Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Neurosci ; 43(2): 319-332, 2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36446585

RESUMEN

Mechanical impact-induced primary injury after traumatic brain injury (TBI) leads to acute microglial pro-inflammatory activation and consequently mediates neurodegeneration, which is a major secondary brain injury mechanism. However, the detailed pathologic cascades have not been fully elucidated, partially because of the pathologic complexity in animal TBI models. Although there are several in vitro TBI models, none of them closely mimic post-TBI microglial activation. In the present study, we aimed to establish an in vitro TBI model, specifically reconstituting the pro-inflammatory activation and associated neurodegeneration following TBI. We proposed three sets of experiments. First, we established a needle scratch injured neuron-induced microglial activation and neurodegeneration in vitro model of TBI. Second, we compared microglial pro-inflammatory cytokines profiles between the in vitro TBI model and TBI in male mice. Additionally, we validated the role of injured neurons-derived damage-associated molecular patterns in amplifying microglial pro-inflammatory pathways using the in vitro TBI model. Third, we applied the in vitro model for the first time to characterize the cellular metabolic profile of needle scratch injured-neuron-activated microglia, and define the role of metabolic reprogramming in mediating pro-inflammatory microglial activation and mediated neurodegeneration. Our results showed that we successfully established a novel in vitro TBI model, which closely mimics primary neuronal injury-triggered microglial pro-inflammatory activation and mediated neurodegeneration after TBI. This in vitro model provides an advanced and highly translational platform for dissecting interactions in the pathologic processes of neuronal injury-microglial activation-neuronal degeneration cascade, and elucidating the detailed underlying cellular and molecular insights after TBI.SIGNIFICANCE STATEMENT Microglial activation is a key component of acute neuroinflammation that leads to neurodegeneration and long-term neurologic outcome deficits after TBI. However, it is not feasible to truly dissect primary neuronal injury-induced microglia activation, and consequently mediated neurodegeneration in vivo Furthermore, there is currently lacking of in vitro TBI models closely mimicking the TBI primary injury-mediated microglial activation. In this study, we successfully established and validated a novel in vitro TBI model of microglial activation, and for the first time, characterized the cellular metabolic profile of microglia in this model. This novel microglial activation in vitro TBI model will help in elucidating microglial inflammatory activation and consequently associated neurodegeneration after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Microglía , Ratones , Masculino , Animales , Microglía/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Macrófagos/metabolismo , Neuronas/metabolismo , Ratones Endogámicos C57BL
2.
Circ Res ; 128(1): 62-75, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33070717

RESUMEN

RATIONALE: Hemorrhagic complications represent a major limitation of intravenous thrombolysis using tPA (tissue-type plasminogen activator) in patients with ischemic stroke. The expression of tPA receptors on immune cells raises the question of what effects tPA exerts on these cells and whether these effects contribute to thrombolysis-related hemorrhagic transformation. OBJECTIVE: We aim to determine the impact of tPA on immune cells and investigate the association between observed immune alteration with hemorrhagic transformation in ischemic stroke patients and in a rat model of embolic stroke. METHODS AND RESULTS: Paired blood samples were collected before and 1 hour after tPA infusion from 71 patients with ischemic stroke. Control blood samples were collected from 27 ischemic stroke patients without tPA treatment. A rat embolic middle cerebral artery occlusion model was adopted to investigate the underlying mechanisms of hemorrhagic transformation. We report that tPA induces a swift surge of circulating neutrophils and T cells with profoundly altered molecular features in ischemic stroke patients and a rat model of focal embolic stroke. tPA exacerbates endothelial injury, increases adhesion and migration of neutrophils and T cells, which are associated with brain hemorrhage in rats subjected to embolic stroke. Genetic ablation of annexin A2 in neutrophils and T cells diminishes the effect of tPA on these cells. Decoupling the interaction between mobilized neutrophils/T cells and the neurovascular unit, achieved via a S1PR (sphingosine-1-phosphate receptor) 1 modulator RP101075 and a CCL2 (C-C motif chemokine ligand 2) synthesis inhibitor bindarit, which block lymphocyte egress and myeloid cell recruitment, respectively, attenuates hemorrhagic transformation and improves neurological function after tPA thrombolysis. CONCLUSIONS: Our findings suggest that immune invasion of the neurovascular unit represents a previously unrecognized mechanism underlying tPA-mediated brain hemorrhage, which can be overcome by precise immune modulation during thrombolytic therapy.


Asunto(s)
Accidente Cerebrovascular Embólico/tratamiento farmacológico , Fibrinolíticos/toxicidad , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Hemorragias Intracraneales/inducido químicamente , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Neutrófilos/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Terapia Trombolítica , Activador de Tejido Plasminógeno/toxicidad , Animales , Anexina A2/metabolismo , Línea Celular , Quimiocina CCL2/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Modelos Animales de Enfermedad , Accidente Cerebrovascular Embólico/sangre , Accidente Cerebrovascular Embólico/inmunología , Femenino , Fibrinolíticos/administración & dosificación , Humanos , Infarto de la Arteria Cerebral Media/sangre , Infarto de la Arteria Cerebral Media/inmunología , Infusiones Intravenosas , Hemorragias Intracraneales/sangre , Hemorragias Intracraneales/inmunología , Accidente Cerebrovascular Isquémico/sangre , Accidente Cerebrovascular Isquémico/inmunología , Masculino , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Ratas Wistar , Receptores de Esfingosina-1-Fosfato/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Activador de Tejido Plasminógeno/administración & dosificación
3.
J Neuroinflammation ; 18(1): 173, 2021 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-34372870

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) is a significant cause of death and disability worldwide. The TLR4-NFκB signaling cascade is the critical pro-inflammatory activation pathway of leukocytes after TBI, and modulating this signaling cascade may be an effective therapeutic target for treating TBI. Previous studies indicate that recombinant annexin A2 (rA2) might be an interactive molecule modulating the TLR4-NFκB signaling; however, the role of rA2 in regulating this signaling pathway in leukocytes after TBI and its subsequent effects have not been investigated. METHODS: C57BL/6 mice were subjected to TBI and randomly divided into groups that received intraperitoneal rA2 or vehicle at 2 h after TBI. The peripheral leukocyte activation and infiltrating immune cells were examined by flow cytometry, RT-qPCR, and immunostaining. The neutrophilic TLR4 expression on the cell membrane was examined by flow cytometry and confocal microscope, and the interaction of annexin A2 with TLR4 was assessed by co-immunoprecipitation coupled with Western blotting. Neuroinflammation was measured via cytokine proteome profiler array and RT-qPCR. Neurodegeneration was determined by Western blotting and immunostaining. Neurobehavioral assessments were used to monitor motor and cognitive function. Brain tissue loss was assessed via MAP2 staining. RESULTS: rA2 administration given at 2 h after TBI significantly attenuates neutrophil activation and brain infiltration at 24 h of TBI. In vivo and in vitro data show that rA2 binds to and reduces TLR4 expression on the neutrophil surface and suppresses TLR4/NFκB signaling pathway in neutrophils at 12 h after TBI. Furthermore, rA2 administration also reduces pro-inflammation of brain tissues within 24 h and neurodegeneration at 48 h after TBI. Lastly, rA2 improves long-term sensorimotor ability and cognitive function, and reduces brain tissue loss at 28 days after TBI. CONCLUSIONS: Systematic rA2 administration at 2 h after TBI significantly inhibits activation and brain infiltration of peripheral leukocytes, especially neutrophils at the acute phase. Consequently, rA2 reduces the detrimental brain pro-inflammation-associated neurodegeneration and ultimately ameliorates neurological deficits after TBI. The underlying molecular mechanism might be at least in part attributed to rA2 bindings to pro-inflammatory receptor TLR4 in peripheral leukocytes, thereby blocking NFκB signaling activation pathways following TBI.


Asunto(s)
Anexina A2/administración & dosificación , Lesiones Traumáticas del Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Cognición/efectos de los fármacos , Leucocitos/efectos de los fármacos , Animales , Encéfalo/metabolismo , Citocinas/metabolismo , Leucocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo
4.
Stroke ; 51(3): 967-974, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32019481

RESUMEN

Background and Purpose- Microglia are among the first cells to respond to intracerebral hemorrhage (ICH), but the mechanisms that underlie their activity following ICH remain unclear. IL (interleukin)-15 is a proinflammatory cytokine that orchestrates homeostasis and the intensity of the immune response following central nervous system inflammatory events. The goal of this study was to investigate the role of IL-15 in ICH injury. Methods- Using brain slices of patients with ICH, we determined the presence and cellular source of IL-15. A transgenic mouse line with targeted expression of IL-15 in astrocytes was generated to determine the role of astrocytic IL-15 in ICH. The expression of IL-15 was controlled by a glial fibrillary acidic protein promoter (GFAP-IL-15tg). ICH was induced by intraparenchymal injection of collagenase or autologous blood. Results- In patients with ICH and wild-type mice subjected to experimental ICH, we found a significant upregulation of IL-15 in astrocytes. In GFAP-IL-15tg mice, we found that astrocyte-targeted expression of IL-15 exacerbated brain edema and neurological deficits following ICH. This aggravated ICH injury in GFAP-IL-15tg mice is accompanied by increased microglial accumulation in close proximity to astrocytes in perihematomal tissues. Additionally, microglial expression of CD86, IL-1ß, and TNF-α is markedly increased in GFAP-IL-15tg mice following ICH. Furthermore, depletion of microglia using a colony stimulating factor 1 receptor inhibitor diminishes the exacerbation of ICH injury in GFAP-IL-15tg mice. Conclusions- Our findings identify IL-15 as a mediator of the crosstalk between astrocytes and microglia that exacerbates brain injury following ICH.


Asunto(s)
Astrocitos/inmunología , Lesiones Encefálicas/inmunología , Hemorragia Cerebral/inmunología , Interleucina-15/inmunología , Microglía/inmunología , Anciano , Anciano de 80 o más Años , Animales , Astrocitos/patología , Lesiones Encefálicas/etiología , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/genética , Hemorragia Cerebral/patología , Femenino , Humanos , Interleucina-15/genética , Masculino , Ratones , Ratones Transgénicos , Microglía/patología
5.
FASEB J ; 32(5): 2757-2767, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29401578

RESUMEN

Stroke-induced immune suppression predisposes the host to infections and can contribute to high morbidity and mortality in stroke patients. Because ischemic stroke has a profound effect on the systemic immune response, which may explain the increased susceptibility of stroke patients to infection, an urgent need persists for a better understanding of mechanisms associated with immune suppression; new and effective treatments for stroke can then be identified. NK cells play a key role in early host defense against pathogens by killing infected cells and/or producing cytokines such as IFN-γ. Because the phenotype and function of peripheral NK cells have been widely investigated in ischemic stroke, nCounter Inflammation Gene Array Analysis was used to build immune-related gene profiles of NK cells to comprehensively analyze the molecular signature of NK cells after ischemic brain injury. We observed distinct gene expression profiles reflecting different splenic NK-cell phenotypes and functional properties across the time course of transient middle cerebral artery occlusion (MCAO). Based on gene expression and pathway-network analysis, lower expression levels of signal transducer and activator of transcription-3 (STAT3) were observed in animals with MCAO compared with sham control animals. Genetic activation of STAT3 through the introduction of STAT3 clustered regularly interspaced short palindromic repeats (CRISPR) plasmid prevented the loss of NK-cell-derived IFN-γ production after MCAO, together with reduced bacterial burden and mortality. Our data suggest that brain ischemia impairs NK-cell-mediated immune defense in the periphery, at least in part through the JAK-STAT3 pathway, which can be readdressed by modulating STAT3 activation status.-Jin, W.-N., Ducruet, A. F., Liu, Q., Shi, S. X.-Y., Waters, M., Zou, M., Sheth, K. N., Gonzales, R., Shi, F.-D. Activation of JAK/STAT3 restores NK-cell function and improves immune defense after brain ischemia.


Asunto(s)
Isquemia Encefálica/inmunología , Regulación de la Expresión Génica/inmunología , Interferón gamma/inmunología , Quinasas Janus/inmunología , Células Asesinas Naturales/inmunología , Factor de Transcripción STAT3/inmunología , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Interferón gamma/genética , Quinasas Janus/genética , Células Asesinas Naturales/patología , Masculino , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Transcripción STAT3/genética
6.
J Immunol ; 197(7): 2610-7, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27543617

RESUMEN

Myasthenia gravis (MG) is a chronic humoral immunity-mediated autoimmune disorder of the neuromuscular junction characterized by muscle weakness. Follicular helper T (Tfh) cells may be the key Th cell subset that promotes MG development, as their major function is helping B cell activation and Ab production. Aberrance of thymus-derived Tfh cells might be implicated in autoimmune diseases including MG; just how circulating Tfh cells, especially those from patients with a normal thymus, contribute to MG pathogenesis remains to be uncovered. In this article, we characterize a population of circulating CD4(+)CXCR5(+)PD-1(+) Tfh cells in ocular and generalized MG patients without thymic abnormalities and demonstrate that the circulating Tfh cells are significantly enriched in generalized MG patients but not in ocular MG patients compared with healthy subjects, whereas a proportion of follicular regulatory T cells decreased in MG patients. In addition, the frequency of plasma cells and B cells was higher and the serum levels of IL-6/IL-21 were also elevated in these MG patients. The activated Tfh1 and Tfh17 in Tfh cells are the major source for IL-21 production in MG patients. A strong correlation between Tfh cells and the plasma cell frequency and anti-acetylcholine receptor Ab titers was evident in generalized MG patients. In particular, we found that Tfh cells derived from MG patients promoted B cells to produce Abs in an IL-21 signaling-dependent manner. Collectively, our results suggest that circulating Tfh cells may act on autoreactive B cells and thus contribute to the development of MG in patients without thymic abnormalities.


Asunto(s)
Linfocitos B/inmunología , Miastenia Gravis/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/patología , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miastenia Gravis/patología , Estudios Prospectivos
8.
Muscle Nerve ; 49(4): 487-94, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23868194

RESUMEN

INTRODUCTION: A subset of regulatory B cells in humans and mice has been defined functionally by their ability to produce interleukin (IL)-10. We characterized IL-10-producing B (B10) cells in myasthenia gravis (MG) patients and correlated them with disease activity and responsiveness to rituximab therapy. METHODS: Frequencies of B10 cells from MG patients and healthy controls were monitored by fluorescence-activated cell sorting (FACS). RESULTS: MG patients had fewer B10 cells than controls, which was associated with more severe disease status. Moreover, patients who responded well to rituximab therapy exhibited rapid repopulation of B10 cells, whereas in patients who did not respond well to rituximab, B10 cell repopulation was delayed. The kinetics of B10 cells were related to the responsiveness to rituximab in MG. CONCLUSIONS: We have characterized a specific subset of B10 cells in MG patients which may serve as a marker for disease activity and responsiveness to immune therapy.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Linfocitos B/metabolismo , Factores Inmunológicos/uso terapéutico , Interleucina-10/biosíntesis , Miastenia Gravis/tratamiento farmacológico , Miastenia Gravis/metabolismo , Adulto , Anticuerpos Monoclonales de Origen Murino/farmacología , Linfocitos B/efectos de los fármacos , Biomarcadores/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Factores Inmunológicos/farmacología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Rituximab , Resultado del Tratamiento , Adulto Joven
9.
Aging Dis ; 14(2): 560-571, 2023 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-37008068

RESUMEN

Patients with intracerebral hemorrhage (ICH) often suffer from heterogeneous long-term neurological deficits, such as cognitive decline. Our ability to measure secondary brain injury to predict the long-term outcomes of these patients is limited. We investigated whether the blood neurofilament light chain (NfL) can monitor brain injury and predict long-term outcomes in patients with ICH. We enrolled 300 patients with first-episode ICH within 24 h recruited in the Chinese Cerebral Hemorrhage Mechanisms and Intervention study cohort from January 2019 to June 2020. Patients were prospectively followed up for 12 months. Blood samples were collected from 153 healthy participants. Plasma NfL levels determined using a single-molecule array revealed a biphasic increase in plasma NfL in ICH patients compared to healthy controls, with the first peak at around 24 h and a second elevation from day 7 through day 14 post-ICH. Plasma NfL levels were positively correlated with hemorrhage volume, National Institute of Health Stroke Scale, and Glasgow Coma Scale scores of ICH patients. Higher NfL concentration within 72 h after ictus was independently associated with 6- and 12-month worsened functional outcomes (modified Rankin Scale ≥ 3) and higher all-cause mortality. Magnetic resonance imaging and cognitive function evaluation were available for 26 patients at 6 months post-ICH, and NfL levels measured 7 days post-ictus correlated with decreased white matter fiber integrity and poor cognitive function at 6 months after stroke. These findings suggest that blood NfL is a sensitive marker for monitoring axonal injury post-ICH and can predict long-term functional ability and survival.

10.
Sci Adv ; 9(23): eabq0712, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-37285421

RESUMEN

Leukocyte infiltration accelerates brain injury following intracerebral hemorrhage (ICH). Yet, the involvement of T lymphocytes in this process has not been fully elucidated. Here, we report that CD4+ T cells accumulate in the perihematomal regions in the brains of patients with ICH and ICH mouse models. T cells activation in the ICH brain is concurrent with the course of perihematomal edema (PHE) development, and depletion of CD4+ T cells reduced PHE volumes and improved neurological deficits in ICH mice. Single-cell transcriptomic analysis revealed that brain-infiltrating T cells exhibited enhanced proinflammatory and proapoptotic signatures. Consequently, CD4+ T cells disrupt the blood-brain barrier integrity and promote PHE progression through interleukin-17 release; furthermore, the TRAIL-expressing CD4+ T cells engage DR5 to trigger endothelial death. Recognition of T cell contribution to ICH-induced neural injury is instrumental for designing immunomodulatory therapies for this dreadful disease.


Asunto(s)
Lesiones Encefálicas , Linfocitos T , Ratones , Animales , Linfocitos T/metabolismo , Encéfalo/metabolismo , Hemorragia Cerebral/etiología , Hemorragia Cerebral/metabolismo , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Modelos Animales de Enfermedad
11.
Ann Neurol ; 69(4): 721-34, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21425186

RESUMEN

OBJECTIVE: The role of natural killer (NK) cells in regulating multiple sclerosis (MS) is not well understood. Additional studies with NK cells might provide insight into the mechanism of action of MS therapies such as daclizumab, an antibody against the interleukin (IL)-2R α-chain, which induces expansion of CD56(bright) NK cells. METHODS: In a relapsing-remitting form of the experimental autoimmune encephalomyelitis (EAE) model of MS induced in SJL mice, we expanded NK cells with IL-2 coupled with an anti-IL-2 monoclonal antibody (mAb) and evaluated the effects of these NK cells on EAE. Further, we investigated the effect of the human version of IL-2/IL-2 mAb on NK cells from MS patients and its effect on central nervous system (CNS) inflammation and pathology in a human-mouse chimera model and assessed the underlying mechanisms. RESULTS: IL-2/IL-2 mAb dramatically expands NK cells both in the peripheral lymphoid organs and in the CNS, and attenuates CNS inflammation and neurological deficits. Disease protection is conferred by CNS-resident NK cells. Importantly, the human version of IL-2/IL-2 mAb restored the defective CD56(+) NK cells from MS patients in a human-mouse chimera model. Both the CD56(bright) and CD56(dim) subpopulations were required to attenuate disease in this model. INTERPRETATION: These findings unveil the immunotherapeutic potential of NK cells, which can act as critical suppressor cells in target organs of autoimmunity. These results also have implications to better understand the mechanism of action of daclizumab in MS.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inmunoterapia/métodos , Interleucina-2/inmunología , Interleucina-2/farmacología , Células Asesinas Naturales/inmunología , Adulto , Animales , Anticuerpos Monoclonales/uso terapéutico , Autoinmunidad , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Sistema Nervioso Central/patología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Citometría de Flujo , Granzimas/metabolismo , Humanos , Inflamación/inmunología , Células Asesinas Naturales/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/inmunología , Perforina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores/inmunología
12.
Cells ; 11(13)2022 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-35805099

RESUMEN

In the pathophysiology of hemorrhagic stroke, the perturbation of the neurovascular unit (NVU), a functional group of the microvascular and brain intrinsic cellular components, is implicated in the progression of secondary injury and partially informs the ultimate patient outcome. Given the broad NVU functions in maintaining healthy brain homeostasis through its maintenance of nutrients and energy substrates, partitioning central and peripheral immune components, and expulsion of protein and metabolic waste, intracerebral hemorrhage (ICH)-induced dysregulation of the NVU directly contributes to numerous destructive processes in the post-stroke sequelae. In ICH, the damaged NVU precipitates the emergence and evolution of perihematomal edema as well as the breakdown of the blood-brain barrier structural coherence and function, which are critical facets during secondary ICH injury. As a gateway to the central nervous system, the NVU is among the first components to interact with the peripheral immune cells mobilized toward the injured brain. The release of signaling molecules and direct cellular contact between NVU cells and infiltrating leukocytes is a factor in the dysregulation of NVU functions and further adds to the acute neuroinflammatory environment of the ICH brain. Thus, the interactions between the NVU and immune cells, and their reverberating consequences, are an area of increasing research interest for understanding the complex pathophysiology of post-stroke injury. This review focuses on the interactions of T-lymphocytes, a major cell of the adaptive immunity with expansive effector function, with the NVU in the context of ICH. In cataloging the relevant clinical and experimental studies highlighting the synergistic actions of T-lymphocytes and the NVU in ICH injury, this review aimed to feature emergent knowledge of T cells in the hemorrhagic brain and their diverse involvement with the neurovascular unit in this disease.


Asunto(s)
Accidente Cerebrovascular , Linfocitos T , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Hemorragia Cerebral/metabolismo , Humanos , Accidente Cerebrovascular/metabolismo , Linfocitos T/metabolismo
13.
Transl Stroke Res ; 13(2): 311-325, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34523038

RESUMEN

Type 2 diabetes mellitus (T2DM) is a major comorbidity exacerbating ischemic brain injury and impairing post-stroke recovery. Our previous study suggested that recombinant human fibroblast growth factor (rFGF) 21 might be a potent therapeutic targeting multiple aspects of pathophysiology in T2DM stroke. This study aims to evaluate the potential effects of rFGF21 on cerebrovascular remodeling after T2DM stroke. Permanent distal middle cerebral artery occlusion was performed in heterozygous non-diabetic db/ + and homozygous diabetic db/db mice. Daily rFGF21 administration was initiated 1 week after stroke induction and maintained for up to 2 weeks thereafter. Multiple markers associated with post-stroke recovery, including angiogenesis, oligodendrogenesis, white matter integrity, and neurogenesis, were assessed up to 3 weeks after stroke. Our results showed an impairment in post-stroke vascular remodeling under T2DM condition, reflected by the decreased expression of trophic factors in brain microvessels and impairments of angiogenesis. The defected cerebrovascular remodeling was accompanied by the decreased oligodendrogenesis and neurogenesis. However, delayed rFGF21 administration normalized post-stroke hyperglycemia and improved neurological outcomes, which may partially be via the promotion of pro-angiogenic trophic factor expression in brain microvessels and cerebrovascular remodeling. The better cerebrovascular remodeling may also contribute to oligodendrogenesis, white matter integrity, and neurogenesis after T2DM stroke. Therefore, delayed rFGF21 administration may improve neurological outcomes in T2DM stroke mice, at least in part by normalizing the metabolic abnormalities and promoting cerebrovascular remodeling and white matter repair.


Asunto(s)
Isquemia Encefálica , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Accidente Cerebrovascular , Sustancia Blanca , Animales , Isquemia Encefálica/complicaciones , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Ratones , Ratones Endogámicos C57BL , Accidente Cerebrovascular/complicaciones
14.
Sci Transl Med ; 13(589)2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33853930

RESUMEN

Acute brain injury mobilizes circulating leukocytes to transmigrate into the perivascular space and brain parenchyma. This process amplifies neural injury. Bone marrow hematopoiesis replenishes the exhausted peripheral leukocyte pools. However, it is not known whether brain injury influences the development of bone marrow lineages and how altered hematopoietic cell lineages affect neurological outcome. Here, we showed that bone marrow hematopoietic stem cells (HSCs) can be swiftly skewed toward the myeloid lineage in patients with intracerebral hemorrhage (ICH) and experimental ICH models. Lineage tracing revealed a predominantly augmented hematopoiesis of Ly6Clow monocytes infiltrating the ICH brain, where they generated alternatively activated macrophages and suppressed neuroinflammation and brain injury. The ICH brain uses ß3-adrenergic innervation that involves cell division cycle 42 to promote bone marrow hematopoiesis of Ly6Clow monocytes, which could be further potentiated by the U.S. Food and Drug Administration-approved ß3-adrenergic agonist mirabegron. Our results suggest that brain injury modulates HSC lineage development to curb distal brain inflammation, implicating the bone marrow as a unique niche for self-protective neuroimmune interaction that might be exploited to obtain therapeutic effects.


Asunto(s)
Médula Ósea , Lesiones Encefálicas , Células de la Médula Ósea , Linaje de la Célula , Hematopoyesis , Células Madre Hematopoyéticas , Humanos , Nicho de Células Madre
15.
J Clin Med ; 10(17)2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34501242

RESUMEN

Blood-brain barrier (BBB) disruption following ischemic stroke (IS) contributes to hemorrhagic transformation, brain edema, increased neural dysfunction, secondary injury, and mortality. Brain endothelial cells form a para and transcellular barrier to most blood-borne solutes via tight junctions (TJs) and rare transcytotic vesicles. The prevailing view attributes the destruction of TJs to the resulting BBB damage following IS. Recent studies define a stepwise impairment of the transcellular barrier followed by the paracellular barrier which accounts for the BBB leakage in IS. The increased endothelial transcytosis that has been proven to be caveolae-mediated, precedes and is independent of TJs disintegration. Thus, our understanding of post stroke BBB deficits needs to be revised. These recent findings could provide a conceptual basis for the development of alternative treatment strategies. Presently, our concept of how BBB endothelial transcytosis develops is incomplete, and treatment options remain limited. This review summarizes the cellular structure and biological classification of endothelial transcytosis at the BBB and reviews related molecular mechanisms. Meanwhile, relevant transcytosis-targeted therapeutic strategies for IS and research entry points are prospected.

16.
Neurosci Lett ; 735: 135160, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32561451

RESUMEN

Endothelial sphingosine-1-phosphate receptors are emerging as relevant therapeutic targets during acute ischemic stroke (AIS). Physiologically, the cerebrovascular endothelium plays a vital role in maintaining barrier integrity and cerebrovascular homeostasis. During a cerebral ischemic event, products from parenchymal cell death are released and trigger vascular endothelial dysfunction and vascular inflammation leading to barrier integrity disruption. Endothelial dysfunction, inflammation, and a breach in barrier property play a significant role in contributing to a vicious cycle which promotes brain edema formation and exacerbates neuronal injury post stroke. Data from experimental stroke models and clinical trials suggest that selective sphingosine-1-phosphate receptor type 1 (S1PR1) modulation improves endothelial health and function and, as a result, contributes to improved neurological outcome post ischemic injury. This review highlights the impact of sphingosine-1-phosphate (S1P)/S1PR1 signaling involved in blood brain barrier (BBB) integrity and cerebrovascular inflammation following AIS. We focus on the beneficial actions of S1PR1 signaling during ischemic injury including barrier protection to lessen brain edema formation and reduction in the development and progression of vascular inflammation by attenuating endothelial cell activation resulting in reduced neurovascular inflammation. Potential gaps and future directions related to the role of S1PR during AIS are also discussed.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Inflamación/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Animales , Barrera Hematoencefálica/patología , Humanos , Inflamación/patología , Accidente Cerebrovascular Isquémico/patología
17.
J Exp Med ; 217(12)2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-32870258

RESUMEN

Perihematomal edema (PHE) occurs within hours after intracerebral hemorrhage (ICH), leading to secondary injury manifested by impaired blood-brain barrier (BBB) integrity and destruction of adjacent tissue. To dissect the mechanisms underlying PHE formation, we profiled human and mouse perihematomal tissues and identified natural killer (NK) cells as the predominant immune cell subset that outnumbers other infiltrating immune cell types during early stages of ICH. Unbiased clustering of single-cell transcriptional profiles revealed two major NK cell subsets that respectively possess high cytotoxicity or robust chemokine production features in the brain after ICH, distinguishing them from NK cells of the periphery. NK cells exacerbate BBB disruption and brain edema after ICH via cytotoxicity toward cerebral endothelial cells and recruitment of neutrophils that augment focal inflammation. Thus, brain-bound NK cells acquire new features that contribute to PHE formation and neurological deterioration following ICH.


Asunto(s)
Edema Encefálico/etiología , Encéfalo/patología , Hemorragia Cerebral/complicaciones , Progresión de la Enfermedad , Células Asesinas Naturales/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Edema Encefálico/inmunología , Hemorragia Cerebral/inmunología , Quimiocina CXCL2/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Femenino , Humanos , Inflamación/patología , Células Asesinas Naturales/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Poli I-C/farmacología , Transcriptoma/genética
18.
Curr Pharm Des ; 24(22): 2556-2563, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30058481

RESUMEN

BACKGROUND: Conventional antidepressants are thought to produce their impact on clinical symptoms by increasing the central availability of biogenic amine neurotransmitters (the monoamine hypothesis of depression). These drugs continue to be the primary medicines used in major depressive disorder. Although they have biological effects after acute dosing, full antidepressant response generally takes weeks of daily administration. Lack of rapid onset is a large limitation in antidepressant therapy (e.g., suicide, lack of medication compliance, difficulty switching medications). METHODS: The present review of the literature discusses the preclinical and clinical findings on compounds that can produce immediate symptom relief. RESULTS: These compounds include ketamine, scopolamine, and mechanistically-related drugs. Newer additions to the list of potential rapid-acting agents include antagonists of metabotropic (mGlu) 2/3 receptors, negative allosteric modulators of α5-containing GABAA receptors, and psychedelic compounds. An additional benefit of these compounds is that they have demonstrated large effect sizes and, importantly, demonstrated efficacy in patient's refractory to other treatments. A drawback of some of these compounds, to date, is finding ways to expand the duration of clinical efficacy. In addition, for some compounds, the side-effect profile requires management. A primary mechanism by which rapid effects might be produced is through the amplification of excitatory neurotransmission through activation of AMPA receptors. The extracellular efflux of glutamate induced by these drugs has been documented and provides the hypothesized triggering mechanism for AMPA receptor amplification. CONCLUSION: The preclinical and clinical literature strongly suggests that rapid-acting antidepressants are the current focus of antidepressant drug discovery. Promising clinical findings exist for several compounds including ketamine and other NMDA receptor antagonists, scopolamine, and psilocybin. Two compounds are in late stage clinical development: GLYX-13 (Rapastinel) and eskekamine.


Asunto(s)
Antidepresivos/uso terapéutico , Trastorno Depresivo Mayor/tratamiento farmacológico , Alucinógenos/farmacología , Humanos , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Resultado del Tratamiento
19.
J Cereb Blood Flow Metab ; 37(6): 2224-2236, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28273719

RESUMEN

Brain ischemia elicits microglial activation and microglia survival depend on signaling through colony-stimulating factor 1 receptor (CSF1R). Although depletion of microglia has been linked to worse stroke outcomes, it remains unclear to what extent and by what mechanisms activated microglia influence ischemia-induced inflammation and injury in the brain. Using a mouse model of transient focal cerebral ischemia and reperfusion, we demonstrated that depletion of microglia via administration of the dual CSF1R/c-Kit inhibitor PLX3397 exacerbates neurodeficits and brain infarction. Depletion of microglia augmented the production of inflammatory mediators, leukocyte infiltration, and cell death during brain ischemia. Of note, microglial depletion-induced exacerbation of stroke severity did not solely depend on lymphocytes and monocytes. Importantly, depletion of microglia dramatically augmented the production of inflammatory mediators by astrocytes after brain ischemia . In vitro studies reveal that microglia restricted ischemia-induced astrocyte response and provided neuroprotective effects. Our findings suggest that neuroprotective effects of microglia may result, in part, from its inhibitory action on astrocyte response after ischemia.


Asunto(s)
Isquemia Encefálica/inmunología , Isquemia Encefálica/patología , Mediadores de Inflamación/metabolismo , Microglía/inmunología , Microglía/patología , Aminopiridinas/farmacología , Animales , Isquemia Encefálica/diagnóstico por imagen , Células Cultivadas , Modelos Animales de Enfermedad , Imagen por Resonancia Magnética , Masculino , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Pirroles/farmacología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores
20.
J Cereb Blood Flow Metab ; 36(8): 1464-76, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26661207

RESUMEN

Recent studies have demonstrated that lymphocytes play a key role in ischemic brain injury. However, there is still a lack of viable approaches to non-invasively track infiltrating lymphocytes and reveal their key spatiotemporal events in the inflamed central nervous system (CNS). Here we describe an in vivo imaging approach for sequential monitoring of brain-infiltrating CD4(+) T cells in experimental ischemic stroke. We show that magnetic resonance imaging (MRI) or Xenogen imaging combined with labeling of SPIO-Molday ION Rhodamine-B (MIRB) can be used to monitor the dynamics of CD4(+) T cells in a passive transfer model. MIRB-labeled CD4(+) T cells can be longitudinally visualized in the mouse brain and peripheral organs such as the spleen and liver after cerebral ischemia. Immunostaining of tissue sections showed similar kinetics of MIRB-labeled CD4(+) T cells when compared with in vivo observations. Our results demonstrated the use of MIRB coupled with in vivo imaging as a valid method to track CD4(+) T cells in ischemic brain injury. This approach will facilitate future investigations to identify the dynamics and key spatiotemporal events for brain-infiltrating lymphocytes in CNS inflammatory diseases.


Asunto(s)
Isquemia Encefálica/sangre , Linfocitos T CD4-Positivos/citología , Rastreo Celular/métodos , Colorantes Fluorescentes/química , Imagen por Resonancia Magnética/métodos , Nanopartículas de Magnetita/química , Animales , Isquemia Encefálica/diagnóstico por imagen , Isquemia Encefálica/inmunología , Proteínas de Unión al ADN/genética , Fluorescencia , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Rodaminas/química , Coloración y Etiquetado/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA