Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 25(1): 155-165, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38102487

RESUMEN

In mouse peritoneal and other serous cavities, the transcription factor GATA6 drives the identity of the major cavity resident population of macrophages, with a smaller subset of cavity-resident macrophages dependent on the transcription factor IRF4. Here we showed that GATA6+ macrophages in the human peritoneum were rare, regardless of age. Instead, more human peritoneal macrophages aligned with mouse CD206+ LYVE1+ cavity macrophages that represent a differentiation stage just preceding expression of GATA6. A low abundance of CD206+ macrophages was retained in C57BL/6J mice fed a high-fat diet and in wild-captured mice, suggesting that differences between serous cavity-resident macrophages in humans and mice were not environmental. IRF4-dependent mouse serous cavity macrophages aligned closely with human CD1c+CD14+CD64+ peritoneal cells, which, in turn, resembled human peritoneal CD1c+CD14-CD64- cDC2. Thus, major populations of serous cavity-resident mononuclear phagocytes in humans and mice shared common features, but the proportions of different macrophage differentiation stages greatly differ between the two species, and dendritic cell (DC2)-like cells were especially prominent in humans.


Asunto(s)
Macrófagos Peritoneales , Macrófagos , Humanos , Ratones , Animales , Ratones Endogámicos C57BL , Macrófagos/metabolismo , Macrófagos Peritoneales/metabolismo , Diferenciación Celular , Células Dendríticas
2.
Development ; 149(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35178561

RESUMEN

Tissue-resident macrophages are increasingly recognized as important determinants of organ homeostasis, tissue repair, remodeling and regeneration. Although the ontogeny and function of tissue-resident macrophages has been identified as distinct from postnatal hematopoiesis, the inability to specify, in vitro, similar populations that recapitulate these developmental waves has limited our ability to study their function and potential for regenerative applications. We took advantage of the concept that tissue-resident macrophages and monocyte-derived macrophages originate from distinct extra-embryonic and definitive hematopoietic lineages to devise a system to generate pure cultures of macrophages that resemble tissue-resident or monocyte-derived subsets. We demonstrate that human pluripotent stem cell-derived extra-embryonic-like and intra-embryonic-like hematopoietic progenitors differentiate into morphologically, transcriptionally and functionally distinct macrophage populations. Single-cell RNA sequencing of developing and mature cultures uncovered distinct developmental trajectories and gene expression programs of macrophages derived from extra-embryonic-like and intra-embryonic-like hematopoietic progenitors. These findings establish a resource for the generation of human tissue resident-like macrophages to study their specification and function under defined conditions and to explore their potential use in tissue engineering and regenerative medicine applications.


Asunto(s)
Macrófagos , Células Madre Pluripotentes , Diferenciación Celular/genética , Hematopoyesis , Homeostasis , Humanos , Macrófagos/metabolismo
3.
Immunity ; 45(4): 817-830, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27760338

RESUMEN

Macrophage activation status is intrinsically linked to metabolic remodeling. Macrophages stimulated by interleukin 4 (IL-4) to become alternatively (or, M2) activated increase fatty acid oxidation and oxidative phosphorylation; these metabolic changes are critical for M2 activation. Enhanced glucose utilization is also characteristic of the M2 metabolic signature. Here, we found that increased glucose utilization is essential for M2 activation. Increased glucose metabolism in IL-4-stimulated macrophages required the activation of the mTORC2 pathway, and loss of mTORC2 in macrophages suppressed tumor growth and decreased immunity to a parasitic nematode. Macrophage colony stimulating factor (M-CSF) was implicated as a contributing upstream activator of mTORC2 in a pathway that involved PI3K and AKT. mTORC2 operated in parallel with the IL-4Rα-Stat6 pathway to facilitate increased glycolysis during M2 activation via the induction of the transcription factor IRF4. IRF4 expression required both mTORC2 and Stat6 pathways, providing an underlying mechanism to explain how glucose utilization is increased to support M2 activation.


Asunto(s)
Factores Reguladores del Interferón/metabolismo , Activación de Macrófagos/fisiología , Macrófagos/fisiología , Complejos Multiproteicos/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Interleucina-4/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción STAT6/metabolismo
4.
Circ Res ; 133(3): 200-219, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37350264

RESUMEN

BACKGROUND: The mTOR (mechanistic target of rapamycin) pathway is a complex signaling cascade that regulates cellular growth, proliferation, metabolism, and survival. Although activation of mTOR signaling has been linked to atherosclerosis, its direct role in lesion progression and in plaque macrophages remains poorly understood. We previously demonstrated that mTORC1 (mTOR complex 1) activation promotes atherogenesis through inhibition of autophagy and increased apoptosis in macrophages. METHODS: Using macrophage-specific Rictor- and mTOR-deficient mice, we now dissect the distinct functions of mTORC2 pathways in atherogenesis. RESULTS: In contrast to the atheroprotective effect seen with blockade of macrophage mTORC1, macrophage-specific mTORC2-deficient mice exhibit an atherogenic phenotype, with larger, more complex lesions and increased cell death. In cultured macrophages, we show that mTORC2 signaling inhibits the FoxO1 (forkhead box protein O1) transcription factor, leading to suppression of proinflammatory pathways, especially the inflammasome/IL (interleukin)-1ß response, a key mediator of vascular inflammation and atherosclerosis. In addition, administration of FoxO1 inhibitors efficiently rescued the proinflammatory response caused by mTORC2 deficiency both in vitro and in vivo. Interestingly, collective deletion of macrophage mTOR, which ablates mTORC1- and mTORC2-dependent pathways, leads to minimal change in plaque size or complexity, reflecting the balanced yet opposing roles of these signaling arms. CONCLUSIONS: Our data provide the first mechanistic details of macrophage mTOR signaling in atherosclerosis and suggest that therapeutic measures aimed at modulating mTOR need to account for its dichotomous functions.


Asunto(s)
Aterosclerosis , Serina-Treonina Quinasas TOR , Ratones , Animales , Diana Mecanicista del Complejo 2 de la Rapamicina , Serina-Treonina Quinasas TOR/metabolismo , Macrófagos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Factores de Transcripción/metabolismo , Aterosclerosis/genética , Aterosclerosis/metabolismo
5.
Hepatology ; 77(6): 1968-1982, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36059147

RESUMEN

BACKGROUND AND AIMS: Nucleotide-binding oligomerization domain-like receptor-family pyrin domain-containing 3 (NLRP3) inflammasome activation has been shown to result in liver fibrosis. Mechanisms and downstream signaling remain incompletely understood. Here, we studied the role of IL-18 in hepatic stellate cells (HSCs), and its impact on liver fibrosis. APPROACH AND RESULTS: We observed significantly increased serum levels of IL-18 (128.4 pg/ml vs. 74.9 pg/ml) and IL-18 binding protein (BP; 46.50 ng/ml vs. 15.35 ng/ml) in patients with liver cirrhosis compared with healthy controls. Single cell RNA sequencing data showed that an immunoregulatory subset of murine HSCs highly expresses Il18 and Il18r1 . Treatment of cultured primary murine HSC with recombinant mouse IL-18 accelerated their transdifferentiation into myofibroblasts. In vivo , IL-18 receptor-deficient mice had reduced liver fibrosis in a model of fibrosis induced by HSC-specific NLRP3 overactivation. Whole liver RNA sequencing analysis from a murine model of severe NASH-induced fibrosis by feeding a choline-deficient, L-amino acid-defined, high fat diet showed that genes related to IL-18 and its downstream signaling were significantly upregulated, and Il18-/- mice receiving this diet for 10 weeks showed protection from fibrotic changes with decreased number of alpha smooth muscle actin-positive cells and collagen deposition. HSC activation triggered by NLRP3 inflammasome activation was abrogated when IL-18 signaling was blocked by its naturally occurring antagonist IL-18BP. Accordingly, we observed that the severe inflammatory phenotype associated with myeloid cell-specific NLRP3 gain-of-function was rescued by IL-18BP. CONCLUSIONS: Our study highlights the role of IL-18 in the development of liver fibrosis by its direct effect on HSC activation identifying IL-18 as a target to treat liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas , Inflamasomas , Ratones , Animales , Inflamasomas/metabolismo , Células Estrelladas Hepáticas/metabolismo , Interleucina-18 , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Cirrosis Hepática/patología , Fibrosis , Proteínas Portadoras/metabolismo , Hígado/patología
6.
Immunity ; 40(1): 91-104, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24439267

RESUMEN

Cardiac macrophages are crucial for tissue repair after cardiac injury but are not well characterized. Here we identify four populations of cardiac macrophages. At steady state, resident macrophages were primarily maintained through local proliferation. However, after macrophage depletion or during cardiac inflammation, Ly6c(hi) monocytes contributed to all four macrophage populations, whereas resident macrophages also expanded numerically through proliferation. Genetic fate mapping revealed that yolk-sac and fetal monocyte progenitors gave rise to the majority of cardiac macrophages, and the heart was among a minority of organs in which substantial numbers of yolk-sac macrophages persisted in adulthood. CCR2 expression and dependence distinguished cardiac macrophages of adult monocyte versus embryonic origin. Transcriptional and functional data revealed that monocyte-derived macrophages coordinate cardiac inflammation, while playing redundant but lesser roles in antigen sampling and efferocytosis. These data highlight the presence of multiple cardiac macrophage subsets, with different functions, origins, and strategies to regulate compartment size.


Asunto(s)
Macrófagos/inmunología , Monocitos/fisiología , Miocarditis/inmunología , Miocardio/inmunología , Animales , Presentación de Antígeno , Antígenos Ly/metabolismo , Muerte Celular , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Desarrollo Fetal , Corazón/embriología , Homeostasis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/inmunología , Fagocitosis , Receptores CCR2/metabolismo , Transcriptoma , Saco Vitelino/citología
7.
Curr Opin Lipidol ; 33(5): 295-299, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35942818

RESUMEN

PURPOSE OF REVIEW: Nonalcoholic steatohepatitis (NASH) is a multisystem disease that affects not only the liver but also heart, pancreas, and kidney. We currently lack a comprehensive understanding of mechanisms responsible for the development of NASH-associated cardiomyopathy or the influence of sex on pathophysiology. There is a critical need to address these gaps in knowledge in order to accelerate translation of knowledge into clinical practice. RECENT FINDINGS: NASH and cardiovascular disease share common risk factors such as chronic inflammation, hyperlipidemia, and insulin resistance. Early cardiac dysfunction in NASH that is independent of obesity or other cardiometabolic risk factors suggests roles for liver-heart crosstalk in disease pathogenesis. Inflammation is a driving force in the pathogenesis of NASH, and it is likely that 'spill over' of NASH inflammation contributes to the development of cardiomyopathy. However, molecular and cellular mechanisms that mediate NASH-associated cardiomyopathy remain unclear because of inherent limitations of experimental models. Even so, recent studies implicate inflammatory, metabolic, and physiologic mechanisms that enhance our understanding of NASH-associated cardiomyopathy and the role of liver-heart crosstalk. SUMMARY: An innovative, detailed, and mechanistic understanding of NASH-associated cardiomyopathy is relevant to public health and will be fundamental for the comprehensive care of these patients.


Asunto(s)
Cardiomiopatías , Enfermedad del Hígado Graso no Alcohólico , Humanos , Inflamación/complicaciones , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo
8.
Hepatology ; 73(1): 437-448, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32740968

RESUMEN

Single cell transcriptomics has emerged as a powerful lens through which to study the molecular diversity of complex tissues such as the liver, during health and disease, both in animal models and in humans. The earliest gene expression methods measured bulk tissue RNA, but the results were often confusing because they derived from the combined transcriptomes of many different cell types in unknown proportions. To better delineate cell-type-specific expression, investigators developed cell isolation, purification, and sorting protocols, yet still, the RNA derived from ensembles of cells obscured recognition of cellular heterogeneity. Profiling transcriptomes at the single-cell level has opened the door to analyses that were not possible in the past. In this review, we discuss the evolution of single cell transcriptomics and how it has been applied for the study of liver physiology and pathobiology to date.


Asunto(s)
Perfilación de la Expresión Génica , Hígado/patología , Hígado/fisiología , Análisis de la Célula Individual , Animales , Humanos , Análisis de Secuencia de ARN
9.
Am J Transplant ; 21(2): 809-814, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33089906

RESUMEN

Chimeric antigen receptor T cells (CAR-T) are genetically modified T cells with a chimeric antigen receptor directed against a specific tumor-associated antigen like CD19 in lymphoma. CAR-T cells have shown encouraging activity against recurrent and refractory diffuse large B cell lymphomas (DLBCL). However concurrent use of immunosuppressive agents was prohibited in most CAR-T trials effectively excluding patients with prior solid organ transplantation (SOT) and posttransplant lymphoproliferative disorders (PTLD). We report the outcomes for three patients with PTLD refractory to immunochemotherapy 10-20 years after SOT who received CAR-T therapy between January 2018 and December 2019. One patient had an orthotopic heart transplant, the second had a deceased donor kidney transplant, and the third had a pancreas after kidney transplant (PAK). All patients developed complications of CAR-T therapy such as cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and acute kidney injury requiring renal replacement therapy in the two out of three patients. All patients expired after withdrawal of care due to lack of response to CAR-T therapy. In addition, the PAK patient developed acute pancreatitis after CAR-T therapy. This case series identifies the challenges of using CAR-T therapy to manage refractory PTLD in SOT recipients and its possible complications.


Asunto(s)
Trastornos Linfoproliferativos , Trasplante de Órganos , Pancreatitis , Receptores Quiméricos de Antígenos , Enfermedad Aguda , Humanos , Trastornos Linfoproliferativos/etiología , Trastornos Linfoproliferativos/terapia , Trasplante de Órganos/efectos adversos
10.
Clin Transplant ; 35(12): e14471, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34546591

RESUMEN

BACKGROUND: Previous studies in heart transplantation have shown an association between institutional case volume and outcomes. We aim to determine the case volume associated with optimal 1-year survival after transplantation. METHODS AND RESULTS: The United Network for Organ Sharing (UNOS) national database was analyzed for adult patients who underwent orthotopic heart transplantation between January 2013 and December 2017. A total of 11,196 cases at 128 transplant centers were included. Risk-adjusted restricted cubic splines revealed a non-linear association between institutional case volume and 1-year post-transplant survival. In the risk-adjusted, random-effect Cox model with segmented linear splines, higher heart transplant volume up to 24 cases per year was associated with better 1-year survival (HR = .978 every additional case, 95% CI .963-.993), and optimal survival was maintained between 24 and 38 cases per year. However, further increase in volume above 38 transplants per year was associated with mildly decreased 1-year survival (HR = 1.007 every additional case, 95% CI 1.002-1.013). CONCLUSIONS: The relationship between institutional case volume and heart transplant 1-year survival is non-linear, with optimal survival observed at institutional case volume of 24-38 cases per year.


Asunto(s)
Trasplante de Corazón , Adulto , Bases de Datos Factuales , Supervivencia de Injerto , Humanos , Modelos de Riesgos Proporcionales , Sistema de Registros , Estudios Retrospectivos , Resultado del Tratamiento
11.
Clin Transplant ; 35(6): e14296, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33759249

RESUMEN

Acute kidney injury is a common complication following heart transplantation, and the factors contributing to acute kidney injury are not well understood. We conducted a retrospective cohort study evaluating patients who underwent heart transplantation between 2009 and 2016 at a single institution. The primary endpoint was incidence of acute kidney injury as defined by Kidney Disease Improving Global Outcomes criteria. Secondary endpoints included 30-day hospital readmission, 30-day mortality, and 1-year mortality. A total of 228 heart transplant patients were included in the study for analysis. In total, 145 (64%) developed acute kidney injury, where 43 (30%) were classified as stage I, 28 (19%) as stage II, and 74 (51%) as stage III. Risk factors found to be associated with the presence of acute kidney injury included increased use of vasopressors and inotropes post-transplant. Protective factors included cardiopulmonary bypass time <170 min. Acute kidney injury was found to be associated with increased 30-day and 1-year mortality.


Asunto(s)
Lesión Renal Aguda , Trasplante de Corazón , Lesión Renal Aguda/epidemiología , Lesión Renal Aguda/etiología , Trasplante de Corazón/efectos adversos , Humanos , Incidencia , Complicaciones Posoperatorias/etiología , Estudios Retrospectivos , Factores de Riesgo
12.
J Immunol ; 201(7): 2054-2069, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30143592

RESUMEN

Obesity and diabetes modulate macrophage activation, often leading to prolonged inflammation and dysfunctional tissue repair. Increasing evidence suggests that the NLRP3 inflammasome plays an important role in obesity-associated inflammation. We have previously shown that activation of the lipotoxic inflammasome by excess fatty acids in macrophages occurs via a lysosome-dependent pathway. However, the mechanisms that link cellular lipid metabolism to altered inflammation remain poorly understood. PPARγ is a nuclear receptor transcription factor expressed by macrophages that is known to alter lipid handling, mitochondrial function, and inflammatory cytokine expression. To undercover novel links between metabolic signaling and lipotoxic inflammasome activation, we investigated mouse primary macrophages deficient in PPARγ. Contrary to our expectation, PPARγ knockout (KO) macrophages released significantly less IL-1ß and IL-1α in response to lipotoxic stimulation. The suppression occurred at the transcriptional level and was apparent for multiple activators of the NLRP3 inflammasome. RNA sequencing revealed upregulation of IFN-ß in activated PPARγKO macrophages, and this was confirmed at the protein level. A blocking Ab against the type 1 IFNR restored the release of IL-1ß to wild type levels in PPARγKO cells, confirming the mechanistic link between these events. Conversely, PPARγ activation with rosiglitazone selectively suppressed IFN-ß expression in activated macrophages. Loss of PPARγ also resulted in diminished expression of genes involved in sterol biosynthesis, a pathway known to influence IFN production. Together, these findings demonstrate a cross-talk pathway that influences the interplay between metabolism and inflammation in macrophages.


Asunto(s)
Inflamasomas/metabolismo , Inflamación/inmunología , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/fisiología , Obesidad/inmunología , PPAR gamma/genética , Animales , Células Cultivadas , Interferón Tipo I/metabolismo , Activación de Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Rosiglitazona/farmacología , Análisis de Secuencia de ARN
13.
Semin Immunol ; 28(5): 417-424, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27771140

RESUMEN

Macrophages are heterogeneous cells that play a key role in inflammatory and tissue reparative responses. Over the past decade it has become clear that shifts in cellular metabolism are important determinants of macrophage function and phenotype. At the same time, our appreciation of macrophage diversity in vivo has also been increasing. Factors such as cell origin and tissue localization are now recognized as important variables that influence macrophage biology. Whether different macrophage populations also have unique metabolic phenotypes has not been extensively explored. In this article, we will discuss the importance of understanding how macrophage origin can modulate metabolic programming and influence inflammatory responses.


Asunto(s)
Metabolismo Energético , Inmunomodulación , Macrófagos/inmunología , Macrófagos/metabolismo , Animales , Regulación de la Expresión Génica , Humanos , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Macrófagos/citología , Redes y Vías Metabólicas , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Fenotipo
14.
Basic Res Cardiol ; 113(5): 38, 2018 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-30097758

RESUMEN

The CHRNA5 gene encodes a neurotransmitter receptor subunit involved in multiple processes, including cholinergic autonomic nerve activity and inflammation. Common variants in CHRNA5 have been linked with atherosclerotic cardiovascular disease. Association of variation in CHRNA5 and specific haplotypes with cardiovascular outcomes has not been described. The aim of this study was to examine the association of CHRNA5 haplotypes with gene expression and mortality among patients with acute myocardial infarction (AMI) and explore potential mechanisms of this association. Patients (N = 2054) hospitalized with AMI were genotyped for two common variants in CHRNA5. Proportional hazard models were used to estimate independent association of CHRNA5 haplotype with 1-year mortality. Both individual variants were associated with mortality (p = 0.0096 and 0.0004, respectively) and were in tight LD (D' = 0.99). One haplotype, HAP3, was associated with decreased mortality one year after AMI (adjusted HR = 0.42, 95% CI 0.26, 0.68; p = 0.0004). This association was validated in an independent cohort (N = 637) of post-MI patients (adjusted HR = 0.23, 95% CI 0.07, 0.79; p = 0.019). Differences in CHRNA5 expression by haplotype were investigated in human heart samples (n = 28). Compared with non-carriers, HAP3 carriers had threefold lower cardiac CHRNA5 mRNA expression (p = 0.023). Circulating levels of the inflammatory marker hsCRP were significantly lower in HAP3 carriers versus non-carriers (3.43 ± 4.2 versus 3.91 ± 5.1; p = 0.0379). Activation of the inflammasome, an important inflammatory complex involved in cardiovascular disease that is necessary for release of the pro-inflammatory cytokine IL-1 ß, was assessed in bone marrow-derived macrophages (BMDM) from CHRNA5 knockout mice and wild-type controls. In BMDM from CHRNA5 knockout mice, IL-1ß secretion was reduced by 50% compared to wild-type controls (p = 0.004). Therefore, a common haplotype of CHRNA5 that results in reduced cardiac expression of CHRNA5 and attenuated macrophage inflammasome activation is associated with lower mortality after AMI. These results implicate CHRNA5 and the cholinergic anti-inflammatory pathway in survival following AMI.


Asunto(s)
Infarto del Miocardio/genética , Miocarditis/genética , Proteínas del Tejido Nervioso/genética , Receptores Nicotínicos/genética , Anciano , Animales , Células Cultivadas , Femenino , Estudios de Asociación Genética , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Inflamasomas/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/metabolismo , Infarto del Miocardio/mortalidad , Miocarditis/diagnóstico , Miocarditis/metabolismo , Miocarditis/mortalidad , Fenotipo , Pronóstico , Estudios Prospectivos , Factores Protectores , Receptores Nicotínicos/deficiencia , Factores de Riesgo , Factores de Tiempo , Estados Unidos/epidemiología
16.
Proc Natl Acad Sci U S A ; 111(45): 16029-34, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25349429

RESUMEN

The mechanistic basis for why inflammation is simultaneously both deleterious and essential for tissue repair is not fully understood. Recently, a new paradigm has emerged: Organs are replete with resident macrophages of embryonic origin distinct from monocyte-derived macrophages. This added complexity raises the question of whether distinct immune cells drive inflammatory and reparative activities after injury. Previous work has demonstrated that the neonatal heart has a remarkable capacity for tissue repair compared with the adult heart, offering an ideal context to examine these concepts. We hypothesized that unrecognized differences in macrophage composition is a key determinant of cardiac tissue repair. Using a genetic model of cardiomyocyte ablation, we demonstrated that neonatal mice expand a population of embryonic-derived resident cardiac macrophages, which generate minimal inflammation and promote cardiac recovery through cardiomyocyte proliferation and angiogenesis. During homeostasis, the adult heart contains embryonic-derived macrophages with similar properties. However, after injury, these cells were replaced by monocyte-derived macrophages that are proinflammatory and lacked reparative activities. Inhibition of monocyte recruitment to the adult heart preserved embryonic-derived macrophage subsets, reduced inflammation, and enhanced tissue repair. These findings indicate that embryonic-derived macrophages are key mediators of cardiac recovery and suggest that therapeutics targeting distinct macrophage lineages may serve as novel treatments for heart failure.


Asunto(s)
Embrión de Mamíferos/metabolismo , Macrófagos/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Recuperación de la Función , Remodelación Ventricular , Animales , Inflamación/metabolismo , Inflamación/terapia , Ratones , Miocitos Cardíacos/trasplante , Regeneración
17.
J Biol Chem ; 289(13): 9158-71, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24532802

RESUMEN

Macrophage dysfunction and inflammasome activation have been implicated in the pathogenesis of diabetes and its complications. Prolonged inflammation and impaired healing are hallmarks of the diabetic response to tissue injury, and excessive inflammasome activation has been associated in these phenotypes. However, the mechanisms that regulate the inflammasome in response to lipid metabolic and inflammatory stress are incompletely understood. We have shown previously that IL-1ß secretion is induced in primary macrophages exposed to the dietary saturated fatty acid palmitate in combination with LPS. In this study, we sought to unravel the mechanisms underlying the activation of this lipotoxic inflammasome. We demonstrate that palmitate-loaded primary macrophages challenged with LPS activate the NLRP3 inflammasome through a mechanism that involves the lysosome. Interestingly, the lysosome was involved in both the regulation of pro-IL-1ß levels and its subsequent cleavage/release. The lysosomal protease cathepsin B was required for IL-1ß release but not pro-IL-1ß production. In contrast, disrupting lysosomal calcium regulation decreased IL-1ß release by reducing pro-IL-1ß levels. The calcium pathway involved the calcium-activated phosphatase calcineurin, which stabilized IL-1ß mRNA. Our findings provide evidence that the lysosome plays a key role in both the priming and assembly phases of the lipostoxic inflammasome. These findings have potential relevance to the hyperinflammatory phenotypes observed in diabetics during tissue damage or infection and identify lysosomes and calcineurin as potential therapeutic targets.


Asunto(s)
Inflamasomas/metabolismo , Lisosomas/metabolismo , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , Animales , Calcineurina/metabolismo , Inhibidores de la Calcineurina , Calcio/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular , Ácidos Grasos/farmacología , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Palmitatos/farmacología , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/química , ARN Mensajero/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
19.
J Immunol ; 190(3): 1285-96, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23275600

RESUMEN

Macrophage dysfunction in obesity and diabetes may predispose to the development of diabetic complications, such as infection and impaired healing after tissue damage. Saturated fatty acids, such as palmitate, are present at elevated concentrations in the plasma of patients with metabolic disease and may contribute to the pathogenesis of diabetes and its sequelae. To examine the effect of lipid excess on macrophage inflammatory function, we determined the influence of palmitate on LPS-mediated responses in peritoneal macrophages. Palmitate and LPS led to a profound synergistic cell death response in both primary and RAW 264.7 macrophages. The cell death had features of apoptosis and necrosis and was not dependent on endoplasmic reticulum stress, ceramide generation, or reactive oxygen species production. Instead, we uncovered a macrophage death pathway that required TLR4 signaling via TRIF but was independent of NF-κB, MAPKs, and IRF3. A significant decrease in macrophage lysosomal content was observed early in the death pathway, with evidence of lysosomal membrane damage occurring later in the death response. Overexpression of the transcription factor TFEB, which induces a lysosomal biogenic program, rescued the lysosomal phenotype and improved viability in palmitate- and LPS-treated cells. Our findings provide new evidence for cross-talk between lipid metabolism and the innate immune response that converges on the lysosome.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Lisosomas/inmunología , Macrófagos Peritoneales/efectos de los fármacos , Palmitatos/toxicidad , Receptor Toll-Like 4/fisiología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Muerte Celular/fisiología , Línea Celular Transformada/efectos de los fármacos , Línea Celular Transformada/metabolismo , Línea Celular Transformada/patología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Células Cultivadas/patología , Complicaciones de la Diabetes/metabolismo , Células HEK293 , Humanos , Inmunidad Innata , Membranas Intracelulares/patología , Metabolismo de los Lípidos/inmunología , Lipopolisacáridos/toxicidad , Lisosomas/patología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología , Transfección
20.
J Biol Chem ; 288(5): 2923-32, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23250746

RESUMEN

Macrophages play a key role in host defense and in tissue repair after injury. Emerging evidence suggests that macrophage dysfunction in states of lipid excess can contribute to the development of insulin resistance and may underlie inflammatory complications of diabetes. Ceramides are sphingolipids that modulate a variety of cellular responses including cell death, autophagy, insulin signaling, and inflammation. In this study we investigated the intersection between TLR4-mediated inflammatory signaling and saturated fatty acids with regard to ceramide generation. Primary macrophages treated with lipopolysaccharide (LPS) did not produce C16 ceramide, whereas palmitate exposure led to a modest increase in this sphingolipid. Strikingly, the combination of LPS and palmitate led to a synergistic increase in C16 ceramide. This response occurred via cross-talk at the level of de novo ceramide synthesis in the ER. The synergistic response required TLR4 signaling via MyD88 and TIR-domain-containing adaptor-inducing interferon beta (TRIF), whereas palmitate-induced ceramide production occurred independent of these inflammatory molecules. This ceramide response augmented IL-1ß and TNFα release, a process that may contribute to the enhanced inflammatory response in metabolic diseases characterized by dyslipidemia.


Asunto(s)
Ceramidas/biosíntesis , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Palmitatos/farmacología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Vías Biosintéticas/efectos de los fármacos , Extractos Celulares , Células Cultivadas , Interleucina-1beta/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Esfingolípidos/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA