Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 156
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 52(4): 620-634.e6, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32268121

RESUMEN

Innate lymphoid cells (ILCs) play an important role in the control and maintenance of barrier immunity. However, chronic activation of ILCs results in immune-mediated pathology. Here, we show that tissue-resident type 2 ILCs (ILC2s) display a distinct metabolic signature upon chronic activation. In the context of allergen-driven airway inflammation, ILC2s increase their uptake of both external lipids and glucose. Externally acquired fatty acids are transiently stored in lipid droplets and converted into phospholipids to promote the proliferation of ILC2s. This metabolic program is imprinted by interleukin-33 (IL-33) and regulated by the genes Pparg and Dgat1, which are both controlled by glucose availability and mTOR signaling. Restricting dietary glucose by feeding mice a ketogenic diet largely ablated ILC2-mediated airway inflammation by impairing fatty acid metabolism and the formation of lipid droplets. Together, these results reveal that pathogenic ILC2 responses require lipid metabolism and identify ketogenic diet as a potent intervention strategy to treat airway inflammation.


Asunto(s)
Alérgenos/administración & dosificación , Asma/dietoterapia , Diacilglicerol O-Acetiltransferasa/inmunología , Dieta Cetogénica/métodos , Interleucina-33/inmunología , Gotas Lipídicas/metabolismo , Subgrupos de Linfocitos T/inmunología , Alternaria/química , Animales , Asma/inducido químicamente , Asma/inmunología , Asma/patología , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Citocinas/administración & dosificación , Diacilglicerol O-Acetiltransferasa/genética , Modelos Animales de Enfermedad , Ácidos Grasos/inmunología , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Glucosa/inmunología , Glucosa/metabolismo , Inmunidad Innata , Interleucina-33/administración & dosificación , Interleucina-33/genética , Interleucinas/administración & dosificación , Gotas Lipídicas/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR gamma/genética , PPAR gamma/inmunología , Papaína/administración & dosificación , Fosfolípidos/inmunología , Fosfolípidos/metabolismo , Cultivo Primario de Células , Subgrupos de Linfocitos T/clasificación , Subgrupos de Linfocitos T/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/inmunología , Linfopoyetina del Estroma Tímico
2.
J Immunol ; 207(11): 2813-2827, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34740958

RESUMEN

Hypoxia-inducible factor-1α (HIF-1α) is an important regulator of glucose metabolism and inflammatory cytokine production in innate immune responses. Viruses modulate HIF-1α to support viral replication and the survival of infected cells, but it is unclear if this transcription factor also plays an important role in regulating antiviral immune responses. In this study, we found that short and long dsRNA differentially engage TLR3, inducing distinct levels of proinflammatory cytokine production (TNF-α and IL-6) in bone marrow-derived macrophages from C57BL/6 mice. These responses are associated with differential accumulation of HIF-1α, which augments NF-κB activation. Unlike TLR4 responses, increased HIF-1α following TLR3 engagement is not associated with significant alterations in glycolytic activity and was more pronounced in low glucose conditions. We also show that the mechanisms supporting HIF-1α stabilization may differ following stimulation with short versus long dsRNA and that pyruvate kinase M2 and mitochondrial reactive oxygen species play a central role in these processes. Collectively, this work suggests that HIF-1α may fine-tune proinflammatory cytokine production during early antiviral immune responses, particularly when there is limited glucose availability or under other conditions of stress. Our findings also suggest we may be able to regulate the magnitude of proinflammatory cytokine production during antiviral responses by targeting proteins or molecules that contribute to HIF-1α stabilization.


Asunto(s)
Citocinas/biosíntesis , Glucosa/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Macrófagos/inmunología , Ácidos Nucleicos/inmunología , Receptor Toll-Like 3/inmunología , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/inmunología
3.
Eur J Immunol ; 51(2): 354-367, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32926407

RESUMEN

Peritoneal dialysis (PD) employs hypertonic glucose to remove excess water and uremic waste. Peritoneal membrane failure limits its long-term use. T-cell cytokines promote this decline. T-cell differentiation is critically determined by the microenvironment. We here study how PD-range hypertonic glucose regulates T-cell polarization and IL-17 production. In the human peritoneal cavity, CD3+ cell numbers increased in PD. Single cell RNA sequencing detected expression of T helper (Th) 17 signature genes RORC and IL23R. In vitro, PD-range glucose stimulated spontaneous and amplified cytokine-induced Th17 polarization. Osmotic controls l-glucose and d-mannose demonstrate that induction of IL-17A is a substance-independent, tonicity dose-dependent process. PD-range glucose upregulated glycolysis and increased the proportion of dysfunctional mitochondria. Blockade of reactive-oxygen species (ROS) prevented IL-17A induction in response to PD-range glucose. Peritoneal mesothelium cultured with IL-17A or IL17F produced pro-inflammatory cytokines IL-6, CCL2, and CX3CL1. In PD patients, peritoneal IL-17A positively correlated with CX3CL1 concentrations. PD-range glucose-stimulated, but neither identically treated Il17a-/- Il17f-/- nor T cells cultured with the ROS scavenger N-acetylcysteine enhanced mesothelial CX3CL1 expression. Our data delineate PD-range hypertonic glucose as a novel inducer of Th17 polarization in a mitochondrial-ROS-dependent manner. Modulation of tonicity-mediated effects of PD solutions may improve membrane survival.


Asunto(s)
Epitelio/inmunología , Glucosa/inmunología , Inflamación/inmunología , Interleucina-17/inmunología , Peritoneo/inmunología , Células Th17/inmunología , Animales , Células Cultivadas , Quimiocina CCL2/inmunología , Quimiocina CXCL1/inmunología , Femenino , Humanos , Interleucina-6/inmunología , Masculino , Manosa/inmunología , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Mitocondrias/inmunología , Diálisis Peritoneal/métodos , Especies Reactivas de Oxígeno/inmunología , Regulación hacia Arriba/inmunología
4.
Proc Natl Acad Sci U S A ; 116(15): 7439-7448, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30910955

RESUMEN

Cellular metabolism and signaling pathways are key regulators to determine conventional T cell fate and function, but little is understood about the role of cell metabolism for natural killer T (NKT) cell survival, proliferation, and function. We found that NKT cells operate distinct metabolic programming from CD4 T cells. NKT cells are less efficient in glucose uptake than CD4 T cells with or without activation. Gene-expression data revealed that, in NKT cells, glucose is preferentially metabolized by the pentose phosphate pathway and mitochondria, as opposed to being converted into lactate. In fact, glucose is essential for the effector functions of NKT cells and a high lactate environment is detrimental for NKT cell survival and proliferation. Increased glucose uptake and IFN-γ expression in NKT cells is inversely correlated with bacterial loads in response to bacterial infection, further supporting the significance of glucose metabolism for NKT cell function. We also found that promyelocytic leukemia zinc finger seemed to play a role in regulating NKT cells' glucose metabolism. Overall, our study reveals that NKT cells use distinct arms of glucose metabolism for their survival and function.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Mitocondrias/metabolismo , Células T Asesinas Naturales/inmunología , Fosforilación Oxidativa , Vía de Pentosa Fosfato/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Glucosa/genética , Glucosa/inmunología , Ratones , Ratones Noqueados , Mitocondrias/genética , Células T Asesinas Naturales/citología , Vía de Pentosa Fosfato/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc/inmunología
5.
FASEB J ; 34(6): 8044-8056, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32307751

RESUMEN

Islet transplantation in man is limited by multiple factors including islet availability, islet cell damage caused by collagenase during isolation, maintenance of islet function between isolation and transplantation, and allograft rejection. In this study, we describe a new approach for preparing islets that enhances islet function in vitro and reduces immunogenicity. The approach involves culture on native decellularized 3D bone marrow-derived extracellular matrix (3D-ECM), which contains many of the matrix components present in pancreas, prior to islet transplantation. Compared to islets cultured on tissue culture plastic (TCP), islets cultured on 3D-ECM exhibited greater attachment, higher survival rate, increased insulin content, and enhanced glucose-stimulated insulin secretion. In addition, culture of islets on 3D-ECM promoted recovery of vascular endothelial cells within the islets and restored basement membrane-related proteins (eg, fibronectin and collagen type VI). More interestingly, culture on 3D-ECM also selectively decontaminated islets of "passenger" cells (co-isolated with the islets) and restored basement membrane-associated type VI collagen, which were associated with an attenuation in islet immunogenicity. These results demonstrate that this novel approach has promise for overcoming two major issues in human islet transplantation: (a) poor yield of islets from donated pancreas tissue and (b) the need for life-long immunosuppression.


Asunto(s)
Membrana Basal/fisiología , Médula Ósea/fisiología , Matriz Extracelular/fisiología , Tolerancia Inmunológica/fisiología , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/fisiología , Animales , Membrana Basal/inmunología , Membrana Basal/metabolismo , Médula Ósea/inmunología , Médula Ósea/metabolismo , Colágeno Tipo VI/inmunología , Colágeno Tipo VI/metabolismo , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Fibronectinas/inmunología , Fibronectinas/metabolismo , Glucosa/inmunología , Glucosa/metabolismo , Tolerancia Inmunológica/inmunología , Insulina/inmunología , Insulina/metabolismo , Secreción de Insulina/inmunología , Secreción de Insulina/fisiología , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Masculino , Ratas , Ratas Endogámicas F344 , Ratas Endogámicas Lew , Ratas Endogámicas WF
6.
Pharmacol Res ; 167: 105576, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33771700

RESUMEN

Macrophages are a type of functionally plastic cells that can create a pro-/anti-inflammatory microenvironment for organs by producing different kinds of cytokines, chemokines, and growth factors to regulate immunity and inflammatory responses. In addition, they can also be induced to adopt different phenotypes in response to extracellular and intracellular signals, a process defined as M1/M2 polarization. Growing evidence indicates that glycobiology is closely associated with this polarization process. In this research, we review studies of the roles of glycosylation, glucose metabolism, and key lectins in the regulation of macrophages function and polarization to provide a new perspective for immunotherapies for multiple diseases.


Asunto(s)
Activación de Macrófagos , Macrófagos/inmunología , Animales , Citocinas/inmunología , Glucosa/inmunología , Glicosilación , Humanos , Inmunidad , Lectinas/inmunología , Macrófagos/citología
7.
J Immunol ; 202(8): 2451-2459, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30850480

RESUMEN

Macrophages, B cells, and adipocytes are among the adipose tissue (AT) APCs that differentiate and activate naive CD4+ T cells. Mice with adipocyte loss of MHC class II (MHC II) are more insulin sensitive. Because macrophages are professional APCs, mice with genetic myeloid MHC II depletion (myeloid MHC II knockout [mMHCII-/-]) were created and metabolically characterized. FITC+ glucan-coated particles (glucan-encapsulated small interfering RNA [siRNA] particles [GeRPs]) were also used to target MHC II knockout specifically in AT macrophages (ATMs). Mice with total body mMHCII-/- were generated by crossing LyzMCre with H2Ab1 floxed mice. For specific ATM depletion of H2Ab1, GeRPs containing H2Ab1 siRNA were administered to high-fat diet-fed C57BL/6 mice. Unexpectedly, mMHCII-/- mice had loss of both macrophage and adipocyte H2Ab1, one of only two Ag-presenting arms; thus, neither cell could present Ag and activate CD4+ T cells. This inability led to a reduction in AT immunosuppressive regulatory T cells, increased AT CD8+ T cells, and no improvement in systemic metabolism. Thus, with combined systemic myeloid and adipocyte MHC II loss, the impact of ATM-specific alterations in APC activity could not be delineated. Therefore, GeRPs containing H2Ab1 siRNA were administered to specifically reduce ATM H2Ab1 which, in contrast, revealed improved glucose tolerance. In conclusion, loss of either ATM or adipocyte APC function, but not both, improves systemic glucose metabolism because of maintenance of AT regulatory T cells.


Asunto(s)
Adipocitos/inmunología , Tejido Adiposo/inmunología , Presentación de Antígeno , Glucosa/inmunología , Macrófagos/inmunología , Adipocitos/citología , Tejido Adiposo/citología , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Glucosa/genética , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Macrófagos/citología , Ratones , Ratones Noqueados
8.
Biotechnol Appl Biochem ; 68(2): 404-410, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32395846

RESUMEN

Release and storage of energy can be regulated by the metabolic parameter dependent on the central nervous system. Macrophages are one of the most professional antigen-presenting cells that are formed by the accumulation of dead or damaged cells or in response to the infection, which has the main function of phagocytosis, secretion of cytokines, and presenting antigen to T cells. A proper immune response is needed for the production of effector cytokines along with comprehensive and rapid cell proliferation and growth. Activation of the immune system and immune cells is needed to increase glucose metabolism. When the immune system responds to pathogens, chemokines inform immune cells such as macrophages and T cells to travel to the infected area. Although glucose is vital for the proper function of immune cells and their proliferation, a high amount of glucose may lead to impaired function of the immune system and pathological conditions. However, a suitable amount of glucose is indispensable for the immune system, but its elevated amount leads to excessive proinflammatory cytokines production. In this study, we focused on the master regulatory role of glucose on the immune system.


Asunto(s)
Proliferación Celular , Citocinas/inmunología , Glucosa/inmunología , Macrófagos/inmunología , Fagocitosis , Linfocitos T/inmunología , Animales , Humanos
9.
Proc Natl Acad Sci U S A ; 115(43): 11042-11047, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30291189

RESUMEN

Sickness behaviors are a conserved set of stereotypic responses to inflammatory diseases. We recently demonstrated that interfering with inflammation-induced anorexia led to metabolic changes that had profound effects on survival of acute inflammatory conditions. We found that different inflammatory states needed to be coordinated with corresponding metabolic programs to actuate tissue-protective mechanisms. Survival of viral inflammation required intact glucose utilization pathways, whereas survival of bacterial inflammation required alternative fuel substrates and ketogenic programs. We thus hypothesized that organismal metabolism would be important in other classes of infectious inflammation and sought to understand its role in the prototypic parasitic disease malaria. Utilizing the cerebral malaria model, Plasmodium berghei ANKA (PbA) infection in C57BL/6J male mice, we unexpectedly found that inhibition of glycolysis using 2-deoxy glucose (2DG) conferred protection from cerebral malaria. Unlike vehicle-treated animals, 2DG-treated animals did not develop cerebral malaria and survived until ultimately succumbing to fatal anemia. We did not find any differences in parasitemia or pathogen load in affected tissues. There were no differences in the kinetics of anemia. We also did not detect differences in immune infiltration in the brain or in blood-brain barrier permeability. Rather, on pathological analyses performed on the entire brain, we found that 2DG prevented the formation of thrombi and thrombotic complications. Using thromboelastography (TEG), we found that 2DG-treated animals formed clots that were significantly less strong and stable. Together, these data suggest that glucose metabolism is involved in inflammation-induced hemostasis and provide a potential therapeutic target in treatment of cerebral malaria.


Asunto(s)
Encéfalo/inmunología , Encéfalo/parasitología , Glucosa/inmunología , Glucosa/metabolismo , Tolerancia Inmunológica/inmunología , Malaria Cerebral/inmunología , Malaria Cerebral/metabolismo , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/parasitología , Modelos Animales de Enfermedad , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/parasitología , Malaria Cerebral/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Parasitemia/inmunología , Plasmodium berghei/inmunología
10.
J Cell Physiol ; 235(4): 3169-3188, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31565806

RESUMEN

Immune cells are bioenergetically expensive during activation, which requires tightly regulated control of metabolic pathways. Both low and high glycemic conditions can modulate immune function. States of undernourishment depress the immune system, and in the same way, excessive intake of nutrients, such as an obesity state, compromise its functioning. Multicellular organisms depend on two mechanisms to survive: the regulation and ability to store energy to prevent starvation and the ability to fight against infection. Synergic interactions between metabolism and immunity affect many systems underpinning human health. In a chronic way, the breakdown of glycemic homeostasis in the body can influence cells of the immune system and consequently contribute to the onset of diseases such as type II diabetes, obesity, Alzheimer's, and fat and lean mass loss. On the contrary, exercise, recognized as a primary strategy to control hyperglycemic disorders, also induces a coordinated immune-neuro-endocrine response that acutely modulates cardiovascular, respiratory, and muscle functions and the immune response to exercise is widely dependent on the intensity and volume that may affect an immunodepressive state. These altered immune responses induced by exercise are modulated through the "stress hormones" adrenaline and cortisol, which are a threat to leukocyte metabolism. In this context, carbohydrates appear to have a positive acute response as a strategy to prevent depression of the immune system by maintaining plasma glucose concentrations to meet the energy demand from all systems involved during strenuous exercises. Therefore, herein, we discuss the mechanisms through which exercise may promotes changes on glycemic homeostasis in the metabolism and how it affects immune cell functions under higher or lower glucose conditions.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Ejercicio Físico/fisiología , Glucosa/metabolismo , Homeostasis/fisiología , Glucosa/inmunología , Homeostasis/inmunología , Hormonas/inmunología , Hormonas/metabolismo , Humanos , Sistema Inmunológico/metabolismo
11.
J Med Virol ; 92(7): 770-775, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32293710

RESUMEN

Coronavirus disease-2019 (COVID-19) infection and its severity can be explained by the concentration of glycosylated severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) viral particles in the lung epithelium, the concentration of glycosylated angiotensin-converting enzyme receptor 2 (ACE2) in the lung epithelium, and the degree and control of the pulmonary immune response to the SARS-CoV-2 spike protein at approximately day 8 to 10 after symptom onset, which may be related to both. Binding of ACE2 by SARS-CoV-2 in COVID-19 also suggests that prolonged uncontrolled hyperglycemia, and not just a history of diabetes mellitus, may be important in the pathogenesis of the disease. It is tempting to consider that the same mechanism acts in COVID-19 as in SARS, where an overactive macrophage M1 inflammatory response, as neutralizing antibodies to the SARS-CoV-2 spike protein form at day 7 to 10, results in acute respiratory distress syndrome (ARDS) in susceptible patients. It also allows consideration of agents, such as hydroxychloroquine, which may interfere with this overly brisk macrophage inflammatory response and perhaps influence the course of the disease, in particular, those that blunt but do not completely abrogate the M1 to M2 balance in macrophage polarization, as well as viral load, which in SARS appears to be temporally related to the onset of ARDS.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/epidemiología , Hidroxicloroquina/uso terapéutico , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/epidemiología , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/epidemiología , Enzima Convertidora de Angiotensina 2 , Anticuerpos Neutralizantes/biosíntesis , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Azitromicina/uso terapéutico , Betacoronavirus/efectos de los fármacos , Betacoronavirus/metabolismo , Betacoronavirus/patogenicidad , COVID-19 , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/metabolismo , Glucosa/inmunología , Glucosa/metabolismo , Glicosilación/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/metabolismo , Incidencia , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/virología , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/inmunología , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/complicaciones , Neumonía Viral/metabolismo , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo
12.
Acta Biochim Biophys Sin (Shanghai) ; 52(7): 768-775, 2020 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-32445465

RESUMEN

Interleukin-33 (IL-33) is produced by various types of cells under physical or pathological conditions. As a multifunctional partner in health and disease, current evidence reveals that IL-33 also participates in several metabolic processes. IL-33 has been proven to contribute to regulating the activity of ST2+ group 2 innate lymphoid cells and regulatory T cells in adipose, which leads to the shift of insulin sensitivity and glucose clearance in glucose metabolism, thermogenesis, and adipocyte beiging in adipose metabolism. In this review, we briefly summarize the biological characteristics of Il-33 and discuss its regulatory function in glucose and adipose metabolism. By clarifying the underlying mechanism of IL-33, we highlight the crosstalk between immune response and metabolic processes mediated by IL-33.


Asunto(s)
Tejido Adiposo , Glucosa , Resistencia a la Insulina/inmunología , Interleucina-33 , Termogénesis/inmunología , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Animales , Glucosa/inmunología , Glucosa/metabolismo , Humanos , Interleucina-33/inmunología , Interleucina-33/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
13.
J Biol Chem ; 293(25): 9706-9717, 2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29716995

RESUMEN

Mycobacteria synthesize intracellular, 6-O-methylglucose-containing lipopolysaccharides (mGLPs) proposed to modulate bacterial fatty acid metabolism. Recently, it has been shown that Mycobacterium tuberculosis mGLP specifically induces a specific subset of protective γ9δ2 T cells. Mild base treatment, which removes all the base-labile groups, reduces the specific activity of mGLP required for induction of these T cells, suggesting that acylation of the saccharide moieties is required for γ9δ2 T-cell activation. On the basis of this premise, we used analytical LC/MS and NMR methods to identify and locate the acyl functions on the mGLP saccharides. We found that mGLP is heterogeneous with respect to acyl functions and contains acetyl, isobutyryl, succinyl, and octanoyl groups and that all acylations in mGLP, except for succinyl and octanoyl residues, reside on the glucosyl residues immediately following the terminal 3-O-methylglucose. Our analyses also indicated that the octanoyl residue resides at position 2 of an internal glucose toward the reducing end. LC/MS analysis of the residual product obtained by digesting the mGLP with pancreatic α-amylase revealed that the product is an oligosaccharide terminated by α-(1→4)-linked 6-O-methyl-d-glucosyl residues. This oligosaccharide retained none of the acyl groups, except for the octanoyl group, and was unable to induce protective γ9δ2 T cells. This observation confirmed that mGLP induces γ9δ2 T cells and indicated that the acylated glucosyl residues at the nonreducing terminus of mGLP are required for this activity.


Asunto(s)
Antígenos Bacterianos/inmunología , Glucosa/química , Lipopolisacáridos/química , Mycobacterium tuberculosis/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Subgrupos de Linfocitos T/inmunología , Glucosa/inmunología , Glucosa/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Activación de Linfocitos
14.
J Biol Chem ; 293(16): 5934-5946, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29496993

RESUMEN

Chronic low-grade inflammation in the pancreatic islets is observed in individuals with type 2 diabetes, and macrophage levels are elevated in the islets of these individuals. However, the molecular mechanisms underlying the interactions between the pancreatic ß cells and macrophages and their involvement in inflammation are not fully understood. Here, we investigated the role of S100 calcium-binding protein A8 (S100A8), a member of the damage-associated molecular pattern molecules (DAMPs), in ß-cell inflammation. Co-cultivation of pancreatic islets with unstimulated peritoneal macrophages in the presence of palmitate (to induce lipotoxicity) and high glucose (to induce glucotoxicity) synergistically increased the expression and release of islet-produced S100A8 in a Toll-like receptor 4 (TLR4)-independent manner. Consistently, a significant increase in the expression of the S100a8 gene was observed in the islets of diabetic db/db mice. Furthermore, the islet-derived S100A8 induced TLR4-mediated inflammatory cytokine production by migrating macrophages. When human islet cells were co-cultured with U937 human monocyte cells, the palmitate treatment up-regulated S100A8 expression. This S100A8-mediated interaction between islets and macrophages evoked ß-cell apoptosis, which was ameliorated by TLR4 inhibition in the macrophages or S100A8 neutralization in the pancreatic islets. Of note, both glucotoxicity and lipotoxicity triggered S100A8 secretion from the pancreatic islets, which in turn promoted macrophage infiltration of the islets. Taken together, a positive feedback loop between islet-derived S100A8 and macrophages drives ß-cell apoptosis and pancreatic islet inflammation. We conclude that developing therapeutic approaches to inhibit S100A8 may serve to prevent ß-cell loss in patients with diabetes.


Asunto(s)
Apoptosis , Calgranulina A/inmunología , Inflamación/inmunología , Células Secretoras de Insulina/inmunología , Macrófagos/inmunología , Animales , Línea Celular , Células Cultivadas , Glucosa/inmunología , Humanos , Células Secretoras de Insulina/citología , Macrófagos/citología , Masculino , Ratones Endogámicos C57BL , Palmitatos/inmunología , Transducción de Señal , Receptor Toll-Like 4/inmunología
15.
Biochem Biophys Res Commun ; 517(2): 227-232, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31331642

RESUMEN

Interleukin-6 (IL-6) has become a target of interest for drug development aiming to treat diabetic retinopathy. Since IL-6 signaling can promote beneficial as well as detrimental effects via two different signaling pathways, the objective of the present study was to investigate the effects of classical IL-6 and IL-6 trans-signaling on human Müller cells (HMC), which are important for the development of diabetic retinopathy. HMCs were cultured in normal (5 mmol/L) and high (25 mmol/L) glucose plus or minus IL-6 or IL-6/sIL-6R. IL-6 receptor expression using immunohistochemistry and flow cytometry and cytokine release using magnetic bead assays were determined. HMCs express the membrane bound form of the IL-6 receptor (mIL-6R), gp130, and can release the soluble forms sIL-6R and sgp130 demonstrating that HMCs are capable of responding to classical IL-6 and IL-6 trans-signaling. IL-6 protected HMCs from glucose toxicity via VEGF-A signaling. IL-6/sIL-6R caused only modest protection, which was not mediated by VEGF-A. Our data show for the first time that classical IL-6 signaling exerts its beneficial effects through VEGF-A action contrary to IL-6 trans-signaling, which was VEGF-A independent. These results have clinical implications for drug development targeting IL-6 since strict anti-IL-6 therapies might further decrease neuroretinal functions in the diabetic retina.


Asunto(s)
Glucosa/inmunología , Interleucina-6/inmunología , Retina/inmunología , Factor A de Crecimiento Endotelial Vascular/inmunología , Células Cultivadas , Receptor gp130 de Citocinas/inmunología , Retinopatía Diabética/inmunología , Humanos , Inflamación/inmunología , Receptores de Interleucina-6/inmunología , Retina/citología , Transducción de Señal
16.
Cancer Immunol Immunother ; 68(1): 11-22, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30229289

RESUMEN

Primary and secondary lymphoid organs are heavily innervated by the autonomic nervous system. Norepinephrine, the primary neurotransmitter secreted by post-ganglionic sympathetic neurons, binds to and activates ß-adrenergic receptors expressed on the surface of immune cells and regulates the functions of these cells. While it is known that both activated and memory CD8+ T-cells primarily express the ß2-adrenergic receptor (ß2-AR) and that signaling through this receptor can inhibit CD8+ T-cell effector function, the mechanism(s) underlying this suppression is not understood. Under normal activation conditions, T-cells increase glucose uptake and undergo metabolic reprogramming. In this study, we show that treatment of murine CD8+ T-cells with the pan ß-AR agonist isoproterenol (ISO) was associated with a reduced expression of glucose transporter 1 following activation, as well as decreased glucose uptake and glycolysis compared to CD8+ T-cells activated in the absence of ISO. The effect of ISO was specifically dependent upon ß2-AR, since it was not seen in adrb2-/- CD8+ T-cells and was blocked by the ß-AR antagonist propranolol. In addition, we found that mitochondrial function in CD8+ T-cells was also impaired by ß2-AR signaling. This study demonstrates that one mechanism by which ß2-AR signaling can inhibit CD8+ T-cell activation is by suppressing the required metabolic reprogramming events which accompany activation of these immune cells and thus reveals a new mechanism by which adrenergic stress can suppress the effector activity of immune cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos/inmunología , Receptores Adrenérgicos beta 2/inmunología , Transducción de Señal/inmunología , Agonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Femenino , Glucosa/inmunología , Glucosa/metabolismo , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/inmunología , Isoproterenol/farmacología , Activación de Linfocitos/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Propranolol/farmacología , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/efectos de los fármacos
17.
Mol Biol Rep ; 46(4): 3991-3999, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31168669

RESUMEN

Mesenchymal stem cells (MSCs) are self-renewing multipotent cells with immunoregulatory function, which makes them attractive candidates for regenerative medicine. However, the detailed mechanisms of their immunomodulatory capacity are not fully characterized. Here, we found that casein kinase 2 interacting protein-1 (CKIP-1) expression was induced in the murine MSC cell line C3H/10T1/2 by LPS. Knockdown of CKIP-1 did not cause significant differences on the cell cycle or immunophenotype of MSCs. However, MSCs with CKIP-1 knockdown showed enhanced immunosuppressive capacity. Real-time PCR and western blot analyses revealed that compared with the control group, MSCs with CKIP-1-knockdown exhibited higher IL-10 production and p38 MAPK phosphorylation following LPS treatment. Interestingly, the expression of CKIP-1 was decreased in MSCs following high glucose treatment. Furthermore, MSCs became more immunosuppressive after high glucose treatment, as shown by higher IL-10 production and enhanced inhibition of T cell proliferation. Collectively, our data reveal a novel role for CKIP-1 in regulating MSC-mediated immunomodulation, and indicate that MSCs become more immunosuppressive under high glucose conditions. These new insights may help in the development of future applications of MSCs.


Asunto(s)
Proteínas Portadoras/inmunología , Factores Inmunológicos/metabolismo , Células Madre Mesenquimatosas/inmunología , Animales , Proteínas Portadoras/metabolismo , Diferenciación Celular/inmunología , Línea Celular , Proliferación Celular/fisiología , Citocinas/inmunología , Glucosa/inmunología , Glucosa/metabolismo , Inmunomodulación/inmunología , Inmunofenotipificación/métodos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL
18.
Int J Mol Sci ; 20(10)2019 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-31126070

RESUMEN

A correlation between gastrointestinal (GI) inflammation and gut hormones has reported that inflammatory stimuli including bacterial endotoxins, lipopolysaccharides (LPS), TNFα, IL-1ß, and IL-6 induces high levels of incretin hormone leading to glucose dysregulation. Although incretin hormones are immediately secreted in response to environmental stimuli, such as nutrients, cytokines, and LPS, but studies of glucose-induced incretin secretion in an inflamed state are limited. We hypothesized that GI inflammatory conditions induce over-stimulated incretin secretion via an increase of glucose-sensing receptors. To confirm our hypothesis, we observed the alteration of glucose-induced incretin secretion and glucose-sensing receptors in a GI inflammatory mouse model, and we treated a conditioned media (Mϕ 30%) containing inflammatory cytokines in intestinal epithelium cells and enteroendocrine L-like NCI-H716 cells. In GI-inflamed mice, we observed that over-stimulated incretin secretion and insulin release in response to glucose and sodium glucose cotransporter (Sglt1) was increased. Incubation with Mϕ 30% increases Sglt1 and induces glucose-induced GLP-1 secretion with increasing intracellular calcium influx. Phloridzin, an sglt1 inhibitor, inhibits glucose-induced GLP-1 secretion, ERK activation, and calcium influx. These findings suggest that the abnormalities of incretin secretion leading to metabolic disturbances in GI inflammatory disease by an increase of Sglt1.


Asunto(s)
Gastroenteritis/inmunología , Glucosa/inmunología , Insulina/inmunología , Transportador 1 de Sodio-Glucosa/inmunología , Animales , Línea Celular , Células Cultivadas , Femenino , Polipéptido Inhibidor Gástrico/inmunología , Gastroenteritis/patología , Péptido 1 Similar al Glucagón/inmunología , Incretinas/inmunología , Inflamación/inmunología , Inflamación/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Ratones Endogámicos C57BL
19.
Cell Physiol Biochem ; 47(1): 403-413, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29788012

RESUMEN

BACKGROUND/AIMS: Allograft inflammatory factor-1 (AIF-1) is an inflammatory cytokine produced mainly by macrophages within human white adipose tissue. Its expression is increased in obese subjects and positively correlated with insulin resistance. The purpose of this study is to characterize the regulatory role of AIF-1 in insulin signaling of adipocyte. METHODS: AIF-1 was over-expressed via transfection of AIF-1 cDNA into murine RAW 264.7 macrophages, and the constitutive expression of AIF-1 was decreased via transfection of targeting siRNA. Murine 3T3L1 adipocytes were treated with macrophage-conditioned medium or AIF-1 protein. Intracellular lipid accumulation was assayed by oil red O stain. Reactive oxygen species production was determinated by a flow cytometer and adipokine secretion was measured with ELISA. Glucose uptake was detected using the glucose oxidase method and insulin-signal-transduction related molecules were analyzed by Western blot. RESULTS: Short term (48 h) AIF-1 treatment slightly promoted intracellular lipid storage in differentiating 3T3L1 cells. The protein stimulated reactive oxygen species production, provoked TNFα, IL6, resistin, but suppressed adiponectin release and insulin-stimulated glucose uptake both under normal basal and insulin resistance conditions. Furthermore, AIF-1 induced NF-κB activation, inhibited PPARγ expression, GLUT4 translocation to plasma membrane and Akt phosphorylation. CONCLUSION: Macrophage-derived AIF-1 up-regulated reactive oxygen species production, adipokine TNFα, IL6, resistin release, and inhibited adiponectin secretion. Moreover, it suppressed insulin-stimulated glucose uptake by down-regulating insulin signaling. Thus, AIF-1 could be related to obesity-related diseases.


Asunto(s)
Adipocitos/inmunología , Proteínas de Unión al Calcio/inmunología , Inflamación/inmunología , Insulina/inmunología , Macrófagos/inmunología , Proteínas de Microfilamentos/inmunología , Transducción de Señal , Células 3T3-L1 , Animales , Glucosa/inmunología , Ratones , Células RAW 264.7 , Especies Reactivas de Oxígeno/inmunología
20.
Cell Physiol Biochem ; 45(6): 2351-2368, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29554649

RESUMEN

BACKGROUND/AIMS: Recent researches highlighted the protective potential of pioglitazone, a PPAR-γ agonist, in the progression of cerebral ischemia-reperfusion injury. However, there has been no study on the application of pioglitazone in treating ischemic stroke through mechanisms involving pyroptosis. METHODS: The cerebral injury was established by middle cerebral artery occlusion (MCAO). in vitro ischemia in primary cultured astrocytes was induced by the oxygen-glucose deprivation (OGD). ELISA and Western Blot analysis were employed to the levels of PPAR-γ, pyroptosis-related biomarkers and cytoplasmic translocation of HMGB-1 and RAGE expression as well as Rac1 activity, respectively. RESULTS: We demonstrated that repeated intraperitoneal administration of pioglitazone remarkably reduced the infarct volume, improved neurological deficits and suppressed the Rac1 activity with significant reduction of excessive ROS in rat model of middle cerebral artery occlusion (MCAO). Moreover, pioglitazone alleviated the up-regulation of pyroptosis-related biomarkers and the increased cytoplasmic translocation of HMGB-1 and RAGE expression in cerebral penumbra cortex. Similarly, the protective effects of pioglitazone on cultured astrocytes were characterized by reduced Rac1 activity, pyroptosis related protein expressions and lactate dehydrogenase (LDH) release. However, these protective effects of pioglitazone were neutralized with the use of GW9662, a PPAR-γ inhibitor. Interestingly, Rac1 knockdown in lentivirus with the Rac1 small hair RNA (shRNA) could inhibit the OGD-induced pyroptosis of primary cultured astrocytes. Furthermore, the combination of Rac1-shRNA and pioglitazone can further strengthen the inhibitory effects on pyroptosis induced by OGD. CONCLUSION: The neuroprotection of pioglitazone was attributable to the alleviated ischemia/hypoxia-induced pyroptosis and was also associated with the PPARγ-mediated suppression of HGMB-1/RAGE signaling pathway. Moreover, the inhibition of Rac1 promoted this function.


Asunto(s)
Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Piroptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/uso terapéutico , Animales , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Astrocitos/patología , Células Cultivadas , Glucosa/inmunología , Proteína HMGB1/inmunología , Infarto de la Arteria Cerebral Media/inmunología , Infarto de la Arteria Cerebral Media/patología , Masculino , Neuroprotección/efectos de los fármacos , PPAR gamma/inmunología , Pioglitazona , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/inmunología , Receptor para Productos Finales de Glicación Avanzada/inmunología , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Proteína de Unión al GTP rac1/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA