Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 236
Filtrar
1.
Cell Transplant ; 30: 9636897211057440, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34757864

RESUMO

The inflammatory response is an obstacle to success in both allogeneic and autologous islet transplantation. In autologous islet transplantation (AIT), however, the recipient is also the donor, permitting pretreatment of donor/recipient for a controlled duration prior to transplantation. We sought to exploit this feature of (AIT) by pretreating donor/recipients with chronic pancreatitis undergoing total pancreatectomy and autologous islet transplantation (TPAIT) to test the hypothesis that peri-transplant treatment with the FDA-approved anti-inflammatory hydroxychloroquine (HCQ) improves graft function. In this randomized placebo-controlled pilot clinical study, patients (n = 6) were treated with oral HCQ for 30 days prior to and 90 days after TPAIT. In vivo islet function was assessed via Mixed Meal Tolerance Testing before HCQ treatment, 6- and 12-months after surgery. In vitro islet bioenergetics were assessed at the time of transplantation via extracellular flux analysis of islet preparation samples from the clinical trial cohort and six additional patients (n = 12). Our study shows that HCQ did not alter clinical endpoints, but HCQ-treated patients showed greater spare respiratory capacity (SRC) compared to samples from control patients (P=0.028). Glycolytic metabolism of islet preparations directly correlated with stimulated C-peptide secretion both before and after TPAIT (P=0.01, R2=0.489 and P=0.03, R2=0.674, respectively), and predicted in vivo islet function better than mitochondrial metabolism of islet preps or islet equivalents infused. Overnight culture of islet preparations altered bioenergetic function, significantly decreasing SRC and maximal respiration (P<0.001). In conclusion, while HCQ did not alter clinical outcomes, it was associated with significantly increased SRC in islet preparations. Bioenergetic analyses of islet preparations suggests that culture should be avoided and that glycolysis may be a more sensitive indicator of in vivo islet function than current metrics, including islet oxygen consumption and islet equivalents infused.


Assuntos
Metabolismo Energético/imunologia , Inibidores Enzimáticos/uso terapêutico , Hidroxicloroquina/uso terapêutico , Transplante das Ilhotas Pancreáticas/métodos , Transplante Autólogo/métodos , Ensaios Clínicos como Assunto , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Hidroxicloroquina/farmacologia , Masculino , Projetos Piloto , Resultado do Tratamento
2.
Int Immunopharmacol ; 101(Pt A): 108175, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34689102

RESUMO

Rheumatoid arthritis (RA) is a systemic autoimmune disease. Synovial hyperplasia and persistent inflammation serve as its typical pathological manifestations, which ultimately lead to joint destruction and function loss. Both clinical observations and metabolomics studies have revealed the prevalence of metabolic disorders in RA. In inflammatory immune microenvironments, energy metabolism is profoundly changed. Increasingly evidences suggest that this abnormality is involved in the occurrence and development of RA-related inflammation. Unsurprisingly, many energy metabolism sensors have been confirmed with immunoregulatory properties. As a representative, silent information regulator type 1 (Sirt1) controls many aspects of immune cells, such as cell lifespan, polarization, and secretion by functioning as a transcriptional regulator. Because of the profound clinical implication, researches on Sirt1 in the regulation of energy metabolism and immune functions under RA conditions have gradually gained momentum. This signaling balances glycolysis, lipid metabolism and insulin secretion orchestrating with other metabolism sensors, and consequently affects immune milieu through a so-called metabolism-immune feedback mechanism. This article reviews the involvement of Sirt1 in RA by discussing its impacts on energy metabolism and immune functions, and specially highlights the potential of Sirt1-targeting anti-rheumatic regimens. It also provides a theoretical basis for clarifying the mystery about the high incidence of metabolic complications in RA patients and identifying new anti-rheumatic reagents.


Assuntos
Artrite Reumatoide/imunologia , Metabolismo Energético/imunologia , Sirtuína 1/metabolismo , Animais , Antirreumáticos/farmacologia , Antirreumáticos/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/patologia , Modelos Animais de Doenças , Metabolismo Energético/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Glicólise/efeitos dos fármacos , Glicólise/imunologia , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Sirtuína 1/antagonistas & inibidores , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/imunologia , Membrana Sinovial/patologia
3.
Int J Mol Sci ; 22(19)2021 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-34638886

RESUMO

Peroxisome proliferator-activated receptor α is a potent regulator of systemic and cellular metabolism and energy homeostasis, but it also suppresses various inflammatory reactions. In this review, we focus on its role in the regulation of innate immunity; in particular, we discuss the PPARα interplay with inflammatory transcription factor signaling, pattern-recognition receptor signaling, and the endocannabinoid system. We also present examples of the PPARα-specific immunomodulatory functions during parasitic, bacterial, and viral infections, as well as approach several issues associated with innate immunity processes, such as the production of reactive nitrogen and oxygen species, phagocytosis, and the effector functions of macrophages, innate lymphoid cells, and mast cells. The described phenomena encourage the application of endogenous and pharmacological PPARα agonists to alleviate the disorders of immunological background and the development of new solutions that engage PPARα activation or suppression.


Assuntos
Metabolismo Energético/imunologia , Homeostase/imunologia , Imunidade Inata/imunologia , Inflamação/imunologia , PPAR alfa/imunologia , Transdução de Sinais/imunologia , Imunidade Adaptativa/imunologia , Animais , Humanos , Macrófagos/imunologia , PPAR alfa/metabolismo
4.
Sci Rep ; 11(1): 19258, 2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34584177

RESUMO

Crohn's disease (CD) is a debilitating inflammatory bowel condition of unknown aetiology that is growing in prevalence globally. Large-scale studies have determined associations between female obesity or low body mass index (BMI) with risk of CD at all ages or 8- < 40 years, respectively. For males, low BMI entering adult life is associated with increased incidence of CD or ulcerative colitis up to 40 years later. Body composition analysis has shown that combinations of lean tissue loss and high visceral fat predict poor CD outcomes. Here, we assessed dietary intake, physical activity and whole or regional body composition of patients with CD relapse or remission. This anthropometric approach found people with CD, irrespective of relapse or remission, differed from a large representative healthy population sample in exhibiting elevated gynoid fat and reduced android fat. CD is associated with mesenteric adipose tissue, or "creeping fat", that envelops affected intestine exclusive of other tissue; that fat is localised to the android region of the body. In this context, CD mesenteric adiposity represents a stark juxtaposition of organ-specific and regional adiposity. Although our study population was relatively small, we suggest tentatively that there is a rationale to refer to Crohn's disease as a fatty intestine condition, akin to fatty liver conditions. We suggest that our data provide early insight into a subject that potentially warrants further investigation across a larger patient cohort.


Assuntos
Adiposidade/imunologia , Doença de Crohn/metabolismo , Metabolismo Energético/imunologia , Absorciometria de Fóton , Adulto , Índice de Massa Corporal , Doença de Crohn/imunologia , Inquéritos sobre Dietas/estatística & dados numéricos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Recidiva
5.
Front Immunol ; 12: 700374, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34434191

RESUMO

Invariant natural killer T (iNKT) cells comprise a unique subset of lymphocytes that are primed for activation and possess innate NK-like functional features. Currently, iNKT cell-based immunotherapies remain in early clinical stages, and little is known about the ability of these cells to survive and retain effector functions within the solid tumor microenvironment (TME) long-term. In conventional T cells (TCONV), cellular metabolism is linked to effector functions and their ability to adapt to the nutrient-poor TME. In contrast, the bioenergetic requirements of iNKT cells - particularly those of human iNKT cells - at baseline and upon stimulation are not well understood; neither is how these requirements affect effector functions such as production of cytokines and cytolytic proteins. We find that unlike TCONV, human iNKT cells are not dependent upon glucose or glutamine for these effector functions upon stimulation with anti-CD3 and anti-CD28. Additionally, transcriptional profiling revealed that stimulated human iNKT cells are less glycolytic than TCONV and display higher expression of fatty acid oxidation (FAO) and adenosine monophosphate-activated protein kinase (AMPK) pathway genes. Furthermore, stimulated iNKT cells displayed higher mitochondrial mass and membrane potential relative to TCONV. Real-time Seahorse metabolic flux analysis revealed that stimulated human iNKT cells utilize fatty acids as substrates for oxidation more than stimulated TCONV. Together, our data suggest that human iNKT cells possess different bioenergetic requirements from TCONV and display a more oxidative metabolic program relative to effector TCONV. Importantly, iNKT cell-based immunotherapeutic strategies could co-opt such unique features of iNKT cells to improve their efficacy and longevity of anti-tumor responses.


Assuntos
Metabolismo Energético/imunologia , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Células Cultivadas , Humanos , Ativação Linfocitária/imunologia
6.
Cancer Lett ; 518: 94-101, 2021 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-34153401

RESUMO

In recent years, tumor metabolism has become a prevalent research topic for scientists and pharmaceutical companies. As research in the field has progressed, the metabolism-based therapy of tumors has ushered in new opportunities. Most tumors emerge and evolve under selective pressure from their microenvironment, which promotes the diversification of both neoplastic and non-neoplastic compartments of the tumor microenvironment (TME), and finally reaches a certain degree of intratumoral heterogeneity. As a result of the tumor intratumoral heterogeneity, tumor cells often possess a complex energy metabolism phenotype. During tumor progression, the metabolism for both tumor parenchyma and stroma is reprogrammed. The tumor stroma mainly consists of the extracellular matrix, fibroblasts, and immune cells. Interestingly, tumor-infiltrating immune cells utilize different metabolites based on their subtype and function, and these immunometabolic pathways can be modified in the TME. In particular, interleukins play a vital role in the activation and differentiation of immune cells and have exhibited multiple effects on tumor cell neoplasia, invasion, and metastasis. In this review, we summarize the common mechanisms of interleukins affecting the tumor and tumor-infiltrating immune cells metabolically and discuss how these mechanisms may lead to novel therapeutic opportunities. This review might contribute to the novel development of cancer immunotherapy.


Assuntos
Interleucinas/imunologia , Interleucinas/metabolismo , Redes e Vias Metabólicas/imunologia , Neoplasias/imunologia , Neoplasias/metabolismo , Animais , Diferenciação Celular/imunologia , Metabolismo Energético/imunologia , Matriz Extracelular/imunologia , Matriz Extracelular/metabolismo , Humanos , Imunoterapia/métodos , Neoplasias/terapia , Microambiente Tumoral/imunologia
7.
Clin Sci (Lond) ; 135(11): 1389-1408, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-34086048

RESUMO

The immune system protects the body against harm by inducing inflammation. During the immune response, cells of the immune system get activated, divided and differentiated in order to eliminate the danger signal. This process relies on the metabolic reprogramming of both catabolic and anabolic pathways not only to produce energy in the form of ATP but also to generate metabolites that exert key functions in controlling the response. Equally important to mounting an appropriate effector response is the process of immune resolution, as uncontrolled inflammation is implicated in the pathogenesis of many human diseases, including allergy, chronic inflammation and cancer. In this review, we aim to introduce the reader to the field of cholesterol immunometabolism and discuss how both metabolites arising from the pathway and cholesterol homeostasis are able to impact innate and adaptive immune cells, staging cholesterol homeostasis at the centre of an adequate immune response. We also review evidence that demonstrates the clear impact that cholesterol metabolism has in both the induction and the resolution of the inflammatory response. Finally, we propose that emerging data in this field not only increase our understanding of immunometabolism but also provide new tools for monitoring and intervening in human diseases, where controlling and/or modifying inflammation is desirable.


Assuntos
Colesterol/metabolismo , Sistema Imunitário/imunologia , Inflamação/metabolismo , Metabolismo dos Lipídeos/imunologia , Animais , Colesterol/imunologia , Metabolismo Energético/imunologia , Metabolismo Energético/fisiologia , Humanos , Inflamação/imunologia , Transdução de Sinais/imunologia , Transdução de Sinais/fisiologia
8.
Cell Death Dis ; 12(5): 434, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33934112

RESUMO

The mitochondrial paralog of the Hsp90 chaperone family TRAP1 is often induced in tumors, but the mechanisms controlling its expression, as well as its physiological functions remain poorly understood. Here, we find that TRAP1 is highly expressed in the early stages of Zebrafish development, and its ablation delays embryogenesis while increasing mitochondrial respiration of fish larvae. TRAP1 expression is enhanced by hypoxic conditions both in developing embryos and in cancer models of Zebrafish and mammals. The TRAP1 promoter contains evolutionary conserved hypoxic responsive elements, and HIF1α stabilization increases TRAP1 levels. TRAP1 inhibition by selective compounds or by genetic knock-out maintains a high level of respiration in Zebrafish embryos after exposure to hypoxia. Our data identify TRAP1 as a primary regulator of mitochondrial bioenergetics in highly proliferating cells following reduction in oxygen tension and HIF1α stabilization.


Assuntos
Metabolismo Energético/imunologia , Proteínas de Choque Térmico HSP90/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Chaperonas Moleculares/metabolismo , Animais , Hipóxia Celular , Modelos Animais de Doenças , Humanos , Peixe-Zebra
9.
J Endocrinol Invest ; 44(12): 2819-2830, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33991317

RESUMO

BACKGROUND: Obesity promotes cellular immunometabolism changes that trigger the activation of macrophages and lymphocytes, leading to systemic inflammation. Activated leukocytes undergo metabolic reprogramming, increasing glycolytic activity. OBJECTIVE: To examine whether the reduction in the inflammatory state associated with bariatric surgery is associated with decreased glycolytic activity in leukocytes. Setting Single-center, prospective observational study. METHODS: This study involved 18 patients with obesity undergoing bariatric surgery. All measurements were performed preoperatively and six months postoperatively. Peripheral blood mononuclear cells and plasma were obtained to determine the glycolytic rate and mitochondrial membrane potential as surrogates of the metabolic switching and high-sensitivity C-reactive protein, adipokines, and CD69 expression as inflammatory and activation markers. RESULTS: Glycolytic activity engaged by CD3/CD28 activation was reduced six months after bariatric surgery, associated with decreased levels of T helper (Th) 1 and Th17 signature cytokines. An overall reduction in inflammatory markers was observed, which correlated with a higher adiponectin/leptin ratio. CONCLUSIONS: Metabolic and bariatric surgery-induced weight loss leads to reprogramming in T cells' metabolic machinery, resulting in reduced stimulation of glycolysis after activation, which may explain the decrease in systemic inflammation mediated by cytokines such as interferon-γ and interleukin-17A.


Assuntos
Ativação Metabólica/imunologia , Cirurgia Bariátrica/métodos , Glicólise/imunologia , Leucócitos Mononucleares , Obesidade Mórbida , Células Th1 , Células Th17 , Adulto , Contagem de Células/métodos , Reprogramação Celular , Metabolismo Energético/imunologia , Feminino , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Masculino , Obesidade Mórbida/metabolismo , Obesidade Mórbida/cirurgia , Período Pós-Operatório , Células Th1/metabolismo , Células Th1/patologia , Células Th17/metabolismo , Células Th17/patologia
10.
Front Immunol ; 12: 645242, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33815400

RESUMO

Emerging reports show that metabolic pathways can be targeted to enhance T cell-mediated immunity to tumors. Yet, tumors consume key metabolites in the host to survive, thus robbing T cells of these nutrients to function and thrive. T cells are often deprived of basic building blocks for energy in the tumor, including glucose and amino acids needed to proliferate or produce cytotoxic molecules against tumors. Immunosuppressive molecules in the host further compromise the lytic capacity of T cells. Moreover, checkpoint receptors inhibit T cell responses by impairing their bioenergetic potential within tumors. In this review, we discuss the fundamental metabolic pathways involved in T cell activation, differentiation and response against tumors. We then address ways to target metabolic pathways to improve the next generation of immunotherapies for cancer patients.


Assuntos
Metabolismo Energético/imunologia , Ativação Linfocitária , Neoplasias , Linfócitos T , Microambiente Tumoral/imunologia , Humanos , Imunoterapia , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/terapia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia
11.
Int J Mol Sci ; 22(6)2021 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-33804765

RESUMO

The recent pandemic Sars-CoV2 infection and studies on previous influenza epidemic have drawn attention to the association between the obesity and infectious diseases susceptibility and worse outcome. Metabolic complications, nutritional aspects, physical inactivity, and a chronic unbalance in the hormonal and adipocytokine microenvironment are major determinants in the severity of viral infections in obesity. By these pleiotropic mechanisms obesity impairs immune surveillance and the higher leptin concentrations produced by adipose tissue and that characterize obesity substantially contribute to such immune response dysregulation. Indeed, leptin not only controls energy balance and body weight, but also plays a regulatory role in the interplay between energy metabolism and immune system. Since leptin receptor is expressed throughout the immune system, leptin may exert effects on cells of both innate and adaptive immune system. Chronic inflammatory states due to metabolic (i.e., obesity) as well as infectious diseases increase leptin concentrations and consequently lead to leptin resistance further fueling inflammation. Multiple factors, including inflammation and ER stress, contribute to leptin resistance. Thus, if leptin is recognized as one of the adipokines responsible for the low grade inflammation found in obesity, on the other hand, impairments of leptin signaling due to leptin resistance appear to blunt the immunologic effects of leptin and possibly contribute to impaired vaccine-induced immune responses. However, many aspects concerning leptin interactions with inflammation and immune system as well as the therapeutical approaches to overcome leptin resistance and reduced vaccine effectiveness in obesity remain a challenge for future research.


Assuntos
Leptina/imunologia , Leptina/metabolismo , Obesidade/complicações , Obesidade/virologia , Viroses/complicações , Animais , Antivirais/uso terapêutico , COVID-19/complicações , COVID-19/imunologia , COVID-19/metabolismo , Metabolismo Energético/imunologia , Humanos , Sistema Imunitário/metabolismo , Sistema Imunitário/virologia , Obesidade/imunologia , Obesidade/metabolismo , Vacinas Virais/uso terapêutico , Viroses/tratamento farmacológico , Viroses/imunologia , Viroses/metabolismo , Tratamento Farmacológico da COVID-19
12.
Nat Rev Nephrol ; 17(7): 465-480, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33828286

RESUMO

Insights into the relationship between immunometabolism and inflammation have enabled the targeting of several immunity-mediated inflammatory processes that underlie infectious diseases and cancer or drive transplant rejection, but this field remains largely unexplored in kidney diseases. The kidneys comprise heterogeneous cell populations, contain distinct microenvironments such as areas of hypoxia and hypersalinity, and are responsible for a functional triad of filtration, reabsorption and secretion. These distinctive features create myriad potential metabolic therapeutic targets in the kidney. Immune cells have crucial roles in the maintenance of kidney homeostasis and in the response to kidney injury, and their function is intricately connected to their metabolic properties. Changes in nutrient availability and biomolecules, such as cytokines, growth factors and hormones, initiate cellular signalling events that involve energy-sensing molecules and other metabolism-related proteins to coordinate immune cell differentiation, activation and function. Disruption of homeostasis promptly triggers the metabolic reorganization of kidney immune and non-immune cells, which can promote inflammation and tissue damage. The metabolic differences between kidney and immune cells offer an opportunity to specifically target immunometabolism in the kidney.


Assuntos
Metabolismo Energético/imunologia , Sistema Imunitário/fisiologia , Nefropatias/imunologia , Imunidade Adaptativa/fisiologia , Humanos , Imunidade Inata/fisiologia
13.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33771921

RESUMO

Despite their close genetic relatedness, apes and African and Asian monkeys (AAMs) differ in their susceptibility to severe bacterial and viral infections that are important causes of human disease. Such differences between humans and other primates are thought to be a result, at least in part, of interspecies differences in immune response to infection. However, because of the lack of comparative functional data across species, it remains unclear in what ways the immune systems of humans and other primates differ. Here, we report the whole-genome transcriptomic responses of ape species (human and chimpanzee) and AAMs (rhesus macaque and baboon) to bacterial and viral stimulation. We find stark differences in the responsiveness of these groups, with apes mounting a markedly stronger early transcriptional response to both viral and bacterial stimulation, altering the transcription of ∼40% more genes than AAMs. Additionally, we find that genes involved in the regulation of inflammatory and interferon responses show the most divergent early transcriptional responses across primates and that this divergence is attenuated over time. Finally, we find that relative to AAMs, apes engage a much less specific immune response to different classes of pathogens during the early hours of infection, up-regulating genes typical of anti-viral and anti-bacterial responses regardless of the nature of the stimulus. Overall, these findings suggest apes exhibit increased sensitivity to bacterial and viral immune stimulation, activating a broader array of defense molecules that may be beneficial for early pathogen killing at the potential cost of increased energy expenditure and tissue damage.


Assuntos
Bactérias/imunologia , Metabolismo Energético/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/genética , Vírus/imunologia , Adulto , Animais , Evolução Biológica , Metabolismo Energético/genética , Feminino , Regulação da Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno/genética , Humanos , Macaca mulatta/genética , Macaca mulatta/imunologia , Masculino , Pessoa de Meia-Idade , Pan troglodytes/genética , Pan troglodytes/imunologia , Papio/genética , Papio/imunologia , RNA-Seq , Especificidade da Espécie , Sequenciamento do Exoma , Adulto Jovem
14.
FASEB J ; 35(4): e21312, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33742689

RESUMO

The decrease in the regulatory T cells (Tregs) population is highly involved in adipose tissue inflammation and insulin resistance in obesity. Tregs depend on fatty acids via ß-oxidation for immunosuppressive function adapting their antioxidant systems to allow survival to oxidative stress. In this study, we have hypothesized that a dietary supplementation with alpha-lipoic acid (ALA), a powerful antioxidant, would improve immunometabolism when added to the classical strategy of obesity treatment. First, we showed by in vitro experiments that ALA favors the polarization of mice CD4 + T cells toward Tregs. Next, we have carried out a translational study where female obese mice and women were supplemented with ALA or vehicle/placebo (mice: 2.5 gALA /kgfood ; 6 weeks; women: 600 mgALA /day, 8 weeks) while following a protocol including regular exercise and a change in diet. Fatty acid oxidation potential and activity of nuclear erythroid-related factor 2 (NRF2) of mouse secondary lymphoid tissues were improved by ALA supplementation. ALA reduced visceral adipose tissue (VAT) mass and preserved Tregs in VAT in mice. In women, ALA supplementation induced significant metabolic changes of circulating CD4 + T cells including increased oxidative capacity and fatty acid oxidation, ameliorated their redox status, and improved the reduction of visceral fat mass. While appropriate biological markers are still required to be used in clinics to judge the effectiveness of long-term obesity treatment, further studies in female mice and women are needed to determine whether these immunometabolic changes would reduce VAT mass-associated risk for secondary health issues arising from obesity.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Suplementos Nutricionais , Exercício Físico , Obesidade/terapia , Condicionamento Físico Animal , Ácido Tióctico/farmacologia , Idoso , Animais , Composição Corporal , Linfócitos T CD4-Positivos , Metabolismo Energético/imunologia , Feminino , Teste de Tolerância a Glucose , Humanos , Peroxidação de Lipídeos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Palmitatos/metabolismo , Distribuição Aleatória , Ácido Tióctico/administração & dosagem
15.
Mol Cell Biochem ; 476(6): 2337-2344, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33586093

RESUMO

Sepsis is described as a systemic immune response of the body to an infectious process that might result in dysfunctional organs that may lead to death. In clinical practice, sepsis is considered a medical emergency. The initial event in sepsis caused by a deregulated host response towards harmful microorganisms that leads to an aggravated systemic inflammatory response syndrome (SIRS) to tackle with pathogen invasion and a compensatory anti-inflammatory response syndrome (CARS) that lasts for several days. The inflammatory response and the cellular damage as well as the risk of an organ dysfunction are in direct proportion. Even though, the pathogenesis of sepsis remains unclear, many studies have shown evidence of role of oxidants and antioxidants in sepsis. The altered innate and adaptive immune cell and upregulated production and release of cytokines and chemokines most probably due to involvement of JAK-STAT pathway, disturbance in redox homeostasis due to low clearance of lactate and other oxidative stressors, contributes to sepsis process to organ dysfunction which contribute to increase rates of mortality among these patients. Hence, the treatment strategies for sepsis include antibiotics, ventilator and blood glucose management and other strategies for resuscitation are rapidly progressing. In the current review, we mainly concentrate on throwing light on the main molecular aspects and chemico-biological interactions that shows involvement in pathways manipulating alteration in physiology of immune cells (innate and adaptive) that change the bioenergetics/cellular metabolism to organ dysfunction and correlation of these altered pathway, improve the understating for new therapeutic target for sepsis.


Assuntos
Citocinas/imunologia , Metabolismo Energético/imunologia , Insuficiência de Múltiplos Órgãos/imunologia , Estresse Oxidativo/imunologia , Sepse/imunologia , Humanos , Insuficiência de Múltiplos Órgãos/patologia , Sepse/patologia , Pesquisa Translacional Biomédica
16.
Clin Exp Immunol ; 204(1): 134-143, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33423291

RESUMO

Interferon regulatory factor 5 (IRF5) is a master regulator of macrophage phenotype and a key transcription factor involved in expression of proinflammatory cytokine responses to microbial and viral infection. Here, we show that IRF5 controls cellular and metabolic responses. By integrating ChIP sequencing (ChIP-Seq) and assay for transposase-accessible chromatin using sequencing (ATAC)-seq data sets, we found that IRF5 directly regulates metabolic genes such as hexokinase-2 (Hk2). The interaction of IRF5 and metabolic genes had a functional consequence, as Irf5-/- airway macrophages but not bone marrow-derived macrophages (BMDMs) were characterized by a quiescent metabolic phenotype at baseline and had reduced ability to utilize oxidative phosphorylation after Toll-like receptor (TLR)-3 activation, in comparison to controls, ex vivo. In a murine model of influenza infection, IRF5 deficiency had no effect on viral load in comparison to wild-type controls but controlled metabolic responses to viral infection, as IRF5 deficiency led to reduced expression of Sirt6 and Hk2. Together, our data indicate that IRF5 is a key component of AM metabolic responses following influenza infection and TLR-3 activation.


Assuntos
Metabolismo Energético/imunologia , Regulação da Expressão Gênica/imunologia , Fatores Reguladores de Interferon/imunologia , Macrófagos/imunologia , Sistema Respiratório/citologia , Animais , Células Cultivadas , Sequenciamento de Cromatina por Imunoprecipitação/métodos , Metabolismo Energético/genética , Feminino , Hexoquinase/genética , Hexoquinase/imunologia , Hexoquinase/metabolismo , Humanos , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sirtuínas/genética , Sirtuínas/imunologia , Sirtuínas/metabolismo , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/imunologia , Receptor 3 Toll-Like/metabolismo
17.
Genes (Basel) ; 12(1)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33478058

RESUMO

Mitochondrial integrity and homeostasis in the midgut are key factors controlling mosquito fitness and anti-pathogen resistance. Targeting genes that regulate mitochondrial dynamics represents a potential strategy for limiting mosquito-borne diseases. AMP-activated protein kinase (AMPK) is a key cellular energy sensor found in nearly all eukaryotic cells. When activated, AMPK inhibits anabolic pathways that consume ATP and activates catabolic processes that synthesize ATP. In this study, we overexpressed a truncated and constitutively active α-subunit of AMPK under the control of the midgut-specific carboxypeptidase promotor in the midgut of female Anopheles stephensi. As expected, AMPK overexpression in homozygous transgenic mosquitoes was associated with changes in nutrient storage and metabolism, decreasing glycogen levels at 24 h post-blood feeding when transgene expression was maximal, and concurrently increasing circulating trehalose at the same time point. When transgenic lines were challenged with Plasmodium falciparum, we observed a significant decrease in the prevalence and intensity of infection relative to wild type controls. Surprisingly, we did not observe a significant difference in the survival of adult mosquitoes fed either sugar only or both sugar and bloodmeals throughout adult life. This may be due to the limited period that the transgene was activated before homeostasis was restored. However, we did observe a significant decrease in egg production, suggesting that manipulation of AMPK activity in the mosquito midgut resulted in the re-allocation of resources away from egg production. In summary, this work identifies midgut AMPK activity as an important regulator of metabolism, reproduction, and innate immunity in An. stephensi, a highly invasive and important malaria vector species.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Anopheles/genética , Proteínas de Insetos/genética , Mucosa Intestinal/enzimologia , Malária Falciparum/prevenção & controle , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Animais Geneticamente Modificados , Anopheles/enzimologia , Anopheles/metabolismo , Anopheles/parasitologia , Resistência à Doença/genética , Resistência à Doença/imunologia , Metabolismo Energético/genética , Metabolismo Energético/imunologia , Feminino , Engenharia Genética , Interações Hospedeiro-Parasita/genética , Imunidade Inata/genética , Proteínas de Insetos/metabolismo , Mucosa Intestinal/parasitologia , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Mitocôndrias/metabolismo , Mosquitos Vetores/enzimologia , Mosquitos Vetores/genética , Mosquitos Vetores/metabolismo , Mosquitos Vetores/parasitologia , Plasmodium falciparum/patogenicidade , Reprodução
18.
Genes (Basel) ; 12(1)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33478117

RESUMO

Gene dysfunction and immune cell infiltration play an essential role in the pathogenesis of idiopathic pulmonary arterial hypertension (IPAH). We aimed to investigate the immune landscape and novel differentially expressed genes (DEGs) of IPAH. In addition, potential druggable molecular targets for IPAH were also explored. In this study, the GSE117261 dataset was reanalyzed to explore the immune landscape and hub DEGs of IPAH. Lasso Cox regression analysis and receiver operating characteristic curve analysis were performed to detect the predictive value of IPAH. Additionally, the underlying drug targets for IPAH treatment were determined by drug-gene analysis. IPAH was significantly associated with the transforming growth factor-ß (TGF-ß) signaling pathway and Wnt signaling pathway as well as energetic metabolism dysfunction. We identified 31 upregulated and 39 downregulated DEGs in IPAH patients. Six hub genes, namely, SAA1, CCL5, CXCR1, CXCR2, CCR1, and ADORA3, were related to IPAH pathogenesis regardless of sex differences. Prediction model analysis showed that the area under the curve values of the hub DEGs except CXCR2 were all above 0.9 for distinguishing IPAH patients. In addition, the relative proportions of 5 subtypes of immune cells, namely, CD8+ T cells, CD4+ memory resting T cells, γ delta T cells, M1 macrophages, and resting mast cells, were significantly upregulated in the IPAH samples, while 6 subtypes of immune cells, namely, CD4+ naive T cells, resting NK cells, monocytes, M0 macrophages, activated mast cells, and neutrophils, were downregulated. Additionally, a total of 17 intersecting drugs targeting 5 genes, CCL5, CXCR1, CXCR2, CCR1, and ADORA3, were generated as potential druggable molecular targets for IPAH. Our study revealed the underlying correlations between genes and immune cells in IPAH and demonstrated for the first time that SAA1, CCL5, CXCR1, CCR1, and ADORA3 may be novel genetic targets for IPAH.


Assuntos
Hipertensão Pulmonar Primária Familiar/genética , Regulação da Expressão Gênica/imunologia , Redes Reguladoras de Genes/imunologia , Predisposição Genética para Doença , Análise de Sequência com Séries de Oligonucleotídeos , Biópsia , Estudos de Casos e Controles , Conjuntos de Dados como Assunto , Metabolismo Energético/genética , Metabolismo Energético/imunologia , Hipertensão Pulmonar Primária Familiar/diagnóstico , Hipertensão Pulmonar Primária Familiar/imunologia , Hipertensão Pulmonar Primária Familiar/patologia , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Pulmão/patologia , Masculino , Mapeamento de Interação de Proteínas , Mapas de Interação de Proteínas/genética , Mapas de Interação de Proteínas/imunologia , Fatores de Risco , Transdução de Sinais/genética , Transdução de Sinais/imunologia
19.
Sci China Life Sci ; 64(4): 534-547, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32815067

RESUMO

Mounting evidence has revealed that the therapeutic efficacy of immunotherapies is restricted to a small portion of cancer patients. A deeper understanding of how metabolic reprogramming in the tumor microenvironment (TME) regulates immunity remains a major challenge to tumor eradication. It has been suggested that metabolic reprogramming in the TME may affect metabolism in immune cells and subsequently suppress immune function. Tumor cells compete with infiltrating immune cells for nutrients and metabolites. Notably, the immunosuppressive TME is characterized by catabolic and anabolic processes that are critical for immune cell function, and elevated inhibitory signals may favor cancer immune evasion. The major energy sources that supply different immune cell subtypes also undergo reprogramming. We herein summarize the metabolic remodeling in tumor cells and different immune cell subtypes and the latest advances underlying the use of metabolic checkpoints in antitumor immunotherapies. In this context, targeting both tumor and immune cell metabolic reprogramming may enhance therapeutic efficacy.


Assuntos
Antineoplásicos/uso terapêutico , Reprogramação Celular/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Imunoterapia/métodos , Neoplasias/terapia , Evasão Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Animais , Reprogramação Celular/imunologia , Metabolismo Energético/imunologia , Humanos , Neoplasias/imunologia , Neoplasias/metabolismo , Evasão Tumoral/imunologia , Microambiente Tumoral/imunologia , Macrófagos Associados a Tumor/efeitos dos fármacos , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/metabolismo
20.
Shock ; 55(3): 407-417, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-32826816

RESUMO

ABSTRACT: In activated immune cells, differentiation and function are determined by cell type-specific modifications of metabolic patterns. After traumatic brain injury both immune cell activation and suppression were reported. Therefore, we sought to explore immune cell energy metabolism in a long-term, resuscitated porcine model of acute subdural hematoma (ASDH)-induced acute brain injury devoid of impaired systemic hemodynamics and oxygen transport.Before and up to 50 h after induction of ASDH, peripheral blood mononuclear cells (PBMCs) were separated by density gradient centrifugation, and cell metabolism was analyzed using high-resolution respirometry for mitochondrial respiration and electron spin resonance for reactive oxygen species production. After incubation with stable isotope-labeled 1,2-13C2-glucose or 13C5-glutamine, distinct labeling patterns of intermediates of glycolysis or tricarboxylic acid (TCA) cycle and 13CO2 production were measured by gas chromatography-mass spectroscopy. Principal component analysis was followed by a varimax rotation on the covariance across all measured variables and all measured time points.After ASDH induction, average PBMC metabolic activity remained unaffected, possibly because strict adherence to intensive care unit guidelines limited trauma to ASDH induction without any change in parameters of systemic hemodynamics, oxygen transport, and whole-body metabolism. Despite decreased glycolytic activity fueling the TCA cycle, the principal component analysis indicated a cell type-specific activation pattern with biosynthetic and proliferative characteristics.


Assuntos
Lesões Encefálicas/etiologia , Metabolismo Energético/imunologia , Hematoma Subdural Agudo/complicações , Leucócitos Mononucleares/imunologia , Animais , Leucócitos Mononucleares/metabolismo , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA