Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 566(7744): 383-387, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30760925

RESUMEN

Sleep is integral to life1. Although insufficient or disrupted sleep increases the risk of multiple pathological conditions, including cardiovascular disease2, we know little about the cellular and molecular mechanisms by which sleep maintains cardiovascular health. Here we report that sleep regulates haematopoiesis and protects against atherosclerosis in mice. We show that mice subjected to sleep fragmentation produce more Ly-6Chigh monocytes, develop larger atherosclerotic lesions and produce less hypocretin-a stimulatory and wake-promoting neuropeptide-in the lateral hypothalamus. Hypocretin controls myelopoiesis by restricting the production of CSF1 by hypocretin-receptor-expressing pre-neutrophils in the bone marrow. Whereas hypocretin-null and haematopoietic hypocretin-receptor-null mice develop monocytosis and accelerated atherosclerosis, sleep-fragmented mice with either haematopoietic CSF1 deficiency or hypocretin supplementation have reduced numbers of circulating monocytes and smaller atherosclerotic lesions. Together, these results identify a neuro-immune axis that links sleep to haematopoiesis and atherosclerosis.


Asunto(s)
Aterosclerosis/prevención & control , Hematopoyesis/fisiología , Sueño/fisiología , Animales , Antígenos Ly/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células de la Médula Ósea/metabolismo , Femenino , Hematopoyesis/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Factor Estimulante de Colonias de Macrófagos/biosíntesis , Factor Estimulante de Colonias de Macrófagos/deficiencia , Factor Estimulante de Colonias de Macrófagos/metabolismo , Masculino , Ratones , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Mielopoyesis/efectos de los fármacos , Neutrófilos/metabolismo , Receptores de Orexina/deficiencia , Receptores de Orexina/metabolismo , Orexinas/biosíntesis , Orexinas/deficiencia , Orexinas/metabolismo , Orexinas/farmacología , Sueño/efectos de los fármacos , Privación de Sueño/metabolismo , Privación de Sueño/fisiopatología , Privación de Sueño/prevención & control
2.
Nature ; 566(7742): 115-119, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30700910

RESUMEN

The biochemical response to food intake must be precisely regulated. Because ingested sugars and fats can feed into many anabolic and catabolic pathways1, how our bodies handle nutrients depends on strategically positioned metabolic sensors that link the intrinsic nutritional value of a meal with intermediary metabolism. Here we describe a subset of immune cells-integrin ß7+ natural gut intraepithelial T lymphocytes (natural IELs)-that is dispersed throughout the enterocyte layer of the small intestine and that modulates systemic metabolism. Integrin ß7- mice that lack natural IELs are metabolically hyperactive and, when fed a high-fat and high-sugar diet, are resistant to obesity, hypercholesterolaemia, hypertension, diabetes and atherosclerosis. Furthermore, we show that protection from cardiovascular disease in the absence of natural IELs depends on the enteroendocrine-derived incretin GLP-12, which is normally controlled by IELs through expression of the GLP-1 receptor. In this metabolic control system, IELs modulate enteroendocrine activity by acting as gatekeepers that limit the bioavailability of GLP-1. Although the function of IELs may prove advantageous when food is scarce, present-day overabundance of diets high in fat and sugar renders this metabolic checkpoint detrimental to health.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Progresión de la Enfermedad , Intestino Delgado/citología , Linfocitos Intraepiteliales/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/prevención & control , Modelos Animales de Enfermedad , Ingestión de Alimentos , Enterocitos/citología , Enterocitos/metabolismo , Femenino , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Cadenas beta de Integrinas/genética , Cadenas beta de Integrinas/metabolismo , Masculino , Síndrome Metabólico/genética , Síndrome Metabólico/metabolismo , Síndrome Metabólico/prevención & control , Ratones
3.
Brain Behav Immun ; 54: 95-109, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26774527

RESUMEN

Traumatic brain injury (TBI) elicits immediate neuroinflammatory events that contribute to acute cognitive, motor, and affective disturbance. Despite resolution of these acute complications, significant neuropsychiatric and cognitive issues can develop and progress after TBI. We and others have provided novel evidence that these complications are potentiated by repeated injuries, immune challenges and stressors. A key component to this may be increased sensitization or priming of glia after TBI. Therefore, our objectives were to determine the degree to which cognitive deterioration occurred after diffuse TBI (moderate midline fluid percussion injury) and ascertain if glial reactivity induced by an acute immune challenge potentiated cognitive decline 30 days post injury (dpi). In post-recovery assessments, hippocampal-dependent learning and memory recall were normal 7 dpi, but anterograde learning was impaired by 30 dpi. Examination of mRNA and morphological profiles of glia 30 dpi indicated a low but persistent level of inflammation with elevated expression of GFAP and IL-1ß in astrocytes and MHCII and IL-1ß in microglia. Moreover, an acute immune challenge 30 dpi robustly interrupted memory consolidation specifically in TBI mice. These deficits were associated with exaggerated microglia-mediated inflammation with amplified (IL-1ß, CCL2, TNFα) and prolonged (TNFα) cytokine/chemokine expression, and a marked reactive morphological profile of microglia in the CA3 of the hippocampus. Collectively, these data indicate that microglia remain sensitized 30 dpi after moderate TBI and a secondary inflammatory challenge elicits robust microglial reactivity that augments cognitive decline. STATEMENT OF SIGNIFICANCE: Traumatic brain injury (TBI) is a major risk factor in development of neuropsychiatric problems long after injury, negatively affecting quality of life. Mounting evidence indicates that inflammatory processes worsen with time after a brain injury and are likely mediated by glia. Here, we show that primed microglia and astrocytes developed in mice 1 month following moderate diffuse TBI, coinciding with cognitive deficits that were not initially evident after injury. Additionally, TBI-induced glial priming may adversely affect the ability of glia to appropriately respond to immune challenges, which occur regularly across the lifespan. Indeed, we show that an acute immune challenge augmented microglial reactivity and cognitive deficits. This idea may provide new avenues of clinical assessments and treatments following TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/psicología , Mediadores de Inflamación/metabolismo , Microglía/patología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Traumatismos Difusos del Encéfalo/inmunología , Traumatismos Difusos del Encéfalo/metabolismo , Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/metabolismo , Quimiocinas/metabolismo , Cognición/fisiología , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Masculino , Memoria/fisiología , Ratones , Ratones Endogámicos BALB C , Microglía/metabolismo , Calidad de Vida
4.
J Neurosci ; 34(26): 8904-17, 2014 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-24966389

RESUMEN

Alternative activation of microglia/macrophages (M2a) by interleukin (IL)-4 is purported to support intrinsic growth and repair processes after CNS injury. Nonetheless, alternative activation of microglia is poorly understood in vivo, particularly in the context of inflammation, injury, and aging. Here, we show that aged mice (18-19 months) had reduced functional recovery after spinal cord injury (SCI) associated with impaired induction of IL-4 receptor α (IL-4Rα) on microglia. The failure to successfully promote an IL-4/IL-4Rα response in aged mice resulted in attenuated arginase (M2a associated), IL-1ß, and chemokine ligand 2 (CCL2) expression, and diminished recruitment of IL-4Rα(+) macrophages to the injured spinal cord. Furthermore, the link between reduced IL-4Rα expression and reduced arginase, IL-1ß, and CCL2 expression was confirmed using adult IL-4Rα knock-out (IL-4Rα(KO)) mice. To better understand IL-4Rα-mediated regulation of active microglia, a series of studies was completed in mice that were peripherally injected with lipopolysaccharide and later provided IL-4 by intracerebroventricular infusion. These immune-based studies demonstrate that inflammatory-induced IL-4Rα upregulation on microglia was required for the induction of arginase by IL-4. In addition, IL-4-mediated reprogramming of active microglia enhanced neurite growth ex vivo and increased inflammatory gene expression (i.e., IL-1ß and CCL2) and the corresponding recruitment of CCR2(+)/IL-4Rα(+)/arginase(+) myeloid cells in vivo. IL-4 reprogrammed active microglia to a unique and previously unreported phenotype (arginase(+)/IL-1ß(+)) that augmented neurite growth and enhanced recruitment of peripheral IL-4Rα(+) myeloid cells to the CNS. Moreover, this key signaling cascade was impaired with age corresponding with reduced functional recovery after SCI.


Asunto(s)
Envejecimiento/metabolismo , Interleucina-4/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Receptores de Interleucina-4/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Animales , Arginasa/metabolismo , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-4/farmacología , Activación de Macrófagos , Macrófagos/efectos de los fármacos , Ratones , Microglía/efectos de los fármacos , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
5.
Glia ; 62(6): 881-95, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24616125

RESUMEN

While there clearly is an intimate relationship between astrocytes and microglia, few studies have examined these potentially dynamic interactions. In this study, cytokine-mediated communication between microglia and astrocytes under inflammatory conditions was investigated. We have previously shown that activated microglia produce Interleukin (IL)-10, a regulatory cytokine that plays an important role in resolving neuroinflammation. Nonetheless, the mechanism by which IL-10 attenuates pro-inflammatory cytokine expression in the brain is unclear. Here, we show that IL-10 redirected astrocytes regulate the activation of microglia in a transforming growth factor (TGF)-ß dependent manner. In support of this concept, astrocytes in the brain maintained higher IL-10 receptor (IL-10R1) expression and primary astrocytes in culture were markedly more sensitive to the anti-inflammatory effects of IL-10 compared with microglia. Moreover, studies using primary cultures and an astrocyte-microglia coculture system revealed that astrocytes mediated the anti-inflammatory effects of IL-10 on microglia through the production of TGFß. For instance, only when astrocytes were present did IL-10 stimulation reduce the expression of IL-1ß and increase expression of anti-inflammatory mediators fractalkine receptor (CX3 CR1) and interleukin 4 receptor-α (IL-4Rα) in microglia. Importantly, these IL-10-astrocyte dependent effects on microglia were blocked by a TGFß inhibitor. Furthermore, inhibition of TGFß signaling in the brain resulted in prolonged sickness behavior and amplified pro-inflammatory cytokine expression in mice challenged with lipopolysaccharide. Taken together, IL-10 stimulated the production of TGFß by astrocytes, which in turn, attenuated microglial activation. Overall, these findings provide novel insight into the mechanisms by which astrocytes modulate microglia under inflammatory conditions.


Asunto(s)
Astrocitos/metabolismo , Interleucina-10/farmacología , Microglía/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Ratones , Ratones Endogámicos BALB C , Microglía/efectos de los fármacos
7.
Brain Behav Immun ; 26(5): 766-77, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22024136

RESUMEN

In several models of aging, microglia become more inflammatory and reactive to immune challenges. For example, peripheral LPS injection causes exaggerated microglial activation associated with prolonged sickness and depressive-like behavior in aged BALB/c mice. Therefore, the purpose of this study was to determine the extent to which age-related amplified microglial activation was associated with reduced sensitivity to the anti-inflammatory and M2 promoting cytokines interleukin (IL)-10 and IL-4. In initial studies with adult mice, LPS induced a time-dependent increase in M1 and M2 mRNA profiles in microglia. Furthermore, peripheral LPS injection markedly increased surface expression of IL-4 receptor-alpha (IL-4Rα), but not IL-10 receptor-1 (IL-10R1) on microglia. In BV-2 cells, IL-4, but not IL-10, re-directed LPS-activated microglia towards an M2 phenotype. Based on these findings, comparisons of M1 and M2 activation profiles, induction of IL-4Rα, and sensitivity to IL-4 were determined in microglia from adult (3-4 mo) and aged (18-22 mo) mice. In aged microglia, LPS promoted an exaggerated and prolonged M1 and M2 profile compared to adults. Moreover, IL-4Rα protein was not increased on aged microglia following LPS injection. To determine the consequence of impaired IL-4Rα upregulation, adult and aged mice were injected with LPS and activated microglia were then isolated and treated ex vivo with IL-4. While ex vivo IL-4 induced an M2 profile in activated microglia from adult mice, activated microglia from aged mice retained a prominent M1 profile. These data indicate that activated microglia from aged mice are less sensitive to the anti-inflammatory and M2-promoting effects of IL-4.


Asunto(s)
Envejecimiento/fisiología , Inflamación/metabolismo , Subunidad alfa del Receptor de Interleucina-4/biosíntesis , Interleucina-4/farmacología , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Microglía/metabolismo , Animales , Separación Celular , Células Cultivadas , Citometría de Flujo , Subunidad alfa del Receptor de Interleucina-10/biosíntesis , Masculino , Ratones , Ratones Endogámicos BALB C , Microglía/efectos de los fármacos , ARN/biosíntesis , ARN/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/efectos de los fármacos
8.
J Pineal Res ; 51(2): 180-6, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21486368

RESUMEN

Winter imposes physiological challenges on individuals including increased thermoregulatory demands, risk of infection, and decreased food availability. To survive these challenges, animals living outside the tropics must appropriately distribute their energetic costs across the year, including reproduction and immune function. Individuals of many species use the annual cycle of changing day lengths (photoperiod), which is encoded by the nightly duration of melatonin secretion, to adjust physiology. Siberian hamsters exposed to short days (SD) (long nights/prolonged endogenous melatonin secretion) enhance some aspects of immune function, but curtail other energetically expensive immune functions including the febrile response. The purpose of this study was twofold. First, we determined whether sustained melatonin treatment would inhibit the development of the SD phenotype in female hamsters as it does in males. Second, we examined whether the SD attenuation of fever would be blocked by continuous exposure to exogenous melatonin. Hamsters were implanted with melatonin or empty capsules, housed in either long days (LD) or SD for 8-9 weeks, and then challenged with lipopolysaccharide; body temperature and locomotor activity were recorded. Unlike hamsters with empty capsules, hamsters with melatonin implants did not respond to SD and maintained a LD phenotype including summer-like spleen, uterine and body masses, and pelage characteristics. Further, sustained melatonin treatment blocked the SD attenuation of febrile responses and prolonged the behavioral components of the sickness response. These results suggest that the daily fluctuations in endogenous melatonin may be masked by continuous exposure to exogenous melatonin, thus inhibiting functional photoperiodic responses to SD.


Asunto(s)
Antioxidantes/farmacología , Peso Corporal/efectos de los fármacos , Fiebre/metabolismo , Melatonina/farmacología , Actividad Motora/efectos de los fármacos , Fotoperiodo , Reproducción/efectos de los fármacos , Adaptación Fisiológica/efectos de los fármacos , Animales , Cricetinae , Femenino , Fiebre/inducido químicamente , Fiebre/fisiopatología , Lipopolisacáridos/toxicidad , Masculino , Phodopus
9.
J Exp Biol ; 213(Pt 12): 2031-7, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20511516

RESUMEN

Mammals that hibernate (hibernators) exhibit a circannual rhythm of food intake and body mass. In the laboratory during the winter hibernation period, many hibernators enter a series of multi-day torpor bouts, dropping their body temperature to near ambient, and cease to feed even if food is present in their cage. The mechanism(s) that regulates food intake in hibernators is unclear. Recently, AMP-activated protein kinase (AMPK) has been shown to play a key role in the central regulation of food intake in mammals. We hypothesized that infusing an AMPK activator, 5-aminoimidazole-4-carboxamide 1 B-D-ribofuranoside (AICAR), intracerebroventricularly (ICV) into the third ventricle of the hypothalamus would stimulate yellow-bellied marmots (Marmota flaviventris) to feed during their hibernation season. Infusion of AICAR ICV into marmots at an ambient temperature of 22 degrees C caused a significant (P<0.05) increase in food intake. In addition, animals stimulated to feed did not enter torpor during the infusion period. Marmots ICV infused with saline did not increase food intake and these animals continued to undergo torpor at an ambient temperature of 22 degrees C. Our results suggest that AICAR stimulated the food intake pathway, presumably by activating AMPK. These results support the hypothesis that AMPK may be involved in regulating food intake in hibernators and that there may be common neural pathways involved in regulating feeding and eliciting torpor.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Regulación del Apetito/efectos de los fármacos , Marmota/fisiología , Ribonucleótidos/farmacología , Aminoimidazol Carboxamida/farmacología , Animales , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/fisiología , Peso Corporal/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Femenino , Masculino , Cloruro de Sodio/farmacología
10.
J Exp Med ; 217(10)2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32716519

RESUMEN

The thymus is a primary lymphoid organ necessary for optimal T cell development. Here, we show that liver X receptors (LXRs)-a class of nuclear receptors and transcription factors with diverse functions in metabolism and immunity-critically contribute to thymic integrity and function. LXRαß-deficient mice develop a fatty, rapidly involuting thymus and acquire a shrunken and prematurely immunoinhibitory peripheral T cell repertoire. LXRαß's functions are cell specific, and the resulting phenotypes are mutually independent. Although thymic macrophages require LXRαß for cholesterol efflux, thymic epithelial cells (TECs) use LXRαß for self-renewal and thymocytes for negative selection. Consequently, TEC-derived LXRαß protects against homeostatic premature involution and orchestrates thymic regeneration following stress, while thymocyte-derived LXRαß limits cell disposal during negative selection and confers heightened sensitivity to experimental autoimmune encephalomyelitis. These results identify three distinct but complementary mechanisms by which LXRαß governs T lymphocyte education and illuminate LXRαß's indispensable roles in adaptive immunity.


Asunto(s)
Receptores X del Hígado/fisiología , Hígado/metabolismo , Linfocitos T/fisiología , Timo/fisiología , Inmunidad Adaptativa , Animales , Apoptosis , Femenino , Citometría de Flujo , Homeostasis , Humanos , Metabolismo de los Lípidos , Receptores X del Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T/metabolismo , Timo/metabolismo
11.
J Exp Med ; 216(2): 369-383, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30670465

RESUMEN

Acquisition of self-reactive effector CD4+ T cells is a major component of the autoimmune response that can occur during myocarditis, an inflammatory form of cardiomyopathy. Although the processes by which self-reactive T cells gain effector function have received considerable attention, how these T cells contribute to effector organ inflammation and damage is less clear. Here, we identified an IL-3-dependent amplification loop that exacerbates autoimmune inflammation. In experimental myocarditis, we show that effector organ-accumulating autoreactive IL-3+ CD4+ T cells stimulate IL-3R+ tissue macrophages to produce monocyte-attracting chemokines. The newly recruited monocytes differentiate into antigen-presenting cells that stimulate local IL-3+ CD4+ T cell proliferation, thereby amplifying organ inflammation. Consequently, Il3 -/- mice resist developing robust autoimmune inflammation and myocardial dysfunction, whereas therapeutic IL-3 targeting ameliorates disease. This study defines a mechanism that orchestrates inflammation in myocarditis, describes a previously unknown function for IL-3, and identifies IL-3 as a potential therapeutic target in patients with myocarditis.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Quimiotaxis/inmunología , Interleucina-3/inmunología , Monocitos/inmunología , Miocarditis/inmunología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Linfocitos T CD4-Positivos/patología , Proliferación Celular , Quimiotaxis/genética , Interleucina-3/genética , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Monocitos/patología , Miocarditis/genética , Miocarditis/patología
12.
J Exp Med ; 214(11): 3293-3310, 2017 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-28978634

RESUMEN

Myocardial infarction (MI) elicits massive inflammatory leukocyte recruitment to the heart. Here, we hypothesized that excessive leukocyte invasion leads to heart failure and death during acute myocardial ischemia. We found that shortly and transiently after onset of ischemia, human and mouse cardiac fibroblasts produce granulocyte/macrophage colony-stimulating factor (GM-CSF) that acts locally and distally to generate and recruit inflammatory and proteolytic cells. In the heart, fibroblast-derived GM-CSF alerts its neighboring myeloid cells to attract neutrophils and monocytes. The growth factor also reaches the bone marrow, where it stimulates a distinct myeloid-biased progenitor subset. Consequently, hearts of mice deficient in either GM-CSF or its receptor recruit fewer leukocytes and function relatively well, whereas mice producing GM-CSF can succumb from left ventricular rupture, a complication mitigated by anti-GM-CSF therapy. These results identify GM-CSF as both a key contributor to the pathogenesis of MI and a potential therapeutic target, bolstering the idea that GM-CSF is a major orchestrator of the leukocyte supply chain during inflammation.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inflamación/metabolismo , Leucocitos/metabolismo , Infarto del Miocardio/metabolismo , Animales , Médula Ósea/metabolismo , Quimiocinas/metabolismo , Subunidad beta Común de los Receptores de Citocinas/genética , Subunidad beta Común de los Receptores de Citocinas/metabolismo , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Monocitos/metabolismo , Células Mieloides/metabolismo , Neutrófilos/metabolismo , Análisis de Supervivencia
13.
Nat Commun ; 7: 12313, 2016 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-27460411

RESUMEN

Lipid accumulation in macrophages has profound effects on macrophage gene expression and contributes to the development of atherosclerosis. Here, we report that angiopoietin-like protein 4 (ANGPTL4) is the most highly upregulated gene in foamy macrophages and it's absence in haematopoietic cells results in larger atherosclerotic plaques, characterized by bigger necrotic core areas and increased macrophage apoptosis. Furthermore, hyperlipidemic mice deficient in haematopoietic ANGPTL4 have higher blood leukocyte counts, which is associated with an increase in the common myeloid progenitor (CMP) population. ANGPTL4-deficient CMPs have higher lipid raft content, are more proliferative and less apoptotic compared with the wild-type (WT) CMPs. Finally, we observe that ANGPTL4 deficiency in macrophages promotes foam cell formation by enhancing CD36 expression and reducing ABCA1 localization in the cell surface. Altogether, these findings demonstrate that haematopoietic ANGPTL4 deficiency increases atherogenesis through regulating myeloid progenitor cell expansion and differentiation, foam cell formation and vascular inflammation.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/deficiencia , Aterosclerosis/metabolismo , Aterosclerosis/patología , Progresión de la Enfermedad , Células Madre Hematopoyéticas/metabolismo , Monocitos/metabolismo , Proteína 4 Similar a la Angiopoyetina/metabolismo , Animales , Apoptosis , Aterosclerosis/complicaciones , Trasplante de Médula Ósea , Proliferación Celular , Supervivencia Celular , Células Espumosas/metabolismo , Humanos , Inflamación/complicaciones , Inflamación/patología , Leucocitosis/complicaciones , Leucocitosis/patología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Células Progenitoras Mieloides/metabolismo , Placa Aterosclerótica/complicaciones , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología
14.
Nat Med ; 22(8): 945-51, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27428900

RESUMEN

Iron is an essential component of the erythrocyte protein hemoglobin and is crucial to oxygen transport in vertebrates. In the steady state, erythrocyte production is in equilibrium with erythrocyte removal. In various pathophysiological conditions, however, erythrocyte life span is compromised severely, which threatens the organism with anemia and iron toxicity. Here we identify an on-demand mechanism that clears erythrocytes and recycles iron. We show that monocytes that express high levels of lymphocyte antigen 6 complex, locus C1 (LY6C1, also known as Ly-6C) ingest stressed and senescent erythrocytes, accumulate in the liver via coordinated chemotactic cues, and differentiate into ferroportin 1 (FPN1, encoded by SLC40A1)-expressing macrophages that can deliver iron to hepatocytes. Monocyte-derived FPN1(+)Tim-4(neg) macrophages are transient, reside alongside embryonically derived T cell immunoglobulin and mucin domain containing 4 (Timd4, also known as Tim-4)(high) Kupffer cells (KCs), and depend on the growth factor Csf1 and the transcription factor Nrf2 (encoded by Nfe2l2). The spleen, likewise, recruits iron-loaded Ly-6C(high) monocytes, but these do not differentiate into iron-recycling macrophages, owing to the suppressive action of Csf2. The accumulation of a transient macrophage population in the liver also occurs in mouse models of hemolytic anemia, anemia of inflammation, and sickle cell disease. Inhibition of monocyte recruitment to the liver during stressed erythrocyte delivery leads to kidney and liver damage. These observations identify the liver as the primary organ that supports rapid erythrocyte removal and iron recycling, and uncover a mechanism by which the body adapts to fluctuations in erythrocyte integrity.


Asunto(s)
Eritrocitos/metabolismo , Hepatocitos/metabolismo , Hierro/metabolismo , Macrófagos del Hígado/metabolismo , Hígado/metabolismo , Anemia , Anemia Hemolítica , Anemia de Células Falciformes , Animales , Antígenos Ly/metabolismo , Proteínas de Transporte de Catión/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Eritrocitos/citología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inflamación , Macrófagos del Hígado/citología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Monocitos/citología , Monocitos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Bazo
15.
J Neurotrauma ; 32(2): 127-38, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25070744

RESUMEN

Traumatic brain injury (TBI) is associated with cerebral edema, blood brain barrier breakdown, and neuroinflammation that contribute to the degree of injury severity and functional recovery. Unfortunately, there are no effective proactive treatments for limiting immediate or long-term consequences of TBI. Therefore, the objective of this study was to determine the efficacy of methylene blue (MB), an antioxidant agent, in reducing inflammation and behavioral complications associated with a diffuse brain injury. Here we show that immediate MB infusion (intravenous; 15-30 minutes after TBI) reduced cerebral edema, attenuated microglial activation and reduced neuroinflammation, and improved behavioral recovery after midline fluid percussion injury in mice. Specifically, TBI-associated edema and inflammatory gene expression in the hippocampus were significantly reduced by MB at 1 d post injury. Moreover, MB intervention attenuated TBI-induced inflammatory gene expression (interleukin [IL]-1ß, tumor necrosis factor α) in enriched microglia/macrophages 1 d post injury. Cell culture experiments with lipopolysaccharide-activated BV2 microglia confirmed that MB treatment directly reduced IL-1ß and increased IL-10 messenger ribonucleic acid in microglia. Last, functional recovery and depressive-like behavior were assessed up to one week after TBI. MB intervention did not prevent TBI-induced reductions in body weight or motor coordination 1-7 d post injury. Nonetheless, MB attenuated the development of acute depressive-like behavior at 7 d post injury. Taken together, immediate intervention with MB was effective in reducing neuroinflammation and improving behavioral recovery after diffuse brain injury. Thus, MB intervention may reduce life-threatening complications of TBI, including edema and neuroinflammation, and protect against the development of neuropsychiatric complications.


Asunto(s)
Conducta Animal/efectos de los fármacos , Edema Encefálico/tratamiento farmacológico , Lesiones Encefálicas/complicaciones , Depresión/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Azul de Metileno/uso terapéutico , Animales , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Edema Encefálico/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Depresión/metabolismo , Depresión/patología , Modelos Animales de Enfermedad , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Azul de Metileno/farmacología , Ratones , Microglía/patología , Factor de Necrosis Tumoral alfa/metabolismo
16.
Science ; 347(6227): 1260-5, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25766237

RESUMEN

Sepsis is a frequently fatal condition characterized by an uncontrolled and harmful host reaction to microbial infection. Despite the prevalence and severity of sepsis, we lack a fundamental grasp of its pathophysiology. Here we report that the cytokine interleukin-3 (IL-3) potentiates inflammation in sepsis. Using a mouse model of abdominal sepsis, we showed that innate response activator B cells produce IL-3, which induces myelopoiesis of Ly-6C(high) monocytes and neutrophils and fuels a cytokine storm. IL-3 deficiency protects mice against sepsis. In humans with sepsis, high plasma IL-3 levels are associated with high mortality even after adjusting for prognostic indicators. This study deepens our understanding of immune activation, identifies IL-3 as an orchestrator of emergency myelopoiesis, and reveals a new therapeutic target for treating sepsis.


Asunto(s)
Interleucina-3/inmunología , Sepsis/inmunología , Animales , Subgrupos de Linfocitos B/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inflamación , Interleucina-3/sangre , Interleucina-3/metabolismo , Lipopolisacáridos/inmunología , Tejido Linfoide/inmunología , Ratones , Ratones Endogámicos BALB C , Monocitos/inmunología , Mielopoyesis , Neutrófilos/inmunología , Peritonitis/inmunología , Peritonitis/patología , Pronóstico , Sepsis/mortalidad , Sepsis/patología , Sepsis/terapia
17.
Biol Psychiatry ; 76(7): 575-84, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24289885

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) is associated with a higher incidence of depression. The majority of individuals who suffer a TBI are juveniles and young adults, and thus, the risk of a lifetime of depressive complications is a significant concern. The etiology of increased TBI-associated depression is unclear but may be inflammatory-related with increased brain sensitivity to secondary inflammatory challenges (e.g., stressors, infection, and injury). METHODS: Adult male BALB/c mice received a sham (n = 52) or midline fluid percussion injury (TBI; n = 57). Neuroinflammation, motor coordination (rotarod), and depressive behaviors (social withdrawal, immobility in the tail suspension test, and anhedonia) were assessed 4 hours, 24 hours, 72 hours, 7 days, or 30 days later. Moreover, 30 days after surgery, sham and TBI mice received a peripheral injection of saline or lipopolysaccharide (LPS) and microglia activation and behavior were determined. RESULTS: Diffuse TBI caused inflammation, peripheral cell recruitment, and microglia activation immediately after injury coinciding with motor coordination deficits. These transient events resolved within 7 days. Nonetheless, 30 days post-TBI a population of deramified and major histocompatibility complex II(+) (primed) microglia were detected. After a peripheral LPS challenge, the inflammatory cytokine response in primed microglia of TBI mice was exaggerated compared with microglia of controls. Furthermore, this LPS-induced microglia reactivity 30 days after TBI was associated with the onset of depressive-like behavior. CONCLUSIONS: These results implicate a primed and immune-reactive microglial population as a possible triggering mechanism for the development of depressive complications after TBI.


Asunto(s)
Lesiones Encefálicas/complicaciones , Depresión/etiología , Depresión/inmunología , Encefalitis/inmunología , Microglía/metabolismo , Animales , Encefalitis/etiología , Gliosis , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos BALB C , Actividad Motora , Factores de Tiempo
18.
J Neuroimmune Pharmacol ; 8(5): 1098-105, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23709339

RESUMEN

Impaired memory may result from synaptic glutamatergic dysregulation related to chronic neuroinflammation. GLT1 is the primary excitatory amino acid transporter responsible for regulating extracellular glutamate levels in the hippocampus. We tested the hypothesis that if impaired spatial memory results from increased extracellular glutamate due to age or experimentally induced chronic neuroinflammation in the hippocampus, then pharmacological augmentation of the glutamate transporter GLT1 will attenuate deficits in a hippocampal-dependent spatial memory task. The profile of inflammation-related genes and proteins associated with normal aging, or chronic neuroinflammation experimentally-induced via a four-week LPS infusion into the IV(th) ventricle, were correlated with performance in the Morris water maze following treatment with Riluzole, a drug that can enhance glutamate clearance by increasing GLT1 expression. Age-associated inflammation was qualitatively different from LPS-induced neuro-inflammation in young rats. LPS produced a pro-inflammatory phenotype characterized by increased IL-1ß expression in the hippocampus, whereas aging was not associated with a strong central pro-inflammatory response but with a mixed peripheral immune phenotype. Riluzole attenuated the spatial memory impairment, the elevation of serum cytokines and the decrease in GLT1 gene expression in Aged rats, but had no effect on young rats infused with LPS. Our findings highlight the therapeutic potential of reducing glutamatergic function upon memory impairment in neurodegenerative diseases associated with aging.


Asunto(s)
Envejecimiento/metabolismo , Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Hipocampo/efectos de los fármacos , Trastornos de la Memoria/metabolismo , Fármacos Neuroprotectores/farmacología , Riluzol/farmacología , Animales , Hipocampo/metabolismo , Lipopolisacáridos/toxicidad , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Ratas , Ratas Endogámicas F344 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Neurobiol Aging ; 34(12): 2748-58, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23880139

RESUMEN

Microglia develop an inflammatory phenotype during normal aging. The mechanism by which this occurs is not well understood, but might be related to impairments in several key immunoregulatory systems. Here we show that micro-RNA (miR)-29a and miR-29b, 2 immunoregulatory micro-RNAs, were increased in the brain of aged BALB/c mice compared with adults. Insulin-like growth factor-1 (IGF-1) and fractalkine ligand (CX3CL1) are negative modulators of microglial activation and were identified as targets of miR-29a and miR-29b using luciferase assay and primary microglia transfection. Indeed, higher expression of miR-29b in the brain of aged mice was associated with reduced messenger RNA (mRNA) levels of IGF-1 and CX3CL1. Parallel to these results in mice, miR-29a and miR-29b were also markedly increased in cortical brain tissue of older individuals (mean, 77 years) compared with middle-aged adults (mean, 45 years). Moreover, increased expression of miR-29b in human cortical tissue was negatively correlated with IGF-1 and CX3CL1 expression. Collectively, these data indicate that an age-associated increase in miR-29 corresponded with the reduction of 2 important regulators of microglia, IGF-1 and CX3CL1.


Asunto(s)
Envejecimiento/genética , Corteza Cerebral/metabolismo , Quimiocina CX3CL1/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , MicroARNs/metabolismo , Microglía/metabolismo , Receptores de Quimiocina/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/metabolismo , Animales , Receptor 1 de Quimiocinas CX3C , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Ligandos , Masculino , Ratones Endogámicos BALB C , Persona de Mediana Edad , ARN Mensajero/metabolismo , Receptores de Quimiocina/genética , Adulto Joven
20.
J Neuroimmune Pharmacol ; 7(1): 7-23, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21932047

RESUMEN

A hallmark of the aged immune system is impaired immunoregulation of the innate and adaptive immune system in the periphery and also in the central nervous system (CNS). Impaired immunoregulation may predispose older individuals to an increased frequency of peripheral infections with concomitant cognitive and behavioral complications. Thus, normal aging is hypothesized to alter the highly coordinated interactions between the immune system and the brain. In support of this notion, mounting evidence in rodent models indicate that the increased inflammatory status of the brain is associated with increased reactivity of microglia, the innate immune cells of the CNS. Understanding how immunity is affected with age is important because CNS immune cells play an integral role in propagating inflammatory signals that are initiated in the periphery. Increased reactivity of microglia sets the stage for an exaggerated inflammatory cytokine response following activation of the peripheral innate immune system that is paralleled by prolonged sickness, depressive-like complications and cognitive impairment. Moreover, amplified neuroinflammation negatively affects several aspects of neural plasticity (e.g., neurogenesis, long-term potentiation, and dendritic morphology) that can contribute to the severity of neurological complications. The purpose of this review is to discuss several key peripheral and central immune changes that impair the coordinated response between the immune system and the brain and result in behavioral and cognitive deficits.


Asunto(s)
Envejecimiento/inmunología , Encéfalo/inmunología , Trastornos del Conocimiento/inmunología , Trastornos Mentales/inmunología , Neuroinmunomodulación/fisiología , Envejecimiento/patología , Animales , Encéfalo/patología , Trastornos del Conocimiento/metabolismo , Humanos , Trastornos Mentales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA