Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 21(6): 696, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32303726

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Nat Immunol ; 21(6): 615-625, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32251403

RESUMEN

Increasing age alters innate immune-mediated responses; however, the mechanisms underpinning these changes in humans are not fully understood. Using a human dermal model of acute inflammation, we found that, although inflammatory onset is similar between young and elderly individuals, the resolution phase was substantially impaired in elderly individuals. This arose from a reduction in T cell immunoglobulin mucin receptor-4 (TIM-4), a phosphatidylserine receptor expressed on macrophages that enables the engulfment of apoptotic bodies, so-called efferocytosis. Reduced TIM-4 in elderly individuals was caused by an elevation in macrophage p38 mitogen-activated protein kinase (MAPK) activity. Administering an orally active p38 inhibitor to elderly individuals rescued TIM-4 expression, cleared apoptotic bodies and restored a macrophage resolution phenotype. Thus, inhibiting p38 in elderly individuals rejuvenated their resolution response to be more similar to that of younger people. This is the first resolution defect identified in humans that has been successfully reversed, thereby highlighting the tractability of targeting pro-resolution biology to treat diseases driven by chronic inflammation.


Asunto(s)
Inflamación/etiología , Inflamación/metabolismo , Fagocitosis/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Factores de Edad , Anciano , Animales , Apoptosis , Vesícula/inmunología , Vesícula/metabolismo , Vesícula/patología , Cantaridina , Expresión Génica , Humanos , Inmunidad Innata , Inflamación/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/patología , Receptores de Superficie Celular/metabolismo , Transducción de Señal
3.
Arterioscler Thromb Vasc Biol ; 42(8): 1060-1076, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35510553

RESUMEN

BACKGROUND: Aging enhances most chronic diseases but its impact on human aortic tissue in health and in thoracic aortic aneurysms (TAA) remains unclear. METHODS: We employed a human aortic biorepository of healthy specimens (n=17) and those that underwent surgical repair for TAA (n=20). First, we performed proteomics comparing aortas of healthy donors to aneurysmal specimens, in young (ie, <60 years of age) and old (ie, ≥60 years of age) subjects. Second, we measured proteins, via immunoblotting, involved in mitophagy (ie, Parkin) and also mitochondrial-induced inflammatory pathways, specifically TLR (toll-like receptor) 9, STING (stimulator of interferon genes), and IFN (interferon)-ß. RESULTS: Proteomics revealed that aging transformed the aorta both quantitatively and qualitatively from health to TAA. Whereas young aortas exhibited an enrichment of immunologic processes, older aortas exhibited an enrichment of metabolic processes. Immunoblotting revealed that the expression of Parkin directly correlated to subject age in health but inversely to subject age in TAA. In TAA, but not in health, phosphorylation of STING and the expression of IFN-ß was impacted by aging regardless of whether subjects had bicuspid or tricuspid valves. In subjects with bicuspid valves and TAAs, TLR9 expression positively correlated with subject age. Interestingly, whereas phosphorylation of STING was inversely correlated with subject age, IFN-ß positively correlated with subject age. CONCLUSIONS: Aging transforms the human aortic proteome from health to TAA, leading to a differential regulation of biological processes. Our results suggest that the development of therapies to mitigate vascular diseases including TAA may need to be modified depending on subject age.


Asunto(s)
Aneurisma de la Aorta Torácica , Envejecimiento , Aorta/metabolismo , Aneurisma de la Aorta Torácica/genética , Aneurisma de la Aorta Torácica/metabolismo , Humanos , Interferones , Proteoma , Ubiquitina-Proteína Ligasas
4.
Circ Res ; 126(3): 298-314, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31818196

RESUMEN

Rationale: Aging is one of the strongest risk factors for atherosclerosis. Yet whether aging increases the risk of atherosclerosis independently of chronic hyperlipidemia is not known. Objective: To determine if vascular aging before the induction of hyperlipidemia enhances atherogenesis. Methods and Results: We analyzed the aortas of young and aged normolipidemic wild type, disease-free mice and found that aging led to elevated IL (interleukin)-6 levels and mitochondrial dysfunction, associated with increased mitophagy and the associated protein Parkin. In aortic tissue culture, we found evidence that with aging mitochondrial dysfunction and IL-6 exist in a positive feedback loop. We triggered acute hyperlipidemia in aged and young mice by inducing liver-specific degradation of the LDL (low-density lipoprotein) receptor combined with a 10-week western diet and found that atherogenesis was enhanced in aged wild-type mice. Hyperlipidemia further reduced mitochondrial function and increased the levels of Parkin in the aortas of aged mice but not young mice. Genetic disruption of autophagy in smooth muscle cells of young mice exposed to hyperlipidemia led to increased aortic Parkin and IL-6 levels, impaired mitochondrial function, and enhanced atherogenesis. Importantly, enhancing mitophagy in aged, hyperlipidemic mice via oral administration of spermidine prevented the increase in aortic IL-6 and Parkin, attenuated mitochondrial dysfunction, and reduced atherogenesis. Conclusions: Before hyperlipidemia, aging elevates IL-6 and impairs mitochondrial function within the aorta, associated with enhanced mitophagy and increased Parkin levels. These age-associated changes prime the vasculature to exacerbate atherogenesis upon acute hyperlipidemia. Our work implies that novel therapeutics aimed at improving vascular mitochondrial bioenergetics or reducing inflammation before hyperlipidemia may reduce age-related atherosclerosis.


Asunto(s)
Envejecimiento/metabolismo , Aterosclerosis/metabolismo , Endotelio Vascular/metabolismo , Lipoproteínas LDL/metabolismo , Mitocondrias/metabolismo , Envejecimiento/patología , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Aterosclerosis/etiología , Aterosclerosis/prevención & control , Dieta Alta en Grasa/efectos adversos , Endotelio Vascular/patología , Retroalimentación Fisiológica , Femenino , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/patología , Mitofagia , Receptores de LDL/metabolismo , Espermidina/farmacología , Espermidina/uso terapéutico , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
5.
J Immunol ; 205(2): 313-320, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32493812

RESUMEN

Aging impairs immunity to promote diseases, especially respiratory viral infections. The current COVID-19 pandemic, resulting from SARS-CoV-2, induces acute pneumonia, a phenotype that is alarmingly increased with aging. In this article, we review findings of how aging alters immunity to respiratory viral infections to identify age-impacted pathways common to several viral pathogens, permitting us to speculate about potential mechanisms of age-enhanced mortality to COVID-19. Aging generally leads to exaggerated innate immunity, particularly in the form of elevated neutrophil accumulation across murine and large animal studies of influenza infection. COVID-19 patients who succumb exhibit a 2-fold increase in neutrophilia, suggesting that exaggerated innate immunity contributes to age-enhanced mortality to SARS-CoV-2 infection. Further investigation in relevant experimental models will elucidate the mechanisms by which aging impacts respiratory viral infections, including SARS-CoV-2. Such investigation could identify therapies to reduce the suffering of the population at large, but especially among older people, infected with respiratory viruses.


Asunto(s)
Envejecimiento/patología , Betacoronavirus/fisiología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Neumonía Viral/inmunología , Neumonía Viral/patología , Infecciones del Sistema Respiratorio/virología , COVID-19 , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/virología , Citocinas/inmunología , Humanos , Gripe Humana/inmunología , Gripe Humana/patología , Pandemias , Infecciones del Sistema Respiratorio/patología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , SARS-CoV-2
6.
Am J Physiol Heart Circ Physiol ; 320(1): H323-H337, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33164548

RESUMEN

Interleukin-4 receptor α (IL4Rα) signaling plays an important role in cardiac remodeling during myocardial infarction (MI). However, the target cell type(s) of IL4Rα signaling during this remodeling remains unclear. Here, we investigated the contribution of endogenous myeloid-specific IL4Rα signaling in cardiac remodeling post-MI. We established a murine myeloid-specific IL4Rα knockout (MyIL4RαKO) model with LysM promoter-driven Cre recombination. Macrophages from MyIL4RαKO mice showed significant downregulation of alternatively activated macrophage markers but an upregulation of classical activated macrophage markers both in vitro and in vivo, indicating the successful inactivation of IL4Rα signaling in macrophages. To examine the role of myeloid IL4Rα during MI, we subjected MyIL4RαKO and littermate floxed control (FC) mice to MI. We found that cardiac function was significantly impaired as a result of myeloid-specific IL4Rα deficiency. This deficiency resulted in a dysregulated inflammatory response consisting of decreased production of anti-inflammatory cytokines. Myeloid IL4Rα deficiency also led to reduced collagen 1 deposition and an imbalance of matrix metalloproteinases (MMPs)/tissue inhibitors of metalloproteinases (TIMPs), with upregulated MMPs and downregulated TIMPs, which resulted in insufficient fibrotic remodeling. In conclusion, this study identifies that myeloid-specific IL4Rα signaling regulates inflammation and fibrotic remodeling during MI. Therefore, myeloid-specific activation of IL4Rα signaling could offer protective benefits after MI.NEW & NOTEWORTHY This study showed, for the first time, the role of endogenous IL4Rα signaling in myeloid cells during cardiac remodeling and the underlying mechanisms. We identified myeloid cells are the critical target cell types of IL4Rα signaling during cardiac remodeling post-MI. Deficiency of myeloid IL4Rα signaling causes deteriorated cardiac function post-MI, due to dysregulated inflammation and insufficient fibrotic remodeling. This study sheds light on the potential of activating myeloid-specific IL4Rα signaling to modify remodeling post-MI. This brings hope to patients with MI and diminishes side effects by cell type-specific instead of whole body treatment.


Asunto(s)
Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Receptores de Superficie Celular/metabolismo , Función Ventricular Izquierda , Remodelación Ventricular , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Activación de Macrófagos , Macrófagos/patología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Transducción de Señal
7.
Arterioscler Thromb Vasc Biol ; 40(1): 61-71, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31619062

RESUMEN

OBJECTIVE: CD73 is an ectonucleotidase which catalyzes the conversion of AMP (adenosine monophosphate) to adenosine. Adenosine has been shown to be anti-inflammatory and vasorelaxant. The impact of ectonucleotidases on age-dependent atherosclerosis remains unclear. Our aim was to investigate the role of CD73 in age-dependent accumulation of atherosclerosis. Approach and results: Mice doubly deficient in CD73 and ApoE (apolipoprotein E; (cd73-/-/apoE-/-) were generated, and the extent of aortic atherosclerotic plaque was compared with apoE-/- controls at 12, 20, 32, and 52 weeks. By 12 weeks of age, cd73-/-/apoE-/- mice exhibited a significant increase in plaque (1.4±0.5% of the total vessel surface versus 0.4±0.1% in apoE-/- controls, P<0.005). By 20 weeks of age, this difference disappeared (2.9±0.4% versus 3.3±0.7%). A significant reversal in phenotype emerged at 32 weeks (9.8±1.2% versus 18.3±1.4%; P<0.0001) and persisted at the 52 week timepoint (22.4±2.1% versus 37.0±2.1%; P<0.0001). The inflammatory response to aging was found to be comparable between cd73-/-/apoE-/- mice and apoE-/- controls. A reduction in lipolysis in CD73 competent mice was observed, even with similar plasma lipid levels (cd73-/-/apoE-/- versus apoE-/- at 12 weeks [16.2±0.7 versus 9.5±1.4 nmol glycerol/well], 32 weeks [24.1±1.5 versus 7.4±0.4 nmol/well], and 52 weeks [13.8±0.62 versus 12.7±2.0 nmol/well], P<0.001). CONCLUSIONS: At early time points, CD73 exerts a subtle antiatherosclerotic influence, but with age, the pattern reverses, and the presence of CD73 promoted suppression of lipid catabolism.


Asunto(s)
5'-Nucleotidasa/genética , Aterosclerosis/genética , Regulación del Desarrollo de la Expresión Génica , ARN/genética , 5'-Nucleotidasa/biosíntesis , Animales , Aorta Torácica/metabolismo , Aorta Torácica/patología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo
8.
Curr Opin Lipidol ; 31(5): 265-272, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32773466

RESUMEN

PURPOSE OF REVIEW: The incidence of arterial calcification increases with age, can occur independently of atherosclerosis and hyperlipidemia, contributes to vessel stiffening, and is associated with adverse cardiovascular outcomes. Here, we provide an up-to-date review of how aging leads to arterial calcification and discuss potential therapies. RECENT FINDINGS: Recent research suggests that mitochondrial dysfunction (impaired efficiency of the respiratory chain, increased reactive oxygen species production, and a high mutation rate of mitochondrial DNA), cellular senescence, ectonucleotidases, and extrinsic factors such as hyperglycemia promote age-determined calcification. We discuss the future potential impact of antilipidemics, senolytics, and poly(ADP-ribose)polymerases inhibitors on age-associated arterial calcification. SUMMARY: Understanding how mechanisms of aging lead to arterial calcification will allow us to pinpoint prospective strategies to mitigate arterial calcification, even after the effects of aging have already begun to occur.


Asunto(s)
Envejecimiento/metabolismo , Arterias/metabolismo , Calcificación Vascular , Envejecimiento/patología , Animales , Humanos , Mitocondrias/patología , Calcificación Vascular/metabolismo , Calcificación Vascular/patología , Calcificación Vascular/fisiopatología
9.
Eur Respir J ; 56(6)2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33033152

RESUMEN

Cellular senescence permanently arrests the replication of various cell types and contributes to age-associated diseases. In particular, cellular senescence may enhance chronic lung diseases including COPD and idiopathic pulmonary fibrosis. However, the role cellular senescence plays in the pathophysiology of acute inflammatory diseases, especially viral infections, is less well understood. There is evidence that cellular senescence prevents viral replication by increasing antiviral cytokines, but other evidence shows that senescence may enhance viral replication by downregulating antiviral signalling. Furthermore, cellular senescence leads to the secretion of inflammatory mediators, which may either promote host defence or exacerbate immune pathology during viral infections. In this Perspective article, we summarise how senescence contributes to physiology and disease, the role of senescence in chronic lung diseases, and how senescence impacts acute respiratory viral infections. Finally, we develop a potential framework for how senescence may contribute, both positively and negatively, to the pathophysiology of viral respiratory infections, including severe acute respiratory syndrome due to the coronavirus SARS-CoV-2.


Asunto(s)
Senescencia Celular , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/virología , Virosis/patología , Virosis/virología , Humanos , Pulmón/patología
10.
J Immunol ; 199(3): 1060-1068, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28646038

RESUMEN

Influenza viral infections often lead to increased mortality in older people. However, the mechanisms by which aging impacts immunity to influenza lung infection remain unclear. We employed a murine model of influenza infection to identify these mechanisms. With aging, we found reduced numbers of alveolar macrophages, cells essential for lung homeostasis. We also determined that these macrophages are critical for influenza-induced mortality with aging. Furthermore, aging vastly alters the transcriptional profile and specifically downregulates cell cycling pathways in alveolar macrophages. Aging impairs the ability of alveolar macrophages to limit lung damage during influenza infection. Moreover, aging decreases alveolar macrophage phagocytosis of apoptotic neutrophils, downregulates the scavenging receptor CD204, and induces retention of neutrophils during influenza infection. Thus, aging induces defective phagocytosis by alveolar macrophages and increases lung damage. These findings indicate that therapies that enhance the function of alveolar macrophages may improve outcomes in older people infected with respiratory viruses.


Asunto(s)
Envejecimiento , Gripe Humana/mortalidad , Macrófagos Alveolares/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/mortalidad , Fagocitosis , Animales , Ciclo Celular , Modelos Animales de Enfermedad , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Gripe Humana/virología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Macrófagos Alveolares/metabolismo , Ratones , Neutrófilos/inmunología , Neutrófilos/patología , Neutrófilos/virología , Infecciones por Orthomyxoviridae/virología , Receptores Depuradores de Clase A/genética , Receptores Depuradores de Clase A/metabolismo
11.
Am J Respir Crit Care Med ; 198(2): 256-263, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29546996

RESUMEN

Pneumonia is a complex pulmonary disease in need of new clinical approaches. Although triggered by a pathogen, pneumonia often results from dysregulations of host defense that likely precede infection. The coordinated activities of immune resistance and tissue resilience then dictate whether and how pneumonia progresses or resolves. Inadequate or inappropriate host responses lead to more severe outcomes such as acute respiratory distress syndrome and to organ dysfunction beyond the lungs and over extended time frames after pathogen clearance, some of which increase the risk for subsequent pneumonia. Improved understanding of such host responses will guide the development of novel approaches for preventing and curing pneumonia and for mitigating the subsequent pulmonary and extrapulmonary complications of pneumonia. The NHLBI assembled a working group of extramural investigators to prioritize avenues of host-directed pneumonia research that should yield novel approaches for interrupting the cycle of unhealthy decline caused by pneumonia. This report summarizes the working group's specific recommendations in the areas of pneumonia susceptibility, host response, and consequences. Overarching goals include the development of more host-focused clinical approaches for preventing and treating pneumonia, the generation of predictive tools (for pneumonia occurrence, severity, and outcome), and the elucidation of mechanisms mediating immune resistance and tissue resilience in the lung. Specific areas of research are highlighted as especially promising for making advances against pneumonia.


Asunto(s)
Susceptibilidad a Enfermedades/fisiopatología , Interacciones Microbiota-Huesped/fisiología , Pulmón/fisiopatología , Neumonía/fisiopatología , Informe de Investigación , Síndrome de Dificultad Respiratoria/fisiopatología , Adulto , Anciano , Anciano de 80 o más Años , Infecciones Bacterianas/fisiopatología , Congresos como Asunto , Femenino , Humanos , Masculino , Persona de Mediana Edad , National Heart, Lung, and Blood Institute (U.S.) , Estados Unidos , Virosis/fisiopatología
12.
Immunol Rev ; 258(1): 132-44, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24517430

RESUMEN

Solid organ transplantation is a vital therapy for end stage diseases. Decades of research have established that components of the adaptive immune system are critical for transplant rejection, but the role of the innate immune system in organ transplantation is just emerging. Accumulating evidence indicates that the innate immune system is activated at the time of organ implantation by the release of endogenous inflammatory triggers. This review discusses the nature of these triggers in organ transplantation and also potential mediators that may enhance inflammation resolution after organ implantation.


Asunto(s)
Rechazo de Injerto/inmunología , Mediadores de Inflamación/inmunología , Inflamación/inmunología , Trasplante de Órganos/efectos adversos , Transducción de Señal , Aloinjertos , Animales , Rechazo de Injerto/metabolismo , Humanos , Inmunidad Innata , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Resultado del Tratamiento
13.
Eur J Immunol ; 46(11): 2650-2658, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27546296

RESUMEN

Organ transplantation in older people is increasing, but how aging impacts B-cell responses to organ transplantation is still unknown. Here, we show that the depletion of B cells with anti-CD20 antibodies has disparate effects depending on recipient age. In young murine recipients, anti-CD20 treatment impaired the ability of immune modulation to extend skin allograft survival. In contrast, anti-CD20 treatment extended allograft survival in aged recipients treated with immune modulation. Although regulatory B-cell function and the numbers of marginal and follicular B cells were similar between age groups, a subpopulation of B cells, termed age-associated B cells (ABCs), accumulated upon aging. ABCs isolated from aged mice exhibited upregulation of CD73, CD80, CD106, and TLR2 and an increased capacity to augment T-cell alloimmunity compared to ABCs from young mice. Importantly, ABCs from aged, but not young, mice impaired the ability of immune modulation to enhance allograft survival after adoptive transfer into young transplant recipients. Our study indicates that ABCs impair the immune regulation of allografts. Thus, recipient age needs to be considered when proposing B-cell-depleting immune therapy.


Asunto(s)
Envejecimiento/inmunología , Aloinjertos/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , 5'-Nucleotidasa/genética , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígeno B7-1/genética , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante de Piel , Receptor Toll-Like 2/genética , Trasplante Homólogo , Molécula 1 de Adhesión Celular Vascular/genética
14.
Circ Res ; 116(10): 1670-9, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25801896

RESUMEN

RATIONALE: Early graft inflammation enhances both acute and chronic rejection of heart transplants, but it is unclear how this inflammation is initiated. OBJECTIVE: To identify specific inflammatory modulators and determine their underlying molecular mechanisms after cardiac transplantation. METHODS AND RESULTS: We used a murine heterotopic cardiac transplant model to identify inflammatory modulators of early graft inflammation. Unbiased mass spectrometric analysis of cardiac tissue before and ≤72 hours after transplantation revealed that 22 proteins including haptoglobin, a known antioxidant, are significantly upregulated in our grafts. Through the use of haptoglobin-deficient mice, we show that 80% of haptoglobin-deficient recipients treated with perioperative administration of the costimulatory blocking agent CTLA4 immunoglobulin exhibited >100-day survival of full major histocompatibility complex mismatched allografts, whereas all similarly treated wild-type recipients rejected their transplants by 21 days after transplantation. We found that haptoglobin modifies the intra-allograft inflammatory milieu by enhancing levels of the inflammatory cytokine interleukin-6 and the chemokine MIP-2 (macrophage inflammatory protein 2) but impair levels of the immunosuppressive cytokine interleukin-10. Haptoglobin also enhances dendritic cell graft recruitment and augments antidonor T-cell responses. Moreover, we confirmed that the protein is present in human cardiac allograft specimens undergoing acute graft rejection. CONCLUSIONS: Our findings provide new insights into the mechanisms of inflammation after cardiac transplantation and suggest that, in contrast to its prior reported antioxidant function in vascular inflammation, haptoglobin is an enhancer of inflammation after cardiac transplantation. Haptoglobin may also be a key component in other sterile inflammatory conditions.


Asunto(s)
Rechazo de Injerto/inmunología , Haptoglobinas/inmunología , Trasplante de Corazón/efectos adversos , Mediadores de Inflamación/inmunología , Inflamación/inmunología , Miocardio/inmunología , Abatacept , Animales , Proliferación Celular , Células Cultivadas , Quimiocina CXCL2/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Rechazo de Injerto/sangre , Rechazo de Injerto/genética , Rechazo de Injerto/patología , Rechazo de Injerto/prevención & control , Haptoglobinas/metabolismo , Humanos , Inmunoconjugados/farmacología , Inmunosupresores/farmacología , Inflamación/sangre , Inflamación/patología , Mediadores de Inflamación/sangre , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Activación de Linfocitos , Masculino , Espectrometría de Masas , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Miocardio/metabolismo , Miocardio/patología , Proteómica/métodos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo
15.
J Immunol ; 194(8): 4039-48, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25762783

RESUMEN

Neutrophils are critical mediators of innate immune responses and contribute to tissue injury. However, immune pathways that regulate neutrophil recruitment to injured tissues during noninfectious inflammation remain poorly understood. DAP12 is a cell membrane-associated protein that is expressed in myeloid cells and can either augment or dampen innate inflammatory responses during infections. To elucidate the role of DAP12 in pulmonary ischemia/reperfusion injury (IRI), we took advantage of a clinically relevant mouse model of transplant-mediated lung IRI. This technique allowed us to dissect the importance of DAP12 in tissue-resident cells and those that infiltrate injured tissue from the periphery during noninfectious inflammation. Macrophages in both mouse and human lungs that have been subjected to cold ischemic storage express DAP12. We found that donor, but not recipient, deficiency in DAP12 protected against pulmonary IRI. Analysis of the immune response showed that DAP12 promotes the survival of tissue-resident alveolar macrophages and contributes to local production of neutrophil chemoattractants. Intravital imaging demonstrated a transendothelial migration defect into DAP12-deficient lungs, which can be rescued by local administration of the neutrophil chemokine CXCL2. We have uncovered a previously unrecognized role for DAP12 expression in tissue-resident alveolar macrophages in mediating acute noninfectious tissue injury through regulation of neutrophil trafficking.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Regulación de la Expresión Génica/inmunología , Trasplante de Pulmón , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Disfunción Primaria del Injerto/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Quimiocina CXCL2/genética , Quimiocina CXCL2/inmunología , Humanos , Pulmón/patología , Macrófagos Alveolares/patología , Ratones , Ratones Noqueados , Infiltración Neutrófila/genética , Neutrófilos/patología , Disfunción Primaria del Injerto/genética , Disfunción Primaria del Injerto/patología
16.
Curr Opin Organ Transplant ; 21(3): 253-7, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27077602

RESUMEN

PURPOSE OF REVIEW: Allograft vasculopathy is the leading cause of late allograft loss following solid organ transplantation. Ischemia reperfusion injury and donor-specific antibody-induced complement activation confer heightened risk for allograft vasculopathy via numerous innate immune mechanisms, including MyD88, high-mobility group box 1 (HMGB1), and complement-induced noncanonical nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling. RECENT FINDINGS: The role of MyD88, a signal adaptor downstream of the Toll-like receptors (TLR), has been defined in an experimental heart transplant model, which demonstrated that recipient MyD88 enhanced allograft vasculopathy. Importantly, triggering receptor on myeloid receptor 1, a MyD88 amplifying signal, was present in rejecting human cardiac transplant biopsies and enhanced the development of allograft vasculopathy in mice. HMGB1, a nuclear protein that activates Toll-like receptors, also enhanced the development of allograft vasculopathy. Complement activation elicits assembly of membrane attack complexes on endothelial cells which activate noncanonical NF-κB signaling, a novel complement effector pathway that induces proinflammatory genes and potentiates endothelial cell-mediated alloimmune T-cell activation, processes which enhance allograft vasculopathy. SUMMARY: Innate immune mediators, including HMGB1, MyD88, and noncanonical NF-κB signaling via complement activation contribute to allograft vasculopathy. These pathways represent potential therapeutic targets to reduce allograft vasculopathy after solid organ transplantation.


Asunto(s)
Aloinjertos/trasplante , Trasplante de Corazón/métodos , Músculo Liso Vascular/metabolismo , Trasplante Homólogo/métodos , Animales , Humanos , Inmunidad Innata , Ratones , Músculo Liso Vascular/patología , FN-kappa B , Receptores Toll-Like
18.
J Immunol ; 188(4): 1933-41, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22246631

RESUMEN

Aging is associated with suboptimal CD8 T cell responses to viral infections. It is not clear whether these poor responses are due to environmental influences or quantitative and qualitative changes in the pool of responding CD8 T cells. Our studies demonstrated several deleterious age-related changes in the pool of Ag-specific CD8 T cells that respond to infection. The majority of CD8 T cells from uninfected aged mice was CD44(Hi) and had increased expression of inhibitory receptors including PD1, LAG3, 2B4, and CD160. These aged CD44(Hi) CD8 T cells were transcriptionally similar to exhausted CD8 T cells found during chronic infections. In addition, the number of virus-specific precursors in aged mice prior to infection was decreased up to 10-fold, and many of these Ag-specific precursors had high expression of CD44 and PD1. Finally, TCR transgenic studies demonstrated that the CD44(Hi) Ag-specific CD8 T cells from unimmunized aged and young mice were qualitatively inferior compared with CD44(Lo) CD8 T cells from aged or young donors. Thus, a decrease in precursor frequency as well as qualitative changes of CD8 T cells during aging are directly related to impaired immunity.


Asunto(s)
Envejecimiento/inmunología , Infecciones por Arenaviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Antígenos CD/biosíntesis , Linfocitos T CD8-positivos/metabolismo , Femenino , Proteínas Ligadas a GPI/biosíntesis , Receptores de Hialuranos/análisis , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/metabolismo , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones , Receptor de Muerte Celular Programada 1/biosíntesis , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Inmunológicos/biosíntesis , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Proteína del Gen 3 de Activación de Linfocitos
19.
JCI Insight ; 9(12)2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38814732

RESUMEN

Influenza poses a persistent health burden worldwide. To design equitable vaccines effective across all demographics, it is essential to better understand how host factors such as genetic background and aging affect the single-cell immune landscape of influenza infection. Cytometry by time-of-flight (CyTOF) represents a promising technique in this pursuit, but interpreting its large, high-dimensional data remains difficult. We have developed a new analytical approach, in silico gating annotating training elucidating (iGATE), based on probabilistic support vector machine classification. By rapidly and accurately "gating" tens of millions of cells in silico into user-defined types, iGATE enabled us to track 25 canonical immune cell types in mouse lung over the course of influenza infection. Applying iGATE to study effects of host genetic background, we show that the lower survival of C57BL/6 mice compared with BALB/c was associated with a more rapid accumulation of inflammatory cell types and decreased IL-10 expression. Furthermore, we demonstrate that the most prominent effect of aging is a defective T cell response, reducing survival of aged mice. Finally, iGATE reveals that the 25 canonical immune cell types exhibited differential influenza infection susceptibility and replication permissiveness in vivo, but neither property varied with host genotype or aging. The software is available at https://github.com/UmichWenLab/iGATE.


Asunto(s)
Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae , Análisis de la Célula Individual , Animales , Ratones , Infecciones por Orthomyxoviridae/inmunología , Análisis de la Célula Individual/métodos , Pulmón/inmunología , Pulmón/virología , Pulmón/patología , Gripe Humana/inmunología , Humanos , Modelos Animales de Enfermedad , Envejecimiento/inmunología , Envejecimiento/genética , Citometría de Flujo/métodos , Linfocitos T/inmunología , Simulación por Computador
20.
Sci Adv ; 10(20): eadk6178, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38748794

RESUMEN

Invasive graft biopsies assess the efficacy of immunosuppression through lagging indicators of transplant rejection. We report on a microporous scaffold implant as a minimally invasive immunological niche to assay rejection before graft injury. Adoptive transfer of T cells into Rag2-/- mice with mismatched allografts induced acute cellular allograft rejection (ACAR), with subsequent validation in wild-type animals. Following murine heart or skin transplantation, scaffold implants accumulate predominantly innate immune cells. The scaffold enables frequent biopsy, and gene expression analyses identified biomarkers of ACAR before clinical signs of graft injury. This gene signature distinguishes ACAR and immunodeficient respiratory infection before injury onset, indicating the specificity of the biomarkers to differentiate ACAR from other inflammatory insult. Overall, this implantable scaffold enables remote evaluation of the early risk of rejection, which could potentially be used to reduce the frequency of routine graft biopsy, reduce toxicities by personalizing immunosuppression, and prolong transplant life.


Asunto(s)
Aloinjertos , Biomarcadores , Rechazo de Injerto , Animales , Rechazo de Injerto/inmunología , Ratones , Trasplante de Piel/efectos adversos , Trasplante de Corazón/efectos adversos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Tejido Subcutáneo/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/inmunología , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA