Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 82(15): 2815-2831.e5, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35752171

RESUMEN

Protein import into mitochondria is a highly regulated process, yet how cells clear mitochondria undergoing dysfunctional protein import remains poorly characterized. Here we showed that mitochondrial protein import stress (MPIS) triggers localized LC3 lipidation. This arm of the mitophagy pathway occurs through the Nod-like receptor (NLR) protein NLRX1 while, surprisingly, without the engagement of the canonical mitophagy protein PINK1. Mitochondrial depolarization, which itself induces MPIS, also required NLRX1 for LC3 lipidation. While normally targeted to the mitochondrial matrix, cytosol-retained NLRX1 recruited RRBP1, a ribosome-binding transmembrane protein of the endoplasmic reticulum, which relocated to the mitochondrial vicinity during MPIS, and the NLRX1/RRBP1 complex in turn controlled the recruitment and lipidation of LC3. Furthermore, NLRX1 controlled skeletal muscle mitophagy in vivo and regulated endurance capacity during exercise. Thus, localization and lipidation of LC3 at the site of mitophagosome formation is a regulated step of mitophagy controlled by NLRX1/RRBP1 in response to MPIS.


Asunto(s)
Proteínas Mitocondriales , Mitofagia , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Transporte de Proteínas
2.
Nat Immunol ; 18(1): 26-35, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27893701

RESUMEN

TRAF1 is a signaling adaptor known for its role in tumor necrosis factor receptor-induced cell survival. Here we show that monocytes from healthy human subjects with a rheumatoid arthritis-associated single-nucleotide polymorphism (SNP) in the TRAF1 gene express less TRAF1 protein but greater amounts of inflammatory cytokines in response to lipopolysaccharide (LPS). The TRAF1 MATH domain binds directly to three components of the linear ubiquitination (LUBAC) complex, SHARPIN, HOIP and HOIL-1, to interfere with the recruitment and linear ubiquitination of NEMO. This results in decreased NF-κB activation and cytokine production, independently of tumor necrosis factor. Consistent with this, Traf1-/- mice show increased susceptibility to LPS-induced septic shock. These findings reveal an unexpected role for TRAF1 in negatively regulating Toll-like receptor signaling, providing a mechanistic explanation for the increased inflammation seen with a disease-associated TRAF1 SNP.


Asunto(s)
Artritis Reumatoide/genética , Leucocitos Mononucleares/inmunología , Monocitos/inmunología , Transducción de Señal , Factor 1 Asociado a Receptor de TNF/metabolismo , Animales , Citocinas/metabolismo , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo de Nucleótido Simple , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Factor 1 Asociado a Receptor de TNF/genética , Receptores Toll-Like/metabolismo
3.
Immunity ; 47(5): 943-958.e9, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29150240

RESUMEN

T cell antigen-presenting cell (APC) interactions early during chronic viral infection are crucial for determining viral set point and disease outcome, but how and when different APC subtypes contribute to these outcomes is unclear. The TNF receptor superfamily (TNFRSF) member GITR is important for CD4+ T cell accumulation and control of chronic lymphocytic choriomeningitis virus (LCMV). We found that type I interferon (IFN-I) induced TNFSF ligands GITRL, 4-1BBL, OX40L, and CD70 predominantly on monocyte-derived APCs and CD80 and CD86 predominantly on classical dendritic cells (cDCs). Mice with hypofunctional GITRL in Lyz2+ cells had decreased LCMV-specific CD4+ T cell accumulation and increased viral load. GITR signals in CD4+ T cells occurred after priming to upregulate OX40, CD25, and chemokine receptor CX3CR1. Thus IFN-I (signal 3) induced a post-priming checkpoint (signal 4) for CD4+ T cell accumulation, revealing a division of labor between cDCs and monocyte-derived APCs in regulating T cell expansion.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Coriomeningitis Linfocítica/inmunología , Factores de Necrosis Tumoral/análisis , Animales , Ligando CD27/análisis , Receptor 1 de Quimiocinas CX3C/análisis , Células Dendríticas/inmunología , Femenino , Proteína Relacionada con TNFR Inducida por Glucocorticoide/análisis , Proteína Relacionada con TNFR Inducida por Glucocorticoide/fisiología , Glicoproteínas de Membrana/análisis , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Ligando OX40
4.
J Immunol ; 210(5): 531-535, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36637223

RESUMEN

Secretion of IL-1ß, a potent cytokine that plays a key role in gout pathogenesis, is regulated by inflammasomes. TRAF1 has been linked to heightened risk to inflammatory arthritis. In this article, we show that TRAF1 negatively regulates inflammasome activation to limit caspase-1 and IL-1ß secretion in human and mouse macrophages. TRAF1 reduces linear ubiquitination and subsequent oligomerization of the adapter protein, ASC. i.p. injection of monosodium urate crystals resulted in increased inflammatory cell infiltrates and IL-1ß production in Traf1 knockout mice compared with wild type littermates. In a model of monosodium urate crystal-induced gout, Traf1 knockout mice exhibited more swelling in the knee joints, increased infiltration of inflammatory cells, and higher expression of proinflammatory cytokines. In summary, this study identifies TRAF1 as a key regulator of IL-1ß production and a potential therapeutic target for inflammasome-driven diseases such as gout.


Asunto(s)
Gota , Inflamasomas , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales , Citocinas , Interleucina-1beta , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Factor 1 Asociado a Receptor de TNF/genética , Ácido Úrico
5.
Am J Physiol Cell Physiol ; 326(4): C1011-C1026, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38145301

RESUMEN

Fibrosis is associated with respiratory and limb muscle atrophy in Duchenne muscular dystrophy (DMD). Current standard of care partially delays the progression of this myopathy but there remains an unmet need to develop additional therapies. Adiponectin receptor agonism has emerged as a possible therapeutic target to lower inflammation and improve metabolism in mdx mouse models of DMD but the degree to which fibrosis and atrophy are prevented remain unknown. Here, we demonstrate that the recently developed slow-release peptidomimetic adiponectin analog, ALY688-SR, remodels the diaphragm of murine model of DMD on DBA background (D2.mdx) mice treated from days 7-28 of age during early stages of disease. ALY688-SR also lowered interleukin-6 (IL-6) mRNA but increased IL-6 and transforming growth factor-ß1 (TGF-ß1) protein contents in diaphragm, suggesting dynamic inflammatory remodeling. ALY688-SR alleviated mitochondrial redox stress by decreasing complex I-stimulated H2O2 emission. Treatment also attenuated fibrosis, fiber type-specific atrophy, and in vitro diaphragm force production in diaphragm suggesting a complex relationship between adiponectin receptor activity, muscle remodeling, and force-generating properties during the very early stages of disease progression in murine model of DMD on DBA background (D2.mdx) mice. In tibialis anterior, the modest fibrosis at this young age was not altered by treatment, and atrophy was not apparent at this young age. These results demonstrate that short-term treatment of ALY688-SR in young D2.mdx mice partially prevents fibrosis and fiber type-specific atrophy and lowers force production in the more disease-apparent diaphragm in relation to lower mitochondrial redox stress and heterogeneous responses in certain inflammatory markers. These diverse muscle responses to adiponectin receptor agonism in early stages of DMD serve as a foundation for further mechanistic investigations.NEW & NOTEWORTHY There are limited therapies for the treatment of Duchenne muscular dystrophy. As fibrosis involves an accumulation of collagen that replaces muscle fibers, antifibrotics may help preserve muscle function. We report that the novel adiponectin receptor agonist ALY688-SR prevents fibrosis in the diaphragm of D2.mdx mice with short-term treatment early in disease progression. These responses were related to altered inflammation and mitochondrial functions and serve as a foundation for the development of this class of therapy.


Asunto(s)
Distrofia Muscular de Duchenne , Animales , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Adiponectina/genética , Modelos Animales de Enfermedad , Interleucina-6/metabolismo , Ratones Endogámicos C57BL , Peróxido de Hidrógeno/metabolismo , Receptores de Adiponectina/genética , Receptores de Adiponectina/metabolismo , Ratones Endogámicos DBA , Músculo Esquelético/metabolismo , Diafragma/metabolismo , Fibrosis , Inflamación/metabolismo , Progresión de la Enfermedad , Atrofia/metabolismo , Atrofia/patología
6.
J Immunol ; 208(2): 286-292, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-35017218

RESUMEN

Inflammasomes are important in human health and disease, whereby they control the secretion of IL-1ß and IL-18, two potent proinflammatory cytokines that play a key role in inflammatory responses to pathogens and danger signals. Several inflammasomes have been discovered over the past two decades. NLRP3 inflammasome is the best characterized and can be activated by a wide variety of inducers. It is composed of a sensor, NLRP3, an adapter protein, ASC, and an effector enzyme, caspase-1. After activation, caspase-1 mediates the cleavage and secretion of bioactive IL-1ß and IL-18 via gasdermin-D pores in the plasma membrane. Aberrant activation of NLRP3 inflammasomes has been implicated in a multitude of human diseases, including inflammatory, autoimmune, and metabolic diseases. Therefore, several mechanisms have evolved to control their activity. In this review, we describe the posttranslational modifications that regulate NLRP3 inflammasome components, including ubiquitination, phosphorylation, and other forms of posttranslational modifications.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Procesamiento Proteico-Postraduccional/genética , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Caspasa 1/genética , Caspasa 1/metabolismo , Membrana Celular/metabolismo , Activación Enzimática/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteínas de Unión a Fosfato/metabolismo , Fosforilación/fisiología , Ubiquitinación/fisiología
7.
Am J Physiol Cell Physiol ; 325(2): C429-C442, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37306389

RESUMEN

Despite its importance in protecting the host from infections and injury, excessive inflammation may lead to serious human diseases including autoimmune disorders, cardiovascular diseases, diabetes, and cancer. Exercise is a known immunomodulator; however, whether exercise causes long-term changes in inflammatory responses and how these changes occur are lacking. Here, we show that chronic moderate-intensity training of mice leads to persistent metabolic rewiring and changes to chromatin accessibility in bone marrow-derived macrophages (BMDMs), which, in turn, tempers their inflammatory responses. We show that BMDMs from exercised mice exhibited a decrease in lipopolysaccharide (LPS)-induced NF-κB activation and proinflammatory gene expression along with an increase in M2-like-associated genes when compared with BMDMs from sedentary mice. This was associated with improved mitochondrial quality and increased reliance on oxidative phosphorylation accompanied with reduced mitochondrial reactive oxygen species (ROS) production. Mechanistically, assay for transposase-accessible chromatin (ATAC)-seq analysis showed changes in chromatin accessibility of genes associated with inflammatory and metabolic pathways. Overall, our data suggest that chronic moderate exercise can influence the inflammatory responses of macrophages by reprogramming their metabolic and epigenetic landscape.NEW & NOTEWORTHY In this study, we explain how long-term moderate exercise training can reduce inflammation in mouse macrophages by reprogramming the way they sense and respond to the presence of pathogens. We completed a thorough analysis and showed that these changes persist in macrophages because exercise improves the ability of cells to utilize oxygen without producing damaging compounds, and changes the way they access their DNA.


Asunto(s)
Macrófagos , Inmunidad Entrenada , Humanos , Animales , Ratones , Macrófagos/metabolismo , Inflamación/metabolismo , Transducción de Señal , Cromatina/metabolismo , Lipopolisacáridos/farmacología
8.
Exp Physiol ; 108(9): 1108-1117, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37415288

RESUMEN

NEW FINDINGS: What is the central question of this study? Can adiponectin receptor agonism improve recognition memory in a mouse model of Duchenne muscular dystrophy? What is the main finding and its importance? Short-term treatment with the new adiponectin receptor agonist ALY688 improves recognition memory in D2.mdx mice. This finding suggests that further investigation into adiponectin receptor agonism is warranted, given that there remains an unmet need for clinical approaches to treat this cognitive dysfunction in people with Duchenne muscular dystrophy. ABSTRACT: Memory impairments have been well documented in people with Duchenne muscular dystrophy (DMD). However, the underlying mechanisms are poorly understood, and there is an unmet need to develop new therapies to treat this condition. Using a novel object recognition test, we show that recognition memory impairments in D2.mdx mice are completely prevented by daily treatment with the new adiponectin receptor agonist ALY688 from day 7 to 28 of age. In comparison to age-matched wild-type mice, untreated D2.mdx mice demonstrated lower hippocampal mitochondrial respiration (carbohydrate substrate), greater serum interleukin-6 cytokine content and greater hippocampal total tau and Raptor protein contents. Each of these measures was partly or fully preserved after treatment with ALY688. Collectively, these results indicate that adiponectin receptor agonism improves recognition memory in young D2.mdx mice.


Asunto(s)
Distrofia Muscular de Duchenne , Animales , Ratones , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/metabolismo , Ratones Endogámicos mdx , Receptores de Adiponectina/metabolismo , Receptores de Adiponectina/uso terapéutico , Adiponectina/metabolismo , Respiración , Modelos Animales de Enfermedad , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Músculo Esquelético/metabolismo
9.
J Immunol ; 195(1): 210-216, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26019270

RESUMEN

IFNs, which transduce pivotal signals through Stat1 and Stat2, effectively suppress the replication of Legionella pneumophila in primary murine macrophages. Although the ability of IFN-γ to impede L. pneumophila growth is fully dependent on Stat1, IFN-αß unexpectedly suppresses L. pneumophila growth in both Stat1- and Stat2-deficient macrophages. New studies demonstrating that the robust response to IFN-αß is lost in Stat1-Stat2 double-knockout macrophages suggest that Stat1 and Stat2 are functionally redundant in their ability to direct an innate response toward L. pneumophila. Because the ability of IFN-αß to signal through Stat1-dependent complexes (i.e., Stat1-Stat1 and Stat1-Stat2 dimers) has been well characterized, the current studies focus on how Stat2 is able to direct a potent response to IFN-αß in the absence of Stat1. These studies reveal that IFN-αß is able to drive the formation of a Stat2 and IFN regulatory factor 9 complex that drives the expression of a subset of IFN-stimulated genes, but with substantially delayed kinetics. These observations raise the possibility that this pathway evolved in response to microbes that have devised strategies to subvert Stat1-dependent responses.


Asunto(s)
Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/inmunología , Legionelosis/inmunología , Macrófagos/inmunología , Receptor de Interferón alfa y beta/inmunología , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT2/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/microbiología , Células de la Médula Ósea/patología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Interferón gamma/genética , Interferón gamma/inmunología , Legionella pneumophila/inmunología , Legionelosis/genética , Legionelosis/microbiología , Legionelosis/patología , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cultivo Primario de Células , Multimerización de Proteína , Receptor de Interferón alfa y beta/genética , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT2/deficiencia , Factor de Transcripción STAT2/genética , Transducción de Señal , Factores de Tiempo
10.
EMBO Rep ; 14(10): 900-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24008845

RESUMEN

The cyclic dinucleotides 3'-5'diadenylate (c-diAMP) and 3'-5' diguanylate (c-diGMP) are important bacterial second messengers that have recently been shown to stimulate the secretion of type I Interferons (IFN-Is) through the c-diGMP-binding protein MPYS/STING. Here, we show that physiologically relevant levels of cyclic dinucleotides also stimulate a robust secretion of IL-1ß through the NLRP3 inflammasome. Intriguingly, this response is independent of MPYS/STING. Consistent with most NLRP3 inflammasome activators, the response to c-diGMP is dependent on the mobilization of potassium and calcium ions. However, in contrast to other NLRP3 inflammasome activators, this response is not associated with significant changes in mitochondrial potential or the generation of mitochondrial reactive oxygen species. Thus, cyclic dinucleotides activate the NLRP3 inflammasome through a unique pathway that could have evolved to detect pervasive bacterial pathogen-associated molecular patterns associated with intracellular infections.


Asunto(s)
Proteínas Portadoras/metabolismo , GMP Cíclico/análogos & derivados , Fosfatos de Dinucleósidos/farmacología , Inflamasomas/metabolismo , Animales , Calcio/metabolismo , Proteínas Portadoras/genética , Línea Celular Tumoral , GMP Cíclico/farmacología , Humanos , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Potencial de la Membrana Mitocondrial , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Potasio/metabolismo , Especies Reactivas de Oxígeno/metabolismo
11.
Biomed J ; 47(1): 100655, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37598797

RESUMEN

Nod-like receptors (NLRs) are innate immune receptors that play a key role in sensing components from pathogens and from damaged cells or organelles. NLRs form signaling complexes that can lead to activation of transcription factors or effector caspases - by means of inflammasome activation -Inflammatory arthritis (IA) culminating in promoting inflammation. An increasing body of research supports the role of NLRs in driving pathogenesis of IA, a collection of diseases that include rheumatoid arthritis (RA), psoriatic arthritis (PsA), ankylosing spondylitis, and pediatric arthritis. In this review, we briefly discuss the main drivers of IA diseases and dive into the evidence for - and against - various NLRs in driving these diseases. We also review the studies examining the use of NLR and inflammasome inhibitors as potential therapies for IA.


Asunto(s)
Artritis Psoriásica , Artritis Reumatoide , Espondilitis Anquilosante , Humanos , Niño , Artritis Psoriásica/patología , Artritis Psoriásica/terapia , Inflamasomas , Proteínas NLR
12.
Int Immunopharmacol ; 132: 111890, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38547772

RESUMEN

The diverse beneficial effects of adiponectin-receptor signaling, including its impact on the regulation of inflammatory processes in vivo, have resulted in development of adiponectin receptor agonists as a treatment for metabolic disorders. However, there are no established non-invasive bioassays for detection of adiponectin target engagement in humans or animal models. Here, we designed an assay using small amounts of blood to assess adiponectin action. Specifically, we tested effects of the small 10-amino acid peptide adiponectin receptor agonist, ALY688, in a sublethal LPS endotoxemia model in mice. LPS-induced pro-inflammatory cytokine levels in serum were significantly reduced in mice treated with ALY688, assessed via multiplex ELISA in flow cytometry. Furthermore, ALY688 alone significantly induced TGF-ß release in serum 1 h after treatment and was elevated for up to 24 h. Additionally, using a flow-cytometry panel for detection of changes in circulating immune cell phenotypes, we observed a significant increase in absolute T cell counts in mice after ALY688 treatment. To assess changes in intracellular signaling effectors downstream of adiponectin, phospho-flow cytometry was conducted. There was a significant increase in phosphorylation of AMPK and p38-MAPK in mice after ALY688 treatment. We then used human donor immune cells (PBMCs) treated with ALY688 ex vivo and observed elevation of AMPK and p38-MAPK phosphorylation from baseline in response to ALY688. Together, these results indicate we can detect adiponectin action on immune cells in vivo by assessing adiponectin signaling pathway for AMPK and p38-MAPK, as well as pro-inflammatory cytokine levels. This new approach provides a blood-based bioassay for screening adiponectin action.


Asunto(s)
Adiponectina , Citocinas , Lipopolisacáridos , Ratones Endogámicos C57BL , Transducción de Señal , Animales , Adiponectina/sangre , Adiponectina/metabolismo , Humanos , Transducción de Señal/efectos de los fármacos , Lipopolisacáridos/farmacología , Ratones , Masculino , Citocinas/metabolismo , Citocinas/sangre , Bioensayo/métodos , Endotoxemia/inmunología , Endotoxemia/metabolismo , Receptores de Adiponectina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Modelos Animales de Enfermedad , Femenino
13.
Cell Death Dis ; 14(4): 252, 2023 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-37029101

RESUMEN

Iron overload is associated with various pathological changes which contribute to metabolic syndrome, many of which have been proposed to occur via damaging tissue through an excessive amount of reactive oxygen species (ROS) production. In this study, we established a model of iron overload in L6 skeletal muscle cells and observed that iron enhanced cytochrome c release from depolarized mitochondria, assayed by immunofluorescent colocalization of cytochrome c with Tom20 and the use of JC-1, respectively. This subsequently elevated apoptosis, determined via use of a caspase-3/7 activatable fluorescent probe and western blotting for cleaved caspase-3. Using CellROX deep red and mBBr, we observed that iron increased generation of reactive oxygen species (ROS), and that pretreatment with the superoxide dismutase mimetic MnTBAP reduced ROS production and attenuated iron-induced intrinsic apoptosis and cell death. Furthermore, using MitoSox Red we observed that iron enhanced mROS and the mitochondria-targeted anti-oxidant SKQ1 reduced iron-induced ROS generation and cell death. Western blotting for LC3-II and P62 levels as well as immunofluorescent detection of autophagy flux with LC3B and P62 co-localization indicated that iron acutely (2-8 h) activated and later (12-24 h) attenuated autophagic flux. We used autophagy-deficient cell models generated by overexpressing a dominant-negative Atg5 mutant or CRISPR-mediated ATG7 knock out to test the functional significance of autophagy and observed that autophagy-deficiency exacerbated iron-induced ROS production and apoptosis. In conclusion, our study showed that high iron levels promoted ROS production, blunted the self-protective autophagy response and led to cell death in L6 skeletal muscle cells.


Asunto(s)
Biopelículas , Sobrecarga de Hierro , Humanos , Especies Reactivas de Oxígeno/metabolismo , Caspasa 3/genética , Caspasa 3/metabolismo , Citocromos c , Reactores Biológicos , Autofagia , Apoptosis , Hierro , Músculo Esquelético/metabolismo
14.
PLoS One ; 18(9): e0287741, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37713400

RESUMEN

While inflammation is an important immune response for protection from infections, excessive or prolonged inflammation can lead to a variety of debilitating diseases including skin disease, diabetes, heart disease, stroke, autoimmune diseases and cancer. Inflammation is a graded response that is typically initiated when resident macrophages sense the presence of pathogens or damage in the tissue and produce inflammatory cytokines and chemokines to kill the pathogen, clear debris and dead tissue, and initiate tissue repair. Here we show that copper-infused fabrics can prevent inflammation by blocking the production of inflammatory cytokines from macrophages after being exposed to LPS, a component of bacterial cell wall. Mechanistically, we show that copper-infused fabrics can significantly reduce the NF-κB and IRF3 activation in LPS-stimulated macrophages. Given the importance of excessive inflammation in diabetes, we show that copper can reduce insulin resistance mediated by inflammatory cytokines in muscle cells. Our data show that copper infused fabrics may be useful to reduce excessive inflammation in macrophages and improve insulin sensitivity in skeletal muscles.


Asunto(s)
Cobre , Resistencia a la Insulina , Humanos , Cobre/farmacología , Lipopolisacáridos , Citocinas , Inflamación , Macrófagos
15.
Infect Immun ; 80(12): 4232-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22988022

RESUMEN

Bacterial infections of the mucosal epithelium are a major cause of human disease. The prolonged presence of microbial pathogens stimulates inflammation of the local tissues, which leads to changes in the molecular composition of the extracellular milieu. A well-characterized molecule that is released to the extracellular milieu by stressed or infected cells is extracellular ATP and its ecto-enzymatic degradation products, which function as signaling molecules through ligation of purinergic receptors. There has been little information, however, on the effects of the extracellular metabolites on bacterial growth in inflamed tissues. Millimolar concentrations of ATP have been previously shown to inhibit irreversibly bacterial infection through ligation of P2X(7) receptors. We show here that the proinflammatory mediator, ATP, is released from Chlamydia trachomatis-infected epithelial cells. Moreover, further stimulation of the infected cells with micromolar extracellular ADP or ATP significantly impairs the growth of the bacteria, with a profile characteristic of the involvement of P2X(4) receptors. A specific role for P2X(4) was confirmed using cells overexpressing P2X(4). The chlamydiae remain viable and return to normal growth kinetics after removal of the extracellular stimulus, similar to responses previously described for persistence of chlamydial infection.


Asunto(s)
Adenosina Trifosfato/farmacología , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/patogenicidad , Células Epiteliales/microbiología , Receptores Purinérgicos P2X4/efectos de los fármacos , Receptores Purinérgicos P2X4/metabolismo , Adenosina Trifosfato/metabolismo , Chlamydia trachomatis/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos
16.
Methods Mol Biol ; 2459: 65-72, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35212954

RESUMEN

A growing body of work has recently highlighted the pivotal role of mitochondria in the initiation and modulation of inflammasome activation. Specifically, mitochondrial dysfunction can induce NLRP3 inflammasome activation, where loss of mitochondrial potential leads to production of reactive oxygen species (ROS) and release of Ca2+, which in turn trigger inflammasome assembly. Therefore, several measures of mitochondrial parameters and components are routinely utilized in studies assessing mechanisms of inflammasome activation. In this chapter, we show detailed protocols on how to employ flow cytometry using three distinct mitochondria-specific dyes to measure mitochondrial ROS (MitoSOX), mitochondrial respiration (Mitotracker deep red), and total mitochondria (Mitotracker green), as well as a dye that measures reduced glutathione (mBBr ).


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Biopelículas , Reactores Biológicos , Citometría de Flujo , Mitocondrias , Especies Reactivas de Oxígeno
17.
Methods Mol Biol ; 2459: 73-78, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35212955

RESUMEN

Apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) is an adaptor protein that is essential for the activation of several inflammasome complexes. Activation of inflammasomes leads to pathogenic clearance and inflammatory cell death called pyroptosis. Upon inflammasome activation, ASC oligomerization leads to the recruitment and activation of caspase-1, which in turn converts pro-inflammatory cytokines (e.g., pro-IL-1ß, pro-IL-18) to their mature active form. Given its central role in inflammasome activation, ASC oligomerization is used as an indicator of inflammasome activation. Here we describe how ASC oligomerization can be detected by Western blotting.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Western Blotting , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis
18.
Nutrients ; 14(21)2022 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-36364794

RESUMEN

High-intensity/impact exercise elicits a transient increase in inflammatory biomarkers. Consuming nutrient-dense wholefoods, like milk, following exercise may modulate post-exercise inflammation and aid recovery. We examined the effect of post-exercise skim milk consumption (versus an isoenergetic, isovolumetric carbohydrate [CHO] drink) on acute exercise-induced inflammation in untrained females. Using a randomized crossover design, 13 healthy females (age = 20 ± 2.3 y; BMI = 21.0 ± 1.1 kg/m2) completed two bouts of combined resistance/plyometric exercise followed by either skim milk (MILK) or CHO at 5-min and 1 h post-exercise. Serum interleukin [IL]-1ß, IL-6, IL-10, and tumor necrosis factor-alpha (TNF-α) concentrations were measured at pre-exercise, 15-min, 75-min, 24 h, and 48 h post-exercise. IL-6 increased 15-min post-exercise vs. all other timepoints (time effect, p = 0.017). Between 24 and 48 h, IL-10 decreased and increased in the MILK and CHO conditions, respectively (interaction, p = 0.018). There were no significant effects for IL-1ß or TNF-α. Relative concentrations of IL-1ß (p = 0.049) and IL-10 (p = 0.028) at 48 h post-exercise were lower in MILK vs. CHO. Milk post-exercise did not influence the absolute concentration of pro-inflammatory cytokines; however, there were divergent responses for the anti-inflammatory cytokine, IL-10, and milk reduced the relative inflammatory response at 48 h (vs. CHO) for IL-1ß and IL-10. This demonstrates the potential for milk to modulate inflammation post-exercise in this sample.


Asunto(s)
Interleucina-10 , Ejercicio Pliométrico , Adolescente , Adulto , Femenino , Humanos , Adulto Joven , Citocinas , Inflamación , Interleucina-6 , Factor de Necrosis Tumoral alfa , Estudios Cruzados
19.
Cells ; 12(1)2022 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-36611912

RESUMEN

Iron overload (IO) is associated with cardiovascular diseases, including heart failure. Our study's aim was to examine the mechanism by which IO triggers cell death in H9c2 cells. IO caused accumulation of intracellular and mitochondrial iron as shown by the use of iron-binding fluorescent reporters, FerroOrange and MitoFerroFluor. Expression of cytosolic and mitochondrial isoforms of Ferritin was also induced by IO. IO-induced iron accumulation and cellular ROS was rapid and temporally linked. ROS accumulation was detected in the cytosol and mitochondrial compartments with CellROX, DCF-DA and MitoSOX fluorescent dyes and partly reversed by the general antioxidant N-acetyl cysteine or the mitochondrial antioxidant SkQ1. Antioxidants also reduced the downstream activation of apoptosis and lytic cell death quantified by Caspase 3 cleavage/activation, mitochondrial Cytochrome c release, Annexin V/Propidium iodide staining and LDH release of IO-treated cells. Finally, overexpression of MitoNEET, an outer mitochondrial membrane protein involved in the transfer of Fe-S clusters between mitochondrial and cytosol, was observed to lower iron and ROS accumulation in the mitochondria. These alterations were correlated with reduced IO-induced cell death by apoptosis in MitoNEET-overexpressing cells. In conclusion, IO mediates H9c2 cell death by causing mitochondrial iron accumulation and subsequent general and mitochondrial ROS upregulation.


Asunto(s)
Antioxidantes , Sobrecarga de Hierro , Humanos , Especies Reactivas de Oxígeno/metabolismo , Antioxidantes/metabolismo , Muerte Celular , Mitocondrias/metabolismo , Hierro/metabolismo , Sobrecarga de Hierro/metabolismo
20.
J Appl Physiol (1985) ; 133(6): 1381-1393, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36356257

RESUMEN

Exercise is one of the only nonpharmacological remedies known to counteract genetic and chronic diseases by enhancing health and improving life span. Although the many benefits of regular physical activity have been recognized for some time, the intricate and complex signaling systems triggered at the onset of exercise have only recently begun to be uncovered. Exercising muscles initiate a coordinated, multisystemic, metabolic rewiring, which is communicated to distant organs by various molecular mediators. The field of exercise research has been expanding beyond the musculoskeletal system, with interest from industry to provide realistic models and exercise mimetics that evoke a whole body rejuvenation response. The 18th International Biochemistry of Exercise conference took place in Toronto, Canada, from May 25 to May 28, 2022, with more than 400 attendees. Here, we provide an overview of the most cutting-edge exercise-related research presented by 66 speakers, focusing on new developments in topics ranging from molecular and cellular mechanisms of exercise adaptations to exercise therapy and management of disease and aging. We also describe how the manipulation of these signaling pathways can uncover therapeutic avenues for improving human health and quality of life.


Asunto(s)
Ejercicio Físico , Calidad de Vida , Humanos , Ejercicio Físico/fisiología , Adaptación Fisiológica , Envejecimiento/fisiología , Terapia por Ejercicio , Músculo Esquelético/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA