Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Nature ; 629(8013): 901-909, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38658756

RESUMEN

The liver is the main gateway from the gut, and the unidirectional sinusoidal flow from portal to central veins constitutes heterogenous zones, including the periportal vein (PV) and the pericentral vein zones1-5. However, functional differences in the immune system in each zone remain poorly understood. Here intravital imaging revealed that inflammatory responses are suppressed in PV zones. Zone-specific single-cell transcriptomics detected a subset of immunosuppressive macrophages enriched in PV zones that express high levels of interleukin-10 and Marco, a scavenger receptor that sequesters pro-inflammatory pathogen-associated molecular patterns and damage-associated molecular patterns, and consequently suppress immune responses. Induction of Marco+ immunosuppressive macrophages depended on gut microbiota. In particular, a specific bacterial family, Odoribacteraceae, was identified to induce this macrophage subset through its postbiotic isoallolithocholic acid. Intestinal barrier leakage resulted in inflammation in PV zones, which was markedly augmented in Marco-deficient conditions. Chronic liver inflammatory diseases such as primary sclerosing cholangitis (PSC) and non-alcoholic steatohepatitis (NASH) showed decreased numbers of Marco+ macrophages. Functional ablation of Marco+ macrophages led to PSC-like inflammatory phenotypes related to colitis and exacerbated steatosis in NASH in animal experimental models. Collectively, commensal bacteria induce Marco+ immunosuppressive macrophages, which consequently limit excessive inflammation at the gateway of the liver. Failure of this self-limiting system promotes hepatic inflammatory disorders such as PSC and NASH.


Asunto(s)
Colangitis Esclerosante , Microbioma Gastrointestinal , Inflamación , Hígado , Macrófagos , Enfermedad del Hígado Graso no Alcohólico , Simbiosis , Animales , Femenino , Humanos , Masculino , Ratones , Bacteroidetes/metabolismo , Colangitis Esclerosante/inmunología , Colangitis Esclerosante/microbiología , Colangitis Esclerosante/patología , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Perfilación de la Expresión Génica , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Interleucina-10/inmunología , Interleucina-10/metabolismo , Hígado/inmunología , Hígado/patología , Hígado/microbiología , Macrófagos/citología , Macrófagos/inmunología , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/microbiología , Enfermedad del Hígado Graso no Alcohólico/patología , Vena Porta , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/metabolismo , Análisis de la Célula Individual , Simbiosis/inmunología
2.
Neuron ; 111(19): 2995-3010.e9, 2023 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-37490917

RESUMEN

The brain is generally resistant to regeneration after damage. The cerebral endogenous mechanisms triggering brain self-recovery have remained unclarified to date. We here discovered that the secreted phospholipase PLA2G2E from peri-infarct neurons generated dihomo-γ-linolenic acid (DGLA) as necessary for triggering brain-autonomous neural repair after ischemic brain injury. Pla2g2e deficiency diminished the expression of peptidyl arginine deiminase 4 (Padi4), a global transcriptional regulator in peri-infarct neurons. Single-cell RNA sequencing (scRNA-seq) and epigenetic analysis demonstrated that neuronal PADI4 had the potential for the transcriptional activation of genes associated with recovery processes after ischemic stroke through histone citrullination. Among various DGLA metabolites, we identified 15-hydroxy-eicosatrienoic acid (15-HETrE) as the cerebral metabolite that induced PADI4 in peri-infarct-surviving neurons. Administration of 15-HETrE enhanced functional recovery after ischemic stroke. Thus, our research clarifies the promising potential of brain-autonomous neural repair triggered by the specialized lipids that initiate self-recovery processes after brain injury.


Asunto(s)
Lesiones Encefálicas , Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Animales , Humanos , Ratones , Encéfalo/metabolismo , Lesiones Encefálicas/metabolismo , Infarto/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Metabolismo de los Lípidos
3.
Nat Rev Neurosci ; 24(5): 299-312, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36973481

RESUMEN

The nervous and immune systems control whole-body homeostasis and respond to various types of tissue injury, including stroke, in a coordinated manner. Cerebral ischaemia and subsequent neuronal cell death activate resident or infiltrating immune cells, which trigger neuroinflammation that affects functional prognosis after stroke. Inflammatory immune cells exacerbate ischaemic neuronal injury after the onset of brain ischaemia; however, some of the immune cells thereafter change their function to neural repair. The recovery processes after ischaemic brain injury require additional and close interactions between the nervous and immune systems through various mechanisms. Thus, the brain controls its own inflammation and repair processes after injury via the immune system, which provides a promising therapeutic opportunity for stroke recovery.


Asunto(s)
Lesiones Encefálicas , Isquemia Encefálica , Accidente Cerebrovascular , Humanos , Neuroinmunomodulación , Encéfalo/metabolismo
4.
Neurosci Res ; 187: 67-71, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36206952

RESUMEN

Stroke is a leading cause of death and disability worldwide, but there are a limited number of therapies that improve patients' functional recovery. The complicated mechanisms of post-stroke neuroinflammation, which is responsible for secondary ischemic neuronal damage, have been clarified by extensive research. Activation of microglia and astrocytes due to ischemic insults is implicated in the production of pro-inflammatory factors, formation of the glial scar, and breakdown of the blood-brain barrier. This leads to the infiltration of leukocytes, which are activated by damage-associated molecular patterns (DAMPs) to produce pro-inflammatory factors and induce additional neuronal damage. In this review, we focus on the glial mechanisms underlying sterile post-ischemic inflammation after stroke.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Accidente Cerebrovascular Isquémico/complicaciones , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/metabolismo , Inflamación/etiología , Neuroglía/metabolismo , Microglía/metabolismo , Isquemia Encefálica/complicaciones , Isquemia Encefálica/metabolismo
5.
Semin Immunopathol ; 45(3): 427-435, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36161515

RESUMEN

Severe loss of cerebral blood flow causes hypoxia and glucose deprivation in the brain tissue, resulting in necrotic cell death in the ischemic brain. Several endogenous molecules, called alarmins or damage-associated molecular patterns (DAMPs), are extracellularly released from the dead cells to activate pattern recognition receptors (PRRs) in immune cells that infiltrate into ischemic brain tissue following the disruption of the blood-brain barrier (BBB) after stroke onset. The activated immune cells produce various inflammatory cytokines and chemokines, triggering sterile cerebral inflammation in the ischemic brain that causes further neuronal cell death. Poststroke inflammation is resolved within several days after stroke onset, and neurological functions are restored to some extent as neural repair occurs around peri-infarct neurons. Clearance of DAMPs from the injured brain is necessary for the resolution of poststroke inflammation. Neurons and glial cells also express PRRs and receive DAMP signaling. Although the role of PRRs in neural cells in the ischemic brain has not yet been clarified, the signaling pathway is likely to be contribute to stroke pathology and neural repair after ischemic stroke. This review describes the molecular dynamics, signaling pathways, and functions of DAMPs in poststroke inflammation and its resolution.


Asunto(s)
Alarminas , Accidente Cerebrovascular , Humanos , Inflamación/metabolismo , Encéfalo , Neuronas/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo
6.
Sci Immunol ; 6(64): eabe7915, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34652960

RESUMEN

Immune responses contribute to tissue injury and repair during and after ischemic stroke. However, the spatiotemporal and initiating molecular events remain incompletely understood. Here, we show that mice deficient in the phosphatidylserine receptor CD300a, which is highly expressed on brain myeloid cells including Ly6Chi monocytes, exhibited ameliorated neurological deficit after middle cerebral artery occlusion (MCAO). CD300a inhibited signaling through the CD300b-DNAX-activation protein 12 (DAP12) signaling pathway to prevent efferocytosis of apoptotic cells. Deficiency of CD300a enhanced efferocytosis by myeloid cells infiltrating the brain as early as 1 hour after MCAO and reduced release of damage-associated molecular patterns from dead cells, resulting in milder inflammation in the penumbral region. Treatment with an anti-CD300a neutralizing antibody ameliorated the neurological deficit after MCAO. These findings reveal an important role of efferocytosis in the super-acute phase of ischemic stroke pathology and identified CD300a as a target for immunotherapy in treating ischemic stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico/inmunología , Células Mieloides/inmunología , Neuronas/inmunología , Receptores Inmunológicos/inmunología , Animales , Encéfalo/inmunología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis
7.
PLoS Biol ; 19(5): e3000939, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34014921

RESUMEN

Inflammation is implicated in the onset and progression of various diseases, including cerebral pathologies. Here, we report that DJ-1, which plays a role within cells as an antioxidant protein, functions as a damage-associated molecular pattern (DAMP) and triggers inflammation if released from dead cells into the extracellular space. We first found that recombinant DJ-1 protein induces the production of various inflammatory cytokines in bone marrow-derived macrophages (BMMs) and dendritic cells (BMDCs). We further identified a unique peptide sequence in the αG and αH helices of DJ-1 that activates Toll-like receptor 2 (TLR2) and TLR4. In the ischemic brain, DJ-1 is released into the extracellular space from necrotic neurons within 24 h after stroke onset and makes direct contact with TLR2 and TLR4 in infiltrating myeloid cells. Although DJ-1 deficiency in a murine model of middle cerebral artery occlusion did not attenuate neuronal injury, the inflammatory cytokine expression in infiltrating immune cells was significantly decreased. Next, we found that the administration of an antibody to neutralize extracellular DJ-1 suppressed cerebral post-ischemic inflammation and attenuated ischemic neuronal damage. Our results demonstrate a previously unknown function of DJ-1 as a DAMP and suggest that extracellular DJ-1 could be a therapeutic target to prevent inflammation in tissue injuries and neurodegenerative diseases.


Asunto(s)
Isquemia Encefálica/metabolismo , Proteína Desglicasa DJ-1/metabolismo , Alarminas/metabolismo , Animales , Encéfalo/metabolismo , Isquemia Encefálica/fisiopatología , Citocinas/inmunología , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/inmunología , Infarto de la Arteria Cerebral Media/patología , Inflamación , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Proteína Desglicasa DJ-1/fisiología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/fisiopatología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo
8.
Inflamm Regen ; 40(1): 28, 2020 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-33292860

RESUMEN

Therapeutic strategies for regulating neuroinflammation are expected in the development of novel therapeutic agents to prevent the progression of central nervous system (CNS) pathologies. An understanding of the detailed molecular and cellular mechanisms of neuroinflammation in each CNS disease is necessary for the development of therapeutics. Since the brain is a sterile organ, neuroinflammation in Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS) is triggered by cerebral cellular damage or the abnormal accumulation of inflammatogenic molecules in CNS tissue through the activation of innate and acquired immunity. Inflammation and CNS pathologies worsen each other through various cellular and molecular mechanisms, such as oxidative stress or the accumulation of inflammatogenic molecules induced in the damaged CNS tissue. In this review, we summarize the recent evidence regarding sterile immune responses in neurodegenerative diseases.

9.
Int Immunol ; 32(11): 719-725, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-32300780

RESUMEN

Stroke is one of the major causes of lethality and disability, yet few effective therapies have been established for ischemic stroke. Inflammation in the ischemic brain is induced by the infiltration and subsequent activation of immune cells. Loss of cerebral blood flow and ischemic brain-cell death trigger the activation of infiltrating immune cells and drastic changes in the lipid content of the ischemic brain. In particular, polyunsaturated fatty acids and their metabolites regulate cerebral post-ischemic inflammation and ischemic stroke pathologies. In this review, we discuss the relationships between the lipid mediators and cerebral post-ischemic inflammation and their relevance to possible future therapeutic strategies targeting lipid mediators for ischemic stroke.


Asunto(s)
Mediadores de Inflamación/inmunología , Inflamación/inmunología , Accidente Cerebrovascular Isquémico/inmunología , Lípidos/inmunología , Animales , Humanos
10.
J Biochem ; 165(6): 459-464, 2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-30796426

RESUMEN

Cerebral inflammation is a promising therapeutic target for ischaemic stroke. After ischaemic stroke, inflammatogenic self-molecules, which originate from damaged brain tissue due to ischaemia, activate infiltrating immune cells (neutrophils, macrophages and lymphocytes) and thereby trigger sterile inflammation. Innate immunity plays the central role in sterile inflammation at the acute phase of brain ischaemia, although immune response by T lymphocytes (innate or acquired immunity) is also implicated in inflammation at the subacute phase, which sustains ischaemic brain damage. In the recovery phase, infiltrating macrophages remove the damage-associated molecular patterns (DAMPs) from the ischaemic brain. These pro-resolving myeloid cells also produce neurotrophic factors involved in neural repair. Through a series of inflammatory mechanisms activated by ischaemic stroke, various immune cells change their functions from inflammation to repair in a precise process. In order to establish therapeutic strategies for the improvement of neurological deficits after ischaemic stroke, it is necessary to clarify the detailed molecular and cellular mechanisms of sterile inflammation after ischaemic brain injury.


Asunto(s)
Isquemia Encefálica/metabolismo , Inflamación/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Isquemia Encefálica/patología , Humanos , Inflamación/patología , Macrófagos/metabolismo , Macrófagos/patología , Accidente Cerebrovascular/patología
11.
Nature ; 565(7738): 246-250, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30602786

RESUMEN

In addition to maintaining immune tolerance, FOXP3+ regulatory T (Treg) cells perform specialized functions in tissue homeostasis and remodelling1,2. However, the characteristics and functions of brain Treg cells are not well understood because there is a low number of Treg cells in the brain under normal conditions. Here we show that there is massive accumulation of Treg cells in the mouse brain after ischaemic stroke, and this potentiates neurological recovery during the chronic phase of ischaemic brain injury. Although brain Treg cells are similar to Treg cells in other tissues such as visceral adipose tissue and muscle3-5, they are apparently distinct and express unique genes related to the nervous system including Htr7, which encodes the serotonin receptor 5-HT7. The amplification of brain Treg cells is dependent on interleukin (IL)-2, IL-33, serotonin and T cell receptor recognition, and infiltration into the brain is driven by the chemokines CCL1 and CCL20. Brain Treg cells suppress neurotoxic astrogliosis by producing amphiregulin, a low-affinity epidermal growth factor receptor (EGFR) ligand. Stroke is a leading cause of neurological disability, and there are currently few effective recovery methods other than rehabilitation during the chronic phase. Our findings suggest that Treg cells and their products may provide therapeutic opportunities for neuronal protection against stroke and neuroinflammatory diseases.


Asunto(s)
Astrocitos/patología , Isquemia Encefálica/inmunología , Isquemia Encefálica/patología , Gliosis/patología , Neuroprotección/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Animales , Encéfalo/citología , Encéfalo/inmunología , Movimiento Celular , Proliferación Celular , Quimiocina CCL1/inmunología , Quimiocina CCL20/inmunología , Interleucina-2/inmunología , Interleucina-33/inmunología , Interleucina-6/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/inmunología , Receptores CCR/metabolismo , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Factor de Transcripción STAT3/metabolismo , Serotonina/metabolismo , Transducción de Señal , Linfocitos T Reguladores/metabolismo
12.
Neurochem Int ; 130: 104316, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30342960

RESUMEN

Stroke causes neuronal cell death and destruction of neuronal circuits in the brain and spinal cord. Injury to the brain tissue induces sterile inflammation triggered by the extracellular release of endogenous molecules, but cerebral inflammation after stroke is gradually resolved within several days. In this pro-resolving process, inflammatory cells adopt a pro-resolving or repairing phenotype in the injured brain, activating endogenous repairing programs. Although the mechanisms involved in the transition from inflammation to neural repair after stroke remain largely unknown to date, some of the mechanisms for inflammation and neural repair have been clarified in detail. This review focuses on the molecular or cellular mechanisms involved in sterile inflammation and neural repair after stroke. This accumulation of evidence may be helpful for speculating about the endogenous repairing mechanisms in the brain and identifying therapeutic targets for improving the functional prognoses of stroke patients.


Asunto(s)
Isquemia Encefálica/inmunología , Encéfalo/inmunología , Red Nerviosa/inmunología , Células-Madre Neurales/inmunología , Neurogénesis/fisiología , Accidente Cerebrovascular/inmunología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Red Nerviosa/metabolismo , Red Nerviosa/patología , Células-Madre Neurales/metabolismo , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
13.
Semin Immunopathol ; 40(6): 523-538, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30206661

RESUMEN

Inflammatory responses play a multifaceted role in regulating both disability and recovery after ischemic brain injury. In the acute phase of ischemic stroke, resident microglia elicit rapid inflammatory responses by the ischemic milieu. After disruption of the blood-brain barrier, peripheral-derived neutrophils and mononuclear phagocytes infiltrate into the ischemic brain. These infiltrating myeloid cells are activated by the endogenous alarming molecules released from dying brain cells. Inflammation after ischemic stroke thus typically consists of sterile inflammation triggered by innate immunity, which exacerbates the pathologies of ischemic stroke and worsens neurological prognosis. Infiltrating immune cells sustain the post-ischemic inflammation for several days; after this period, however, these cells take on a repairing function, phagocytosing inflammatory mediators and cellular debris. This time-specific polarization of immune cells in the ischemic brain is a potential novel therapeutic target. In this review, we summarize the current understanding of the phase-dependent role of innate myeloid cells in ischemic stroke and discuss the cellular and molecular mechanisms of their inflammatory or repairing polarization from a therapeutic perspective.


Asunto(s)
Isquemia Encefálica/inmunología , Encéfalo/inmunología , Inflamación/inmunología , Células Mieloides/inmunología , Accidente Cerebrovascular/inmunología , Animales , Encéfalo/patología , Humanos , Inmunidad Innata/fisiología , Inflamación/patología , Células Mieloides/patología , Fagocitosis/fisiología , Accidente Cerebrovascular/patología
14.
Nihon Yakurigaku Zasshi ; 151(1): 9-14, 2018.
Artículo en Japonés | MEDLINE | ID: mdl-29321396

RESUMEN

Inflammation is an essential step for the pathology of ischemic stroke, and is also an important therapeutic target for developing novel therapeutic methods which have a wide therapeutic time window. Since there is no pathogen in the brain, the inflammation will be triggered by some endogenous molecules which are called as danger associated molecular patterns (DAMPs). So far two important DAMPs, high mobility group box 1 (HMGB1) and peroxiredoxin (PRX), have been recently identified in the ischemic brain. HMGB1 exaggerates the disruption of blood brain barrier; on the other hand, PRX activates mononuclear phagocytes and induces the inflammatory cytokine production through the activation of Toll-like receptor 2 (TLR2) and TLR4. Various inflammatory molecules produced from infiltrating immune cells have been known to exacerbate the neurological deficits of ischemic stroke patients. Recently, it has been paid attention that the inflammation after tissue injury also induces tissue repair, while its mechanisms remain to be clarified. Novel therapeutic methods will be established by clarifying detailed molecular mechanisms underlying the induction of neural repair after cerebral post-ischemic inflammation.


Asunto(s)
Isquemia Encefálica/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/inmunología , Humanos , Inflamación/inmunología , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/genética
15.
Nat Commun ; 8: 15338, 2017 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-28530241

RESUMEN

Adoptive T-cell immunotherapy is a promising approach to cancer therapy. Stem cell memory T (TSCM) cells have been proposed as a class of long-lived and highly proliferative memory T cells. CD8+ TSCM cells can be generated in vitro from naive CD8+ T cells via Wnt signalling; however, methods do not yet exist for inducing TSCM cells from activated or memory T cells. Here, we show a strategy for generating TSCM-like cells in vitro (iTSCM cells) from activated CD4+ and CD8+ T cells in mice and humans by coculturing with stromal cells that express a Notch ligand. iTSCM cells lose PD-1 and CTLA-4 expression, and produce a large number of tumour-specific effector cells after restimulation. This method could therefore be used to generate antigen-specific effector T cells for adoptive immunotherapy.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Activación de Linfocitos , Receptores Notch/metabolismo , Linfocitos T/citología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Antígeno CTLA-4/metabolismo , Diferenciación Celular , Línea Celular , Proliferación Celular , Separación Celular , Técnicas de Cocultivo , Citometría de Flujo , Homeostasis , Humanos , Memoria Inmunológica , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptor de Muerte Celular Programada 1/metabolismo , Células Madre/citología
16.
Nat Med ; 23(6): 723-732, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28394332

RESUMEN

Damage-associated molecular patterns (DAMPs) trigger sterile inflammation after tissue injury, but the mechanisms underlying the resolution of inflammation remain unclear. In this study, we demonstrate that common DAMPs, such as high-mobility-group box 1 (HMGB1), peroxiredoxins (PRXs), and S100A8 and S100A9, were internalized through the class A scavenger receptors MSR1 and MARCO in vitro. In ischemic murine brain, DAMP internalization was largely mediated by MSR1. An elevation of MSR1 levels in infiltrating myeloid cells observed 3 d after experimental stroke was dependent on the transcription factor Mafb. Combined deficiency for Msr1 and Marco, or for Mafb alone, in infiltrating myeloid cells caused impaired clearance of DAMPs, more severe inflammation, and exacerbated neuronal injury in a murine model of ischemic stroke. The retinoic acid receptor (RAR) agonist Am80 increased the expression of Mafb, thereby enhancing MSR1 expression. Am80 exhibited therapeutic efficacy when administered, even at 24 h after the onset of experimental stroke. Our findings uncover cellular mechanisms contributing to DAMP clearance in resolution of the sterile inflammation triggered by tissue injury.


Asunto(s)
Alarminas/inmunología , Encéfalo/inmunología , Infarto de la Arteria Cerebral Media/inmunología , Factor de Transcripción MafB/inmunología , Células Mieloides/inmunología , Receptores Inmunológicos/inmunología , Receptores Depuradores de Clase A/inmunología , Animales , Benzoatos/farmacología , Encéfalo/efectos de los fármacos , Isquemia Encefálica/inmunología , Sistemas CRISPR-Cas , Calgranulina A/inmunología , Calgranulina B/inmunología , Inmunoprecipitación de Cromatina , Proteína HMGB1/inmunología , Inflamación , Factor de Transcripción MafB/efectos de los fármacos , Factor de Transcripción MafB/genética , Ratones , Células Mieloides/metabolismo , Peroxirredoxinas/inmunología , Receptores Inmunológicos/genética , Receptores de Ácido Retinoico/agonistas , Receptores Depuradores de Clase A/efectos de los fármacos , Receptores Depuradores de Clase A/genética , Accidente Cerebrovascular/inmunología , Tetrahidronaftalenos/farmacología
17.
Int Immunol ; 29(2): 59-70, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28338748

RESUMEN

Damage-associated molecular patterns (DAMPs) have been implicated in sterile inflammation in various tissue injuries. High-mobility group box 1 (HMGB1) is a representative DAMP, and has been shown to transmit signals through receptors for advanced glycation end products (RAGEs) and TLRs, including TLR2 and TLR4. HMGB1 does not, however, bind to TLRs with high affinity; therefore, the mechanism of HMGB1-mediated TLR activation remains unclear. In this study, we found that fluorescently labeled HMGB1 was efficiently internalized into macrophages through class A scavenger receptors. Although both M1- and M2-type macrophages internalized HMGB1, only M1-type macrophages secreted cytokines in response to HMGB1. The pan-class A scavenger receptor competitive inhibitor, maleylated bovine serum albumin (M-BSA), inhibited HMGB1 internalization and reduced cytokine production from macrophages in response to HMGB1 but not to LPS. The C-terminal acidic domain of HMGB1 is responsible for scavenger receptor-mediated internalization and cytokine production. HMGB1 and TLR4 co-localized in macrophages, and this interaction was disrupted by M-BSA, suggesting that class A scavenger receptors function as co-receptors of HMGB1 for TLR activation. M-BSA ameliorated LPS-induced sepsis and dextran sulfate sodium (DSS)-induced colitis models in which HMGB1 has been shown to play progressive roles. These data suggest that scavenger receptors function as co-receptors along with TLRs for HMGB1 in M1-type inflammatory macrophages.


Asunto(s)
Colitis/inmunología , Macrófagos/fisiología , Receptores Inmunológicos/metabolismo , Receptores Depuradores de Clase A/metabolismo , Sepsis/inmunología , Animales , Bovinos , Diferenciación Celular , Células Cultivadas , Colitis/inducido químicamente , Citocinas/metabolismo , Proteína HMGB1/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptores Inmunológicos/genética , Receptores Depuradores de Clase A/genética , Sepsis/inducido químicamente , Albúmina Sérica Bovina/administración & dosificación , Transducción de Señal , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
18.
Nihon Rinsho ; 74(4): 573-8, 2016 Apr.
Artículo en Japonés | MEDLINE | ID: mdl-27333742

RESUMEN

Post-ischemic inflammation is re-appraised as an important player in the progression of ischemic stroke. Activation of inflammatory cells via Toll-like receptor 2 (TLR2) and TLR4 is caused by several damage-associated molecular patterns (DAMPs), including high mobility group box-1 (HMGB-1) and heat shock proteins. We have recently found that peroxiredoxin (Prx) is one of the strong DAMPs and activates infiltrating macrophages in brain ischemia. We have also found that interleukin-23 (IL-23) from the activated macrophages stimulates γδT cells which release IL-17, thereby causing the delayed expansion of infarct lesions. Further investigation of the innate immune response would lead to development of novel stroke treatment with a broad therapeutic time window.


Asunto(s)
Inflamación/genética , Accidente Cerebrovascular , Encéfalo/patología , Progresión de la Enfermedad , Proteína HMGB1 , Proteínas de Choque Térmico , Humanos , Inmunidad Innata , Inflamación/inmunología , Interleucina-17 , Interleucina-23 , Activación de Macrófagos , Peroxirredoxinas/fisiología , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/inmunología , Receptor Toll-Like 2 , Receptor Toll-Like 4
19.
Int Immunol ; 28(5): 233-43, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26647405

RESUMEN

Lipopolysaccharide (LPS)-induced acute lung injury (ALI) is known as a mouse model of acute respiratory distress syndrome; however, the function of T-cell-derived cytokines in ALI has not yet been established. We found that LPS challenge in one lung resulted in a rapid induction of innate-type pro-inflammatory cytokines such as IL-6 and TNF-α, followed by the expression of T-cell-type cytokines, including IL-17, IL-22 and IFN-γ. We discovered that IL-23 is important for ALI, since blockage of IL-23 by gene disruption or anti-IL-12/23p40 antibody treatment reduced neutrophil infiltration and inflammatory cytokine secretion into the bronchoalveolar lavage fluid (BALF). IL-23 was mostly produced from F4/80(+)CD11c(+) alveolar macrophages, and IL-23 expression was markedly reduced by the pre-treatment of mice with antibiotics, suggesting that the development of IL-23-producing macrophages required commensal bacteria. Unexpectedly, among T-cell-derived cytokines, IL-22 rather than IL-17 or IFN-γ played a major role in LPS-induced ALI. IL-22 protein levels were higher than IL-17 in the BALF after LPS instillation, and the major source of IL-22 was memory Th17 cells. Lung memory CD4(+) T cells had a potential to produce IL-22 at higher levels than IL-17 in response to IL-1ß plus IL-23 without TCR stimulation. Our study revealed an innate-like function of the lung memory Th17 cells that produce IL-22 in response to IL-23 and are involved in exaggeration of ALI.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Citocinas/inmunología , Inmunidad Innata/efectos de los fármacos , Lipopolisacáridos/toxicidad , Pulmón/inmunología , Células Th17/inmunología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Lavado Broncoalveolar , Citocinas/genética , Inmunidad Innata/genética , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Pulmón/patología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Células Th17/patología
20.
J Exp Med ; 212(10): 1623-40, 2015 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-26304965

RESUMEN

Regulatory T (T reg) cells are central mediators of immune suppression. As such, T reg cells are characterized by a distinct pattern of gene expression, which includes up-regulation of immunosuppressive genes and silencing of inflammatory cytokine genes. Although an increasing number of transcription factors that regulate T reg cells have been identified, the mechanisms by which the T reg cell-specific transcriptional program is maintained and executed remain largely unknown. The Nr4a family of nuclear orphan receptors, which we recently identified as essential for the development of T reg cells, is highly expressed in mature T reg cells as well, suggesting that Nr4a factors play important roles even beyond T reg cell development. Here, we showed that deletion of Nr4a genes specifically in T reg cells caused fatal systemic immunopathology. Nr4a-deficient T reg cells exhibited global alteration of the expression of genes which specify the T reg cell lineage, including reduction of Foxp3 and Ikzf4. Furthermore, Nr4a deficiency abrogated T reg cell suppressive activities and accelerated conversion to cells with Th2 and follicular helper T (Tfh) effector-like characteristics, with heightened expression of Th2 and Tfh cytokine genes. These findings demonstrate that Nr4a factors play crucial roles in mature T reg cells by directly controlling a genetic program indispensable for T reg cell maintenance and function.


Asunto(s)
Receptores de Esteroides/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Interleucina-4/genética , Interleucina-4/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Proteínas del Tejido Nervioso/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/inmunología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/inmunología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/inmunología , Receptores de Hormona Tiroidea/metabolismo , Células Th2/inmunología , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA