Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros




Base de datos
Asunto de la revista
Intervalo de año de publicación
1.
Sci Immunol ; 6(56)2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33891558

RESUMEN

Opportunities to interrogate the immune responses in the injured tissue of living patients suffering from acute sterile injuries such as stroke and heart attack are limited. We leveraged a clinical trial of minimally invasive neurosurgery for patients with intracerebral hemorrhage (ICH), a severely disabling subtype of stroke, to investigate the dynamics of inflammation at the site of brain injury over time. Longitudinal transcriptional profiling of CD14+ monocytes/macrophages and neutrophils from hematomas of patients with ICH revealed that the myeloid response to ICH within the hematoma is distinct from that in the blood and occurs in stages conserved across the patient cohort. Initially, hematoma myeloid cells expressed a robust anabolic proinflammatory profile characterized by activation of hypoxia-inducible factors (HIFs) and expression of genes encoding immune factors and glycolysis. Subsequently, inflammatory gene expression decreased over time, whereas anti-inflammatory circuits were maintained and phagocytic and antioxidative pathways up-regulated. During this transition to immune resolution, glycolysis gene expression and levels of the potent proresolution lipid mediator prostaglandin E2 remained elevated in the hematoma, and unexpectedly, these elevations correlated with positive patient outcomes. Ex vivo activation of human macrophages by ICH-associated stimuli highlighted an important role for HIFs in production of both inflammatory and anti-inflammatory factors, including PGE2, which, in turn, augmented VEGF production. Our findings define the time course of myeloid activation in the human brain after ICH, revealing a conserved progression of immune responses from proinflammatory to proresolution states in humans after brain injury and identifying transcriptional programs associated with neurological recovery.


Asunto(s)
Encéfalo/patología , Hemorragia Cerebral/complicaciones , Enfermedades Neuroinflamatorias/inmunología , Adulto , Anciano , Encéfalo/inmunología , Células Cultivadas , Hemorragia Cerebral/inmunología , Hemorragia Cerebral/patología , Femenino , Voluntarios Sanos , Hematoma , Humanos , Estudios Longitudinales , Macrófagos/inmunología , Masculino , Persona de Mediana Edad , Enfermedades Neuroinflamatorias/patología , Neutrófilos/inmunología , Cultivo Primario de Células , RNA-Seq , Transcriptoma/inmunología
2.
BMC Immunol ; 19(1): 42, 2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30577749

RESUMEN

It has been highlighted that the original article [1] contained a typesetting mistake in the middle name of Arthur F. Steinschneider.

3.
BMC Immunol ; 19(1): 30, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30376808

RESUMEN

BACKGROUND: Transcriptional profiling with ultra-low input methods can yield valuable insights into disease, particularly when applied to the study of immune cells using RNA-sequencing. The advent of these methods has allowed for their use in profiling cells collected in clinical trials and other studies that involve the coordination of human-derived material. To date, few studies have sought to quantify what effects that collection and handling of this material can have on resulting data. RESULTS: We characterized the global effects of blood handling, methods for leukocyte isolation, and preservation media on low numbers of immune cells isolated from blood. We found overall that storage/shipping temperature of blood prior to leukocyte isolation and sorting led to global changes in both CD8+ T cells and monocytes, including alterations in immune-related gene sets. We found that the use of a leukocyte filtration system minimized these alterations and we applied this method to generate high-quality transcriptional data from sorted immune cells isolated from the blood of intracerebral hemorrhage patients and matched healthy controls. CONCLUSIONS: Our data underscore the necessity of processing samples with comparably defined protocols prior to transcriptional profiling and demonstrate that a filtration method can be applied to quickly isolate immune cells of interest while minimizing transcriptional bias.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Procedimientos de Reducción del Leucocitos/métodos , Leucocitos Mononucleares/inmunología , Transcriptoma , Linfocitos T CD8-positivos/citología , Perfilación de la Expresión Génica , Humanos , Leucocitos Mononucleares/citología , Análisis de Secuencia de ARN/métodos
4.
J Clin Invest ; 128(2): 607-624, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29251628

RESUMEN

Macrophages are a source of both proinflammatory and restorative functions in damaged tissue through complex dynamic phenotypic changes. Here, we sought to determine whether monocyte-derived macrophages (MDMs) contribute to recovery after acute sterile brain injury. By profiling the transcriptional dynamics of MDMs in the murine brain after experimental intracerebral hemorrhage (ICH), we found robust phenotypic changes in the infiltrating MDMs over time and demonstrated that MDMs are essential for optimal hematoma clearance and neurological recovery. Next, we identified the mechanism by which the engulfment of erythrocytes with exposed phosphatidylserine directly modulated the phenotype of both murine and human MDMs. In mice, loss of receptor tyrosine kinases AXL and MERTK reduced efferocytosis of eryptotic erythrocytes and hematoma clearance, worsened neurological recovery, exacerbated iron deposition, and decreased alternative activation of macrophages after ICH. Patients with higher circulating soluble AXL had poor 1-year outcomes after ICH onset, suggesting that therapeutically augmenting efferocytosis may improve functional outcomes by both reducing tissue injury and promoting the development of reparative macrophage responses. Thus, our results identify the efferocytosis of eryptotic erythrocytes through AXL/MERTK as a critical mechanism modulating macrophage phenotype and contributing to recovery from ICH.


Asunto(s)
Hemorragia Cerebral/patología , Eritrocitos/clasificación , Macrófagos/citología , Animales , Apoptosis , Lesiones Encefálicas , Eritrocitos/citología , Hematoma/metabolismo , Humanos , Inmunidad Innata , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fagocitosis , Fenotipo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Solubilidad , Resultado del Tratamiento , Tirosina Quinasa c-Mer/metabolismo , Tirosina Quinasa del Receptor Axl
5.
J Clin Invest ; 127(1): 280-292, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-27893460

RESUMEN

Intracerebral hemorrhage (ICH) is a devastating form of stroke that results from the rupture of a blood vessel in the brain, leading to a mass of blood within the brain parenchyma. The injury causes a rapid inflammatory reaction that includes activation of the tissue-resident microglia and recruitment of blood-derived macrophages and other leukocytes. In this work, we investigated the specific responses of microglia following ICH with the aim of identifying pathways that may aid in recovery after brain injury. We used longitudinal transcriptional profiling of microglia in a murine model to determine the phenotype of microglia during the acute and resolution phases of ICH in vivo and found increases in TGF-ß1 pathway activation during the resolution phase. We then confirmed that TGF-ß1 treatment modulated inflammatory profiles of microglia in vitro. Moreover, TGF-ß1 treatment following ICH decreased microglial Il6 gene expression in vivo and improved functional outcomes in the murine model. Finally, we observed that patients with early increases in plasma TGF-ß1 concentrations had better outcomes 90 days after ICH, confirming the role of TGF-ß1 in functional recovery from ICH. Taken together, our data show that TGF-ß1 modulates microglia-mediated neuroinflammation after ICH and promotes functional recovery, suggesting that TGF-ß1 may be a therapeutic target for acute brain injury.


Asunto(s)
Lesiones Encefálicas/sangre , Hemorragia Cerebral/sangre , Microglía/metabolismo , Recuperación de la Función , Transducción de Señal , Factor de Crecimiento Transformador beta1/sangre , Animales , Lesiones Encefálicas/etiología , Hemorragia Cerebral/genética , Hemorragia Cerebral/patología , Regulación de la Expresión Génica , Interleucina-6/biosíntesis , Interleucina-6/genética , Ratones , Ratones Transgénicos , Microglía/patología , Factor de Crecimiento Transformador beta1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA