functions of these ubiquitous molecules are largely dependent on their intra- and extracellular concentrations as well as their subcompartmental localisation. Importantly, the signalling functions of these metabolic intermediates extend beyond self-regulatory roles and include cell-to-cell communication and sensing of microenvironmental conditions to elicit stress responses and cellular adaptation.


Assuntos
Ciclo do Ácido Cítrico/imunologia , Glicólise/imunologia , Imunidade Inata , Macrófagos/metabolismo , Transdução de Sinais/imunologia , Linfócitos T/metabolismo , Acetilcoenzima A/imunologia , Acetilcoenzima A/metabolismo , Comunicação Celular/imunologia , Citocinas/biossíntese , Citocinas/imunologia , Células Endoteliais/citologia , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Ácidos Graxos/imunologia , Ácidos Graxos/metabolismo , Humanos , Ácido Láctico/imunologia , Ácido Láctico/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Neurônios/citologia , Neurônios/imunologia , Neurônios/metabolismo , Ácido Succínico/imunologia , Ácido Succínico/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia
19.
Sci Rep ; 5: 13092, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26271607

RESUMO

The biomechanics stress and chronic inflammation in obesity are causally linked to osteoarthritis. However, the metabolic factors mediating obesity-related osteoarthritis are still obscure. Here we scanned and identified at least two elevated metabolites (stearic acid and lactate) from the plasma of diet-induced obese mice. We found that stearic acid potentiated LDH-a-dependent production of lactate, which further stabilized HIF1α protein and increased VEGF and proinflammatory cytokine expression in primary mouse chondrocytes. Treatment with LDH-a and HIF1α inhibitors notably attenuated stearic acid-or high fat diet-stimulated proinflammatory cytokine production in vitro and in vivo. Furthermore, positive correlation of plasma lactate, cartilage HIF1α and cytokine levels with the body mass index was observed in subjects with osteoarthritis. In conclusion, saturated free fatty acid induced proinflammatory cytokine production partly through activation of a novel lactate-HIF1α pathway in chondrocytes. Our findings hold promise of developing novel clinical strategies for the management of obesity-related diseases such as osteoarthritis.


Assuntos
Condrócitos/imunologia , Citocinas/imunologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/imunologia , Mediadores da Inflamação/imunologia , Ácido Láctico/imunologia , Ácidos Esteáricos/imunologia , Animais , Células Cultivadas , Condrócitos/citologia , Dieta Hiperlipídica/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia , Fator A de Crescimento do Endotélio Vascular/imunologia
20.
Immunobiology ; 220(10): 1161-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26101138

RESUMO

The use of short chain fatty acids to modulate gastrointestinal inflammatory conditions such as ulcerative colitis has produced encouraging results either in animal models or also in clinical trials. Identifying the key cellular and molecular targets of this activity will contribute to establish the appropriate combinations/targeting strategies to maximize the efficacy of anti-inflammatory interventions. In the present work, we evaluated in vitro the interaction of lactate, acetate, propionate and butyrate on cells relevant for innate immune response of the gastrointestinal tract. All molecules tested regulate the production of proinflammatory cytokines by TLR-4 and TLR-5 activated intestinal epithelial cells in a dose response manner. Furthermore SCFAs and lactate modulate cytokine secretion of TLR-activated bone marrow derived macrophages and also TLR-dependent CD40 upregulation in bone marrow derived dendritic in a dose-dependent manner. Butyrate and propionate have been effective at concentrations of 1 to 5mM whereas acetate and lactate produced modulatory effects at concentrations higher than 20-50mM in different assays. Our results indicate that in concentrations similar to found in large bowel lumen, all SCFAs tested and lactate can modulate activity of relevant sentinel cell types activated by TLR signals. Modulatory activity was not inhibited by pertussis toxin treatment indicating that the effects are not related to Gi signaling. The use of these molecules in combined or separately as intervention strategy in conditions where epithelial or myeloid cells are main triggers of the inflammatory situation seems appropriate.


Assuntos
Bactérias/imunologia , Regulação para Baixo/imunologia , Ácidos Graxos/imunologia , Mucosa Intestinal/imunologia , Ácido Láctico/imunologia , Células Mieloides/imunologia , Animais , Antígenos CD40/imunologia , Células CACO-2 , Células Epiteliais/imunologia , Feminino , Humanos , Mucosa Intestinal/microbiologia , Camundongos , Receptor 4 Toll-Like/imunologia , Receptor 5 Toll-Like/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA