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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 36(12): e22616, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36394527

RESUMEN

Cerebral ischemia activates neural progenitors that participate in brain remodeling following acute injury. Sphingosine-1-phosphate receptor (S1PR) signaling governs cell proliferation and mobilization, yet its potential impact on neural progenitors and stroke recovery remains poorly understood. The goal of this study was to investigate the impact of S1PR modulation on post-stroke neurogenesis and functional recovery, using a S1PR modulator BAF312. Mice were subjected to 60 min middle cerebral artery occlusion (MCAO) and received BAF312 starting from day 3 after MCAO until the end of experiment. BAF312 facilitated motor function recovery in MCAO mice until day 14 after surgery. Flow cytometry analysis revealed that BAF312 treatment led to an increase of type A cells in subventricular zone (SVZ), but not other progenitor cell subsets in MCAO mice. We found an increase of BrdU incorporation in SVZ DCX+ cells from MCAO mice receiving BAF312 and augmented proliferation of DCX+ cells in cultured neurospheres isolated from SVZ tissues. Notably, a S1PR1 antagonist W146 abolished BAF312-induced increase of SVZ type A cells from MCAO mice and proliferation of DCX+ cells in cultured neurospheres. Additionally, the benefit of BAF312 to improve neurogenesis and stroke recovery remains in Rag2-/- mice lacking of T and B cells. Our results demonstrate that S1PR modulation improves neurogenesis and functional recovery following brain ischemia.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular , Ratones , Animales , Recuperación de la Función , Receptores de Esfingosina-1-Fosfato , Neurogénesis/fisiología , Accidente Cerebrovascular/tratamiento farmacológico , Infarto de la Arteria Cerebral Media
2.
Anal Chem ; 94(23): 8474-8482, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35652329

RESUMEN

A circular nonuniform electric field strategy coupled with gel electrophoresis was proposed to control the precise separation and efficient concentration of nano- and microparticles. The circular nonuniform electric field has the feature of exponential increase in the electric field intensity along the radius, working with three functional zones of migration, acceleration, and concentration. The distribution form of electric field lines is regulated in functional zones to control the migration behaviors of particles for separation and concentration by altering the relative position of the ring electrode (outside) and rodlike electrode (inner). The circular nonuniform electric field promotes the target-type and high-precision separation of nanoparticles based on the difference in charge-to-size ratio. The concentration multiple of nanoparticles is also controlled randomly with the alternation of radius, taking advantage of vertical extrusion and concentric converging of the migration path. This work provides a brand new insight into the simultaneous separation and concentration of particles and is promising for developing a versatile tool for the separation and preparation of various samples instead of conventional methods.


Asunto(s)
Electricidad , Nanopartículas , Electrodos , Electroforesis/métodos , Tamaño de la Partícula
3.
FASEB J ; 35(11): e21856, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34606651

RESUMEN

Neuromyelitis optica spectrum disorder (NMOSD) is a severe central nervous system (CNS) autoimmune disease that primarily damages the optic nerves and spinal cord. Group 2 innate lymphoid cells (ILC2) are potent producers of type 2 cytokines that orchestrate immune and inflammatory responses. However, the role of ILC2 in CNS autoimmune diseases remains unknown. In patients with NMOSD, we identified a significant reduction of ILC2 in peripheral blood, which was correlated with disease severity. Using a mouse model of NMOSD induced by intracerebral injection of NMOSD-IgG with complement, we found CNS infiltration of ILC2 mainly expressing interleukin (IL)-5 and IL-13. The depletion of ILC2 led to increased CNS lesion volume, reduced CNS glucose metabolism, and augmented astrocyte injury and demyelination. The exacerbated NMOSD pathology was accompanied by increased accumulation of Iba1+ cells and complement activity in CNS lesions. In addition, the expansion of ILC2 using IL-33 attenuated NMO pathology. Collectively, these findings suggest a beneficial role of ILC2 in NMOSD, which deserves further investigation for future design of immune therapies to treat patients with NMOSD.


Asunto(s)
Inmunidad Innata , Linfocitos/inmunología , Neuromielitis Óptica/inmunología , Neuromielitis Óptica/patología , Índice de Severidad de la Enfermedad , Adulto , Animales , Astrocitos/metabolismo , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Proteínas del Sistema Complemento/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-33/administración & dosificación , Interleucina-33/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neuromielitis Óptica/sangre , Neuromielitis Óptica/tratamiento farmacológico , Proteínas Recombinantes/administración & dosificación , Resultado del Tratamiento
4.
FASEB J ; 35(6): e21617, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33982343

RESUMEN

Intracerebral hemorrhage (ICH) is a severe stroke subtype without effective pharmacological treatment. Following ICH, peripheral leukocytes infiltrate into the brain and contribute to neuroinflammation and brain edema. However, the intercellular machinery controlling the initiation and propagation of leukocyte infiltration remains elusive. Exosomes are small extracellular vesicles released from donor cells and bridge intercellular communication. In this study, we investigated the effects of inhibition of exosome release on neuroinflammation and ICH injury. Using a mouse model of ICH induced by collagenase injection, we found that ICH induced an increase of exosome level in the brain. Inhibition of exosome release using GW4869 augmented neurological deficits and brain edema after ICH. The exacerbation of ICH injury was accompanied by increased barrier disruption and brain infiltration of leukocytes. The detrimental effects of GW4869 were ablated in ICH mice receiving antibody depletion of Gr-1+ myeloid cells. Extracted exosomes from the ICH brains suppressed the production of inflammatory factors by splenocytes. Additionally, exosomes extracted from brain tissues of donor ICH mice reduced ICH injury in recipient mice. These results demonstrate that inhibition of exosome release augments neuroinflammation and ICH injury. The impact of exosomes released from the ICH brain on the immune system deserves further investigation.


Asunto(s)
Encéfalo/patología , Hemorragia Cerebral/complicaciones , Exosomas/patología , Inflamación/patología , Neuronas/patología , Animales , Encéfalo/metabolismo , Hemorragia Cerebral/inducido químicamente , Exosomas/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo
5.
Proc Natl Acad Sci U S A ; 114(3): E396-E405, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-27994144

RESUMEN

Astrocytes are believed to bridge interactions between infiltrating lymphocytes and neurons during brain ischemia, but the mechanisms for this action are poorly understood. Here we found that interleukin-15 (IL-15) is dramatically up-regulated in astrocytes of postmortem brain tissues from patients with ischemic stroke and in a mouse model of transient focal brain ischemia. We generated a glial fibrillary acidic protein (GFAP) promoter-controlled IL-15-expressing transgenic mouse (GFAP-IL-15tg) line and found enlarged brain infarcts, exacerbated neurodeficits after the induction of brain ischemia. In addition, knockdown of IL-15 in astrocytes attenuated ischemic brain injury. Interestingly, the accumulation of CD8+ T and natural killer (NK) cells was augmented in these GFAP-IL-15tg mice after brain ischemia. Of note, depletion of CD8+ T or NK cells attenuated ischemic brain injury in GFAP-IL-15tg mice. Furthermore, knockdown of the IL-15 receptor α or blockade of cell-to-cell contact diminished the activation and effector function of CD8+ T and NK cells in GFAP-IL-15tg mice, suggesting that astrocytic IL-15 is delivered in trans to target cells. Collectively, these findings indicate that astrocytic IL-15 could aggravate postischemic brain damage via propagation of CD8+ T and NK cell-mediated immunity.


Asunto(s)
Astrocitos/metabolismo , Lesiones Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Inmunidad Celular/inmunología , Interleucina-15/inmunología , Interleucina-15/metabolismo , Anciano de 80 o más Años , Animales , Astrocitos/inmunología , Encéfalo/inmunología , Encéfalo/metabolismo , Lesiones Encefálicas/inmunología , Isquemia Encefálica/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/inmunología , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/inmunología , Ratones Transgénicos/metabolismo , Neuroglía/inmunología , Neuroglía/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Regiones Promotoras Genéticas/inmunología , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/metabolismo , Regulación hacia Arriba/inmunología
6.
FASEB J ; 32(6): 3336-3345, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29401614

RESUMEN

The activation of microglia and the various substances they produce have been linked to the pathologic development of Parkinson's disease (PD), but the precise role of microglia in PD remains to be defined. The survival of microglia depends on colony-stimulating factor 1 receptor (CSF1R) signaling, and CSF1R inhibition results in rapid elimination of microglia in the central nervous system. Using a mouse PD model induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment, we showed that the depletion of microglia via the CSF1R inhibitor PLX3397 exacerbated the impairment of locomotor activities and the loss of dopaminergic neurons. Further, depletion of microglia augmented the production of inflammatory mediators and infiltration of leukocytes in the brain after MPTP exposure. Microglia depletion-induced aggravation of MPTP neurotoxicity was also seen in lymphocyte-deficient mice. In addition, the depletion of microglia did not affect the production of brain-derived neurotrophic factor, but it dramatically augmented the production of inflammatory mediators by astrocytes after MPTP treatment. Our findings suggest microglia play a protective role against MPTP-induced neuroinflammation and dopaminergic neurotoxicity.-Yang, X., Ren, H., Wood, K., Li, M., Qiu, S., Shi, F.-D., Ma, C., Liu, Q. Depletion of microglia augments the dopaminergic neurotoxicity of MPTP.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Mediadores de Inflamación/metabolismo , Leucocitos/metabolismo , Intoxicación por MPTP/metabolismo , Microglía/metabolismo , Aminopiridinas/farmacología , Animales , Neuronas Dopaminérgicas/patología , Leucocitos/patología , Intoxicación por MPTP/patología , Masculino , Ratones , Ratones Noqueados , Microglía/patología , Pirroles/farmacología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores
7.
FASEB J ; 32(1): 220-229, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28877956

RESUMEN

Severe brain injury significantly influences immune responses; however, the levels at which this influence occurs and which neurogenic pathways are involved are not well defined. Here, we used MRI to measure spleen volume and tissue diffusion changes in patients with intracerebral hemorrhage (ICH). We observed increased capillary exchange and spleen shrinkage by d 3 post-ICH, with recovery by d 14. The extent of spleen shrinkage was associated with brain hematoma size, and a reduced progression of perihematomal edema was observed in the presence of severe spleen shrinkage. At the cellular level, lymphopenia was present in patients with ICH at admission and persisted up to 14 d. Lymphopenia did not parallel the observed spleen alteration. In addition, patients with ICH with infection had significant deficiencies of T and NK cells and poor functional outcomes. Finally, in mouse models of ICH, spleen shrinkage could be related to innervations from adrenergic input and the hypothalamus-pituitary-adrenal (HPA) axis. In sum, the profound impact of ICH on the immune system involves the coordinated actions of sympathetic innervation and the HPA axis, which modulate spleen shrinkage and cellular immunity.-Zhang, J., Shi, K., Li, Z., Li, M., Han, Y., Wang, L., Zhang, Z., Yu, C., Zhang, F., Song, L., Dong, J.-F., La Cava, A., Sheth, K. N., Shi, F.-D. Organ- and cell-specific immune responses are associated with the outcomes of intracerebral hemorrhage.


Asunto(s)
Hemorragia Cerebral/inmunología , Anciano , Animales , Edema Encefálico/diagnóstico por imagen , Hemorragia Cerebral/diagnóstico por imagen , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Hematoma/diagnóstico por imagen , Humanos , Sistema Hipotálamo-Hipofisario/inmunología , Inmunidad Celular , Células Asesinas Naturales/inmunología , Linfopenia/inmunología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neuroinmunomodulación , Sistema Hipófiso-Suprarrenal/inmunología , Bazo/diagnóstico por imagen , Bazo/inmunología , Linfocitos T/inmunología
8.
Stroke ; 49(1): 184-192, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29212744

RESUMEN

BACKGROUND AND PURPOSE: Intracerebral hemorrhage (ICH) is a devastating disease without effective treatment. As a key component of the innate immune system, the NOD-like receptor (NLR) family, NLRP3 (pyrin domain-containing protein 3) inflammasome, when activated after ICH, promotes neuroinflammation and brain edema. MCC950 is a potent, selective, small-molecule NLRP3 inhibitor that blocks NLRP3 activation at nanomolar concentrations. Here, we examined the effect of MCC950 on brain injury and inflammation in 2 models of ICH in mice. METHODS: In mice with ICH induced by injection of autologous blood or bacterial collagenase, we determined the therapeutic potential of MCC950 and its mechanisms of neuroprotection. RESULTS: MCC950 reduced IL-1ß (interleukin-1ß) production and attenuated neurodeficits and perihematomal brain edema after ICH induction by injection of either autologous blood or collagenase. In mice with autologous blood-induced ICH, the protection of MCC950 was associated with reduced leukocyte infiltration into the brain and microglial production of IL-6. MCC950 improved blood-brain barrier integrity and diminished cell death. Notably, the protective effect of MCC950 was abolished in mice depleted of either microglia or Gr-1+ myeloid cells. CONCLUSIONS: These results indicate that the NLRP3 inflammasome inhibitor, MCC950, attenuates brain injury and inflammation after ICH. Hence, NLRP3 inflammasome inhibition is a potential therapy for ICH that warrants further investigation.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Hemorragia Cerebral/tratamiento farmacológico , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Animales , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Femenino , Interleucina-1beta/metabolismo , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
9.
FASEB J ; 31(1): 172-179, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27671228

RESUMEN

Parkinson's disease (PD) is characterized by a progressive loss of dopaminergic neurons with limited treatment options. Emerging evidence shows that FTY720 protects against neural injury via modulation of the sphingosine-1-phosphate 1 receptor (S1PR1). However, it remains unclear whether FTY720 could influence neurodegeneration in PD. Therefore, the present study was designed to determine the impact of fingolimod (FTY720), a sphingosine-1-phosphate receptor (S1PR) agonist, on 2 mouse models of PD. We found that FTY720 significantly reduced the deficit of motor function, diminished the loss of tyrosine hydroxylase-positive neurons in the substantia nigra, and attenuated the decrease of striatal dopamine and metabolite levels in mice receiving 6-hydroxydopamine (6-OHDA) or rotenone to simulate PD. An S1PR1-selective antagonist, W146, blocked the neuroprotective effects of FTY720. Of note, FTY720 retained the phosphorylation of ERK, together with a decreased expression of cleaved caspase-3 in mice treated with 6-OHDA or rotenone. In vitro studies revealed that FTY720 also attenuated 6-OHDA- or rotenone-induced toxicity in SH-SY5Y cells. These findings suggest the potential of S1PR modulation as a treatment for PD.-Zhao, P., Yang, X., Yang, L., Li, M., Wood, K., Liu, Q., Zhu, X. Neuroprotective effects of fingolimod in mouse models of Parkinson's disease.


Asunto(s)
Clorhidrato de Fingolimod/farmacología , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Animales , Línea Celular Tumoral , Regulación de la Expresión Génica/fisiología , Humanos , Masculino , Ratones , Neuroblastoma/tratamiento farmacológico , Oxidopamina/farmacología , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Rotenona/toxicidad , Receptores de Esfingosina-1-Fosfato
10.
FASEB J ; 31(8): 3278-3287, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28416580

RESUMEN

Intracerebral hemorrhage (ICH) is a devastating disease without effective treatment. After ICH, the immediate infiltration of leukocytes and activation of microglia are accompanied by a rapid up-regulation of the 18-kDa translocator protein (TSPO). TSPO ligands have shown anti-inflammatory and neuroprotective properties in models of CNS injury. In this study, we determined the impact of a TSPO ligand, etifoxine, on brain injury and inflammation in 2 mouse models of ICH. TSPO was up-regulated in Iba1+ cells from brains of patients with ICH and in CD11b+CD45int cells from mice subjected to collagenase-induced ICH. Etifoxine significantly reduced neurodeficits and perihematomal brain edema after ICH induction by injection of either autologous blood or collagenase. In collagenase-induced ICH mice, the protection of etifoxine was associated with reduced leukocyte infiltration into the brain and microglial production of IL-6 and TNF-α. Etifoxine improved blood-brain barrier integrity and diminished cell death. Notably, the protective effect of etifoxine was abolished in mice depleted of microglia by using a colony-stimulating factor 1 receptor inhibitor. These results indicate that the TSPO ligand etifoxine attenuates brain injury and inflammation after ICH. TSPO may be a viable therapeutic target that requires further investigations in ICH.-Li, M., Ren, H., Sheth, K. N., Shi, F.-D., Liu, Q. A TSPO ligand attenuates brain injury after intracerebral hemorrhage.


Asunto(s)
Ansiolíticos/farmacología , Hemorragia Cerebral/patología , Oxazinas/farmacología , Receptores de GABA/metabolismo , Animales , Edema Encefálico/tratamiento farmacológico , Hemorragia Cerebral/metabolismo , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica/fisiología , Ratones , Regulación hacia Arriba
11.
Muscle Nerve ; 57(5): 814-820, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29193204

RESUMEN

INTRODUCTION: Agrin is essential for the formation and maintenance of neuromuscular junctions (NMJs). NT-1654 is a C-terminal fragment of mouse neural agrin. In this study, we determined the effects of NT-1654 on the severity of experimental autoimmune myasthenia gravis (EAMG). METHODS: EAMG was induced in female Lewis rats by immunization with the Torpedo acetylcholine receptor (tAChR) and complete Freund's adjuvant (CFA). NT-1654 was dissolved in phosphate-buffered saline (PBS) and injected daily subcutaneously into tAChR immunized rats during the first 10 days after immunization, and then every other day for the following 20 days. RESULTS: We showed that NT-1654 attenuated clinical severity, effectively promoted the clustering of AChRs at NMJs, and alleviated the impairment of NMJ transmission and the reduction of muscle-specific kinase (MuSK) in EAMG rats. DISCUSSION: We demonstrated that NT-1654 attenuated clinical severity, effectively promoted the clustering of AChRs at NMJs, and alleviated the impairment of NMJ transmission and the reduction of muscle-specific kinase (MuSK) in EAMG rats. Muscle Nerve 57: 814-820, 2018.


Asunto(s)
Agrina/uso terapéutico , Inmunización/efectos adversos , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Miastenia Gravis Autoinmune Experimental/patología , Fragmentos de Péptidos/uso terapéutico , Potenciales de Acción/fisiología , Agrina/biosíntesis , Agrina/química , Animales , Autoanticuerpos/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Electromiografía , Femenino , Adyuvante de Freund/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/etiología , Atrofia Muscular/terapia , Proteínas del Tejido Nervioso/metabolismo , Neurofibromina 1/metabolismo , Unión Neuromuscular/patología , Fragmentos de Péptidos/biosíntesis , Fragmentos de Péptidos/química , Ratas , Ratas Endogámicas Lew , Receptores Colinérgicos/inmunología , Receptores Colinérgicos/metabolismo
12.
J Neuroinflammation ; 14(1): 151, 2017 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-28754131

RESUMEN

BACKGROUND: Cerebral ischemia is a leading cause of death and disability with limited treatment options. Although inflammatory and immune responses participate in ischemic brain injury, the molecular regulators of neuroinflammation after ischemia remain to be defined. Translocator protein 18 kDa (TSPO) mainly localized to the mitochondrial outer membrane is predominantly expressed in glia within the central nervous system during inflammatory conditions. This study investigated the effect of a TSPO agonist, etifoxine, on neuroinflammation and brain injury after ischemia/reperfusion. METHODS: We used a mouse model of middle cerebral artery occlusion (MCAO) to examine the therapeutic potential and mechanisms of neuroprotection by etifoxine. RESULTS: TSPO was upregulated in Iba1+ or CD11b+CD45int cells from mice subjected to MCAO and reperfusion. Etifoxine significantly attenuated neurodeficits and infarct volume after MCAO and reperfusion. The attenuation was pronounced in mice subjected to 30, 60, or 90 min MCAO. Etifoxine reduced production of pro-inflammatory factors in the ischemic brain. In addition, etifoxine treatment led to decreased expression of interleukin-1ß, interleukin-6, tumor necrosis factor-α, and inducible nitric oxide synthase by microglia. Notably, the benefit of etifoxine against brain infarction was ablated in mice depleted of microglia using a colony-stimulating factor 1 receptor inhibitor. CONCLUSIONS: These findings indicate that the TSPO agonist, etifoxine, reduces neuroinflammation and brain injury after ischemia/reperfusion. The therapeutic potential of targeting TSPO requires further investigations in ischemic stroke.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Oxazinas/uso terapéutico , Receptores de GABA/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Animales , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/etiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Esquema de Medicación , Citometría de Flujo , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Examen Neurológico , ARN Mensajero , Daño por Reperfusión/patología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
13.
Metab Brain Dis ; 32(5): 1395-1402, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28303450

RESUMEN

Traumatic brain injury (TBI) results in severe neurological impairments without effective treatments. Inflammation appears to be an important contributor to key pathogenic events such as secondary brain injury following TBI and therefore serves as a promising target for novel therapies. We have recently demonstrated the ability of a molecular construct comprised of the human leukocyte antigen (HLA)-DRα1 domain linked covalently to mouse (m)MOG-35-55 peptide (DRα1-MOG-35-55 construct) to reduce CNS inflammation and tissue injury in animal models of multiple sclerosis and ischemic stroke. The aim of the current study was to determine if DRα1-MOG-35-55 treatment of a fluid percussion injury (FPI) mouse model of TBI could reduce the lesion size and improve disease outcome measures. Neurodeficits, lesion size, and immune responses were determined to evaluate the therapeutic potential and mechanisms of neuroprotection induced by DRα1-MOG-35-55 treatment. The results demonstrated that daily injections of DRα1-MOG-35-55 given after FPI significantly reduced numbers of infiltrating CD74+ and CD86+ macrophages and increased numbers of CD206+ microglia in the brain concomitant with smaller lesion sizes and improvement in neurodeficits. Conversely, DRα1-MOG-35-55 treatment of TBI increased numbers of circulating CD11b+ monocytes and their expression of CD74 but had no detectable effect on cell numbers or marker expression in the spleen. These results demonstrate that DRα1-MOG-35-55 therapy can reduce CNS inflammation and significantly improve histological and clinical outcomes after TBI. Future studies will further examine the potential of DRα1-MOG-35-55 for treatment of TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Antígenos de Diferenciación de Linfocitos B/metabolismo , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/patología , Antígeno CD11b/metabolismo , Clonación Molecular , Antígenos de Histocompatibilidad Clase II/metabolismo , Recuento de Leucocitos , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Enfermedades del Sistema Nervioso/etiología , Fármacos Neuroprotectores/síntesis química , Proteínas Recombinantes de Fusión/síntesis química , Resultado del Tratamiento
14.
Immunol Cell Biol ; 92(2): 164-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24275856

RESUMEN

Glatiramer acetate (GA) is one of the first-line disease-modifying medications that have been approved for the treatment of multiple sclerosis via immune modulatory mechanisms. However, it remains unclear whether the immunomodulation effect of GA is central nervous system (CNS) antigen specific. Here, we explored the mechanism of action of GA by subcutaneously injecting GA in experimental autoimmune neuritis (EAN) rats, an animal model for Guillain-Barré syndrome (GBS). Clinical, electrophysiological and histological findings showed that neurological deficits, demyelination and axonal injury of sciatic nerves were all significantly attenuated in Lewis rats when GA was administered before immunization with peripheral nervous system antigen P0. Our results further demonstrated that GA treatment inhibited either P0 or myelin basic protein (MBP) (CNS antigen)-stimulated auto-immune T-cell proliferation in vitro. GA administrated at 10 days after induction of EAN when neurological sign became apparent also ameliorated the severity of disease, inhibited T-cell response to P0 and MBP and induced shift of proinflammatory and immune modulatory cytokines. Collectively, our findings suggested that GA attenuated neurological deficits in EAN rats and that the immune modulatory mechanisms of GA were not CNS antigen specific.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Síndrome de Guillain-Barré/tratamiento farmacológico , Inmunosupresores/farmacología , Neuritis Autoinmune Experimental/tratamiento farmacológico , Péptidos/farmacología , Linfocitos T/inmunología , Animales , Acetato de Glatiramer , Síndrome de Guillain-Barré/inmunología , Síndrome de Guillain-Barré/patología , Neuritis Autoinmune Experimental/inmunología , Neuritis Autoinmune Experimental/patología , Ratas , Ratas Endogámicas Lew , Linfocitos T/patología
15.
J Cereb Blood Flow Metab ; 44(3): 355-366, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-37933727

RESUMEN

Intracerebral hemorrhage (ICH) mobilizes circulating leukocytes that contribute to neuroinflammation and neural injury. However, little is known about the endogenous regulatory immune mechanisms to restrict neuroinflammation following ICH. We examined the role of group 2 innate lymphoid cells (ILC2) that are a specialized subset of innate immune modulators in a mouse model of ICH. We found accumulation of ILC2 in the brain following acute ICH and a concomitant increase of ILC2 within the peripheral lymph nodes. Depletion of ILC2 exacerbated neurodeficits and brain edema after ICH in male and female mice. This aggravated ICH injury was accompanied by augmented microglia activity and leukocyte infiltration. In contrast, expansion of ILC2 using IL-33 led to reduced ICH injury, microglia activity and leukocyte infiltration. Notably, elimination of microglia using a colony stimulating factor 1 receptor inhibitor diminished the exacerbation of ICH injury induced by depletion of ILC2. Brain-infiltrating ILC2 had upregulation of IL-13 after ICH. Results from in vitro assays revealed that ILC2 suppressed thrombin-induced inflammatory activity in microglia-like BV2 cells. Thus, our findings demonstrate that ILC2 suppress neuroinflammation and acute ICH injury.


Asunto(s)
Lesiones Encefálicas , Inmunidad Innata , Masculino , Femenino , Ratones , Animales , Enfermedades Neuroinflamatorias , Linfocitos/metabolismo , Hemorragia Cerebral/patología , Lesiones Encefálicas/metabolismo , Microglía/metabolismo
16.
Hum Mol Genet ; 20(7): 1339-52, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21239471

RESUMEN

Parthenogenetic embryonic stem cells (pESCs) have been generated in several mammalian species from parthenogenetic embryos that would otherwise die around mid-gestation. However, previous reports suggest that pESCs derived from in vivo ovulated (IVO) mature oocytes show limited pluripotency, as evidenced by low chimera production, high tissue preference and especially deficiency in germline competence, a critical test for genetic integrity and pluripotency of ESCs. Here, we report efficient generation of germline-competent pESC lines (named as IVM pESCs) from parthenogenetic embryos developed from immature oocytes of adult mouse ovaries following in vitro maturation (IVM) and artificial activation. In contrast, pESCs derived from IVO oocytes show defective germline competence, consistent with previous reports. Further, IVM pESCs resemble more ESCs from fertilized embryos (fESCs) than do IVO pESCs on genome-wide DNA methylation and global protein profiles. In addition, IVM pESCs express higher levels of Blimp1, Lin28 and Stella, relative to fESCs, and in their embryoid bodies following differentiation. This may indicate differences in differentiation potentially to the germline. The mechanisms for acquisition of pluripotency and germline competency of IVM pESCs from immature oocytes remain to be determined.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Células Madre Embrionarias , Oocitos , Ovario , Partenogénesis , Células Madre Pluripotentes , Animales , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Ratones , Oocitos/citología , Oocitos/metabolismo , Ovario/citología , Ovario/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
17.
Anal Methods ; 15(2): 171-178, 2023 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-36504026

RESUMEN

Surface chemistry of electrodes plays a critical role in the fields of electrochemistry and electric-field-assisted separation. In this study, making ingenious use of the ordered mesoporous structure of silica materials and the electrochemical stability of ionic liquids (ILs) when integrated with polyvinylpyrrolidone (PVP), the PVP-modified IL-mesoporous silica/platinum wire (Pt/PVP@meso-SiO2@IL) was fabricated to increase hydroelectric stability and avoid the problem of electrode polarization. The effect of different amounts of mesoporous silica material used to modify the surface of the Pt electrode was systematically investigated. As a result, we successfully obtained a highly ordered mesoporous Pt/PVP@meso-SiO2 material with smooth surface. Because pentyl triethylamine bis(trifluoromethylsulfonyl) imide exhibits a wide electrochemical window between -3 to 3 V, this IL was chosen to modify mesopores under vacuum. Even after repeatedly applying electric field on Pt/PVP@meso-SiO2@IL 100 times, this working electrode remained stable and showed high hydroelectric stability. After verifying the feasibility of this method, it was successfully applied in the electric-field-assisted separation of 2.0 and 3.0 µm polystyrene particles without any impediment from electrode polarization problems. This work provides a brand-new insight for resolving the problem of electrode polarization by developing a versatile tool for the electroseparation of micro-objects.

18.
Stroke Vasc Neurol ; 8(5): 424-434, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37072337

RESUMEN

BACKGROUND: Acute brain ischaemia elicits pronounced inflammation, which aggravates neural injury. However, the mechanisms governing the resolution of acute neuroinflammation remain poorly understood. In contrast to regulatory T and B cells, group 2 innate lymphoid cells (ILC2s) are immunoregulatory cells that can be swiftly mobilised without antigen presentation; whether and how these ILC2s participate in central nervous system inflammation following brain ischaemia is still unknown. METHODS: Leveraging brain tissues from patients who had an ischaemic stroke and a mouse model of focal ischaemia, we characterised the presence and cytokine release of brain-infiltrating ILC2s. The impact of ILC2s on neural injury was evaluated through antibody depletion and ILC2 adoptive transfer experiments. Using Rag2-/-γc-/- mice receiving passive transfer of IL-4-/- ILC2s, we further assessed the contribution of interleukin (IL)-4, produced by ILC2s, in ischaemic brain injury. RESULTS: We demonstrate that ILC2s accumulate in the areas surrounding the infarct in brain tissues of patients with cerebral ischaemia, as well as in mice subjected to focal cerebral ischaemia. Oligodendrocytes were a major source of IL-33, which contributed to ILC2s mobilisation. Adoptive transfer and expansion of ILC2s reduced brain infarction. Importantly, brain-infiltrating ILC2s reduced the magnitude of stroke injury severity through the production of IL-4. CONCLUSIONS: Our findings revealed that brain ischaemia mobilises ILC2s to curb neuroinflammation and brain injury, expanding the current understanding of inflammatory networks following stroke.


Asunto(s)
Lesiones Encefálicas , Isquemia Encefálica , Accidente Cerebrovascular , Humanos , Ratones , Animales , Inmunidad Innata , Interleucina-4 , Linfocitos , Enfermedades Neuroinflamatorias , Inflamación
19.
Int J Pharm ; 611: 121278, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34774693

RESUMEN

Oral delivery of nanoparticles possesses many advantages for delivery of active pharmaceutical ingredients (APIs) to the gastrointestinal tract. However, the poor physical stability of nanoparticles in liquid state is often a challenge. Removing water from the nanosuspensions and transforming the nanoparticles into solid particulate matter in the form of, e.g., tablets could be a potential approach to increase the stability of nanoparticles. The aim of this study was to transform nanoparticles into compacts and to investigate the redispersion of nanoparticles from compacts as well as the dissolution behavior of these compacts. DL-lactide-co-glycolide copolymer (PLGA) nanoparticles and celecoxib (CLX) nanoparticles were used as two model nanoparticle systems and fabricated into nano-embedded microparticles (NEMs) and subsequently compressed into compacts. The compacts were evaluated with respect to the redispersibility of the nanoparticles, as well as the dissolution characteristics of CLX. The results showed that the NEMs could be readily compressed into compacts with sufficient mechanical strength. The size of the redispersed PLGA nanoparticles from the compacts using 2-hydroxypropyl-ß-cyclodextrin (HPßCD) as stabilizer was comparable to the original nanoparticles. In contrast, the redispersibility of CLX nanoparticles from the compacts was not as effective as for the PLGA nanoparticles evidenced by a significant increase in the size and polydispersity index (PDI) of the redispersed nanoparticles. Nonetheless, an obvious enhancement in dissolution rate of CLX was observed from the compacts with CLX nanoparticles. It is concluded that transforming polymeric nanoparticles into compacts via NEMs provides stabilization and allows redispersion into original nanoparticles. Despite the reduced redispersibility, compacts loaded with nanoparticles exhibited improved dissolution rate compared with the crystalline drug. Loading of nanoparticles into compacts is a promising approach to overcome the poor stability of nanoparticle within oral drug delivery of nanoparticles.


Asunto(s)
Nanopartículas , 2-Hidroxipropil-beta-Ciclodextrina , Celecoxib , Sistemas de Liberación de Medicamentos , Polímeros
20.
Cell Death Dis ; 13(1): 33, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013119

RESUMEN

Aged microglia display augmented inflammatory activity after neural injury. Although aging is a risk factor for poor outcome after brain insults, the precise impact of aging-related alterations in microglia on neural injury remains poorly understood. Microglia can be eliminated via pharmacological inhibition of the colony-stimulating factor 1 receptor (CSF1R). Upon withdrawal of CSF1R inhibitors, microglia rapidly repopulate the entire brain, leading to replacement of the microglial compartment. In this study, we investigated the impact of microglial replacement in the aged brain on neural injury using a mouse model of intracerebral hemorrhage (ICH) induced by collagenase injection. We found that replacement of microglia in the aged brain reduced neurological deficits and brain edema after ICH. Microglial replacement-induced attenuation of ICH injury was accompanied with alleviated blood-brain barrier disruption and leukocyte infiltration. Notably, newly repopulated microglia had reduced expression of IL-1ß, TNF-α and CD86, and upregulation of CD206 in response to ICH. Our findings suggest that replacement of microglia in the aged brain restricts neuroinflammation and brain injury following ICH.


Asunto(s)
Envejecimiento/efectos de los fármacos , Encéfalo/efectos de los fármacos , Hemorragia Cerebral/tratamiento farmacológico , Microglía/efectos de los fármacos , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Envejecimiento/patología , Aminopiridinas/administración & dosificación , Aminopiridinas/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Encéfalo/inmunología , Encéfalo/patología , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/etiología , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/patología , Muerte Celular/efectos de los fármacos , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/inmunología , Hemorragia Cerebral/patología , Quimiotaxis de Leucocito/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones , Microglía/inmunología , Microglía/patología , Enfermedades Neuroinflamatorias/etiología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/patología , Pirroles/administración & dosificación , Pirroles/farmacología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA