Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
ACS Macro Lett ; 12(12): 1698-1704, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38039381

RESUMEN

Shape memory polymers (SMPs) are a type of smart shape-shifting material that can respond to various stimuli. Their shape recovery pathway is determined by the internal stress stored in the temporary shapes. Thus, manipulating the internal stress is key to the potential applications of SMPs. This is commonly achieved by the types of deformation forces applied during the programming stage. In contrast, we present here a digital laser direct writing method to selectively induce thermal relaxation of internal stress stored in the two-dimensional (2D) shape of a thermoplastic SMP. The internal stress field, while invisible under natural light, can be visualized under polarized light. Consequently, the digital stress pattern can be used for anticounterfeiting. In addition, further uniform heating induces the release of the programmed internal stress within the 2D film. This triggers its transformation into a three-dimensional (3D) shape, enabling 4D printing. The simplicity and versatility of our approach in manipulating internal stress and shape-shifting make it attractive for potential applications.

2.
Nat Commun ; 14(1): 1313, 2023 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-36899070

RESUMEN

Delicate geometries and suitable mechanical properties are essential for device applications of polymer materials. 3D printing offers unprecedented versatility, but the geometries and mechanical properties are typically fixed after printing. Here, we report a 3D photo-printable dynamic covalent network that can undergo two independently controllable bond exchange reactions, allowing reprogramming the geometry and mechanical properties after printing. Specifically, the network is designed to contain hindered urea bonds and pendant hydroxyl groups. The homolytic exchange between hindered urea bonds allows reconfiguring the printed shape without affecting the network topology and mechanical properties. Under different conditions, the hindered urea bonds are transformed into urethane bonds via exchange reactions with hydroxyl groups, which permits tailoring of the mechanical properties. The freedom to reprogram the shape and properties in an on-demand fashion offers the opportunity to produce multiple 3D printed products from one single printing step.

3.
Adv Mater ; 35(16): e2209824, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36681865

RESUMEN

Living creatures possess complex geometries, exceptional adaptability, and continuous growing and regenerating characteristics, which are difficult for synthetic materials to imitate simultaneously. A living polymer network with these features is reported. The polymer can be digitally printed into arbitrary 3D shapes and subsequently undergoes growth via living polymerization of a monomer as the nutrient. This leads to macroscopic dimensional growth and transforms the printed amorphous network into a crystallizable network, resulting in geometric adaptability via a shape-memory mechanism. By controlling the localized growth, an initial homogeneous structure can be converted into a geometrically different heterogeneous structure composed of materials with different properties (crystallization and mechanical properties). After growth, the original network can be chemically regenerated for regrowth. With this regenerative living 4D printing, one 3D-printed seed template can be turned into different derivatives with distinct geometries and mechanical properties when repeated regeneration is conducted in different localized regions and the degree of regrowth is varied.

4.
J Neuroinflammation ; 19(1): 201, 2022 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-35933343

RESUMEN

BACKGROUND: Histone deacetylases (HDACs) are believed to exacerbate traumatic brain injury (TBI) based on studies using pan-HDAC inhibitors. However, the HDAC isoform responsible for the detrimental effects and the cell types involved remain unknown, which may hinder the development of specific targeting strategies that boost therapeutic efficacy while minimizing side effects. Microglia are important mediators of post-TBI neuroinflammation and critically impact TBI outcome. HDAC3 was reported to be essential to the inflammatory program of in vitro cultured macrophages, but its role in microglia and in the post-TBI brain has not been investigated in vivo. METHODS: We generated HDAC3LoxP mice and crossed them with CX3CR1CreER mice, enabling in vivo conditional deletion of HDAC3. Microglia-specific HDAC3 knockout (HDAC3 miKO) was induced in CX3CR1CreER:HDAC3LoxP mice with 5 days of tamoxifen treatment followed by a 30-day development interval. The effects of HDAC3 miKO on microglial phenotype and neuroinflammation were examined 3-5 days after TBI induced by controlled cortical impact. Neurological deficits and the integrity of white matter were assessed for 6 weeks after TBI by neurobehavioral tests, immunohistochemistry, electron microscopy, and electrophysiology. RESULTS: HDAC3 miKO mice harbored specific deletion of HDAC3 in microglia but not in peripheral monocytes. HDAC3 miKO reduced the number of microglia by 26%, but did not alter the inflammation level in the homeostatic brain. After TBI, proinflammatory microglial responses and brain inflammation were markedly alleviated by HDAC3 miKO, whereas the infiltration of blood immune cells was unchanged, suggesting a primary effect of HDAC3 miKO on modulating microglial phenotype. Importantly, HDAC3 miKO was sufficient to facilitate functional recovery for 6 weeks after TBI. TBI-induced injury to axons and myelin was ameliorated, and signal conduction by white matter fiber tracts was significantly enhanced in HDAC3 miKO mice. CONCLUSION: Using a novel microglia-specific conditional knockout mouse model, we delineated for the first time the role of microglial HDAC3 after TBI in vivo. HDAC3 miKO not only reduced proinflammatory microglial responses, but also elicited long-lasting improvement of white matter integrity and functional recovery after TBI. Microglial HDAC3 is therefore a promising therapeutic target to improve long-term outcomes after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Histona Desacetilasas , Sustancia Blanca , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Histona Desacetilasas/metabolismo , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Sustancia Blanca/metabolismo
5.
J Cereb Blood Flow Metab ; 41(3): 511-529, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32757740

RESUMEN

Long-term neurological recovery after severe traumatic brain injury (TBI) is strongly linked to the repair and functional restoration of injured white matter. Emerging evidence suggests that the anti-inflammatory cytokine interleukin-4 (IL-4) plays an important role in promoting white matter integrity after cerebral ischemic injury. Here, we report that delayed intranasal delivery of nanoparticle-packed IL-4 boosted sensorimotor neurological recovery in a murine model of controlled cortical impact, as assessed by a battery of neurobehavioral tests for up to five weeks. Post-injury IL-4 treatment failed to reduce macroscopic brain lesions after TBI, but preserved the structural and functional integrity of white matter, at least in part through oligodendrogenesis. IL-4 directly facilitated the differentiation of oligodendrocyte progenitor cells (OPCs) into mature myelin-producing oligodendrocytes in primary cultures, an effect that was attenuated by selective PPARγ inhibition. IL-4 treatment after TBI in vivo also failed to stimulate oligodendrogenesis or improve white matter integrity in OPC-specific PPARγ conditional knockout (cKO) mice. Accordingly, IL-4-afforded improvements in sensorimotor neurological recovery after TBI were markedly impaired in the PPARγ cKO mice compared to wildtype controls. These results support IL-4 as a potential novel neurorestorative therapy to improve white matter functionality and mitigate the long-term neurological consequences of TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Interleucina-4/uso terapéutico , Oligodendroglía/metabolismo , PPAR gamma/metabolismo , Sustancia Blanca/patología , Administración Intranasal , Animales , Conducta Animal/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Interleucina-4/química , Interleucina-4/farmacología , Liposomas/química , Masculino , Ratones , Ratones Transgénicos , Vaina de Mielina/metabolismo , Oligodendroglía/citología , PPAR gamma/deficiencia , PPAR gamma/genética , Recuperación de la Función
6.
J Cereb Blood Flow Metab ; 40(1_suppl): S81-S97, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32065074

RESUMEN

Age-dependent alterations in microglia behavior have been implicated in neurodegeneration and CNS injuries. Here, we compared the transcriptional profiles of young versus aged microglia during stroke recovery. CD45intermediateCD11b+ microglia were FACS-isolated from the brains of young (10-week-old) and aged (18-month-old) male mice with sham operation or 14 days after distal middle cerebral artery occlusion and subjected to RNA-sequencing analysis. Functional groups enriched in young microglia are indicative of upregulation in cell movement, cell interactions, inflammatory responses and angiogenesis, while aged microglia exhibited a reduction or no change in these features. We confirmed reduced chemoattractive capacities of aged microglia toward ischemic brain tissue in organotypic slide co-cultures, and delayed accumulation of aged microglia around dead neurons injected into the striatum in vivo. In addition, aging is associated with an overall failure to increase the expression of microglial genes involved in cell-cell interactions, such as CXCL10. Finally, impaired upregulation of pro-angiogenic genes in aged microglia was associated with a decline in neovascularization in aged mice compared to young mice after distal middle cerebral artery occlusion. This study provides a new resource to understand the mechanisms underlying microglial alterations in the aged brain milieu and sheds light on new strategies to improve microglial functions in aged stroke victims.


Asunto(s)
Microglía/metabolismo , Accidente Cerebrovascular/genética , Animales , Modelos Animales de Enfermedad , Masculino , Ratones
7.
PLoS Biol ; 17(6): e3000330, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31226122

RESUMEN

The repair of white matter damage is of paramount importance for functional recovery after brain injuries. Here, we report that interleukin-4 (IL-4) promotes oligodendrocyte regeneration and remyelination. IL-4 receptor expression was detected in a variety of glial cells after ischemic brain injury, including oligodendrocyte lineage cells. IL-4 deficiency in knockout mice resulted in greater deterioration of white matter over 14 d after stroke. Consistent with these findings, intranasal delivery of IL-4 nanoparticles after stroke improved white matter integrity and attenuated long-term sensorimotor and cognitive deficits in wild-type mice, as revealed by histological immunostaining, electron microscopy, diffusion tensor imaging, and electrophysiology. The selective effect of IL-4 on remyelination was verified in an ex vivo organotypic model of demyelination. By leveraging primary oligodendrocyte progenitor cells (OPCs), microglia-depleted mice, and conditional OPC-specific peroxisome proliferator-activated receptor gamma (PPARγ) knockout mice, we discovered a direct salutary effect of IL-4 on oligodendrocyte differentiation that was mediated by the PPARγ axis. Our findings reveal a new regenerative role of IL-4 in the central nervous system (CNS), which lies beyond its known immunoregulatory functions on microglia/macrophages or peripheral lymphocytes. Therefore, intranasal IL-4 delivery may represent a novel therapeutic strategy to improve white matter integrity in stroke and other brain injuries.


Asunto(s)
Interleucina-4/metabolismo , Oligodendroglía/metabolismo , PPAR gamma/metabolismo , Animales , Lesiones Encefálicas , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatología , Diferenciación Celular/fisiología , Enfermedades Desmielinizantes/metabolismo , Interleucina-4/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Vaina de Mielina/metabolismo , Regeneración Nerviosa , Neurogénesis , Oligodendroglía/fisiología , PPAR gamma/fisiología , Recuperación de la Función , Remielinización/fisiología , Transducción de Señal , Accidente Cerebrovascular , Sustancia Blanca
8.
CNS Neurosci Ther ; 25(9): 1018-1029, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31140740

RESUMEN

AIMS: This study determines whether assessment with compound action potentials (CAPs) can distinguish two different forms of cerebral white matter injury at the functional levels. METHODS: A pure demyelination model was induced in C57/BL6 adult mice by dietary supplementation of cuprizone (0.2%) for 6 weeks. Callosal L-N5-(1-Iminoethyl) ornithine (L-NIO) hydrochloride (27 mg/mL) was injected into the corpus callosum (CC) to induce a focal white matter stroke (WMS), resulting in both demyelination and axonal injury. White matter integrity was assessed by performing CAP recording, electron microscopy, and immunohistological and luxol fast blue (LFB) staining. RESULTS: Immunohistological and electron microscopic analyses confirmed the induction of robust demyelination in CC with cuprizone, and mixed demyelination and axonal damage with L-NIO. Electrophysiologically, cuprizone-induced demyelination significantly reduced the amplitude of negative peak 1 (N1), but increased the amplitude of negative peak 2 (N2), of the CAPs compared to the sham controls. However, cuprizone did not affect the axonal conduction velocity. In contrast, the amplitude and area of both N1 and N2 along with N1 axonal conduction velocity were dramatically decreased in L-NIO-induced WMS. CONCLUSIONS: Concertedly, parameters of the CAPs offer a novel functional assessment strategy for cerebral white matter injury in rodent models.


Asunto(s)
Potenciales de Acción/fisiología , Axones/fisiología , Cuerpo Calloso/fisiopatología , Enfermedades Desmielinizantes/fisiopatología , Conducción Nerviosa/fisiología , Sustancia Blanca/fisiopatología , Animales , Axones/ultraestructura , Cuerpo Calloso/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Sustancia Blanca/ultraestructura
9.
Proc Natl Acad Sci U S A ; 116(18): 9115-9124, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-30996120

RESUMEN

Emerging evidence suggests that tissue plasminogen activator (tPA), currently the only FDA-approved medication for ischemic stroke, exerts important biological actions on the CNS besides its well-known thrombolytic effect. In this study, we investigated the role of tPA on primary neurons in culture and on brain recovery and plasticity after ischemic stroke in mice. Treatment with recombinant tPA stimulated axonal growth in culture, an effect independent of its protease activity and achieved through epidermal growth factor receptor (EGFR) signaling. After permanent focal cerebral ischemia, tPA knockout mice developed more severe sensorimotor and cognitive deficits and greater axonal and myelin injury than wild-type mice, suggesting that endogenously expressed tPA promotes long-term neurological recovery after stroke. In tPA knockout mice, intranasal administration of recombinant tPA protein 6 hours poststroke and 7 more times at 2 d intervals mitigated white matter injury, improved axonal conduction, and enhanced neurological recovery. Consistent with the proaxonal growth effects observed in vitro, exogenous tPA delivery increased poststroke axonal sprouting of corticobulbar and corticospinal tracts, which might have contributed to restoration of neurological functions. Notably, recombinant mutant tPA-S478A lacking protease activity (but retaining the EGF-like domain) was as effective as wild-type tPA in rescuing neurological functions in tPA knockout stroke mice. These findings demonstrate that tPA improves long-term functional outcomes in a clinically relevant stroke model, likely by promoting brain plasticity through EGFR signaling. Therefore, treatment with the protease-dead recombinant tPA-S478A holds particular promise as a neurorestorative therapy, as the risk for triggering intracranial hemorrhage is eliminated and tPA-S478A can be delivered intranasally hours after stroke.


Asunto(s)
Plasticidad Neuronal/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Activador de Tejido Plasminógeno/uso terapéutico , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Encéfalo/metabolismo , Lesiones Encefálicas/tratamiento farmacológico , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Infarto Cerebral , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Neurogénesis/efectos de los fármacos , Neuronas/metabolismo , Recuperación de la Función
10.
J Cereb Blood Flow Metab ; 39(7): 1215-1231, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30334662

RESUMEN

Ischemic injury can be alleviated by the judicious use of hypothermia. However, the optimal regimens and the temporal kinetics of post-stroke neurovascular responses to hypothermic intervention have not been systematically studied. These gaps slow the clinical translation of hypothermia as an anti-stroke therapy. Here, we characterized the effects of transient selective brain hypothermia (TSBH) from the hyperacute to chronic stages of focal ischemia/reperfusion brain injury induced by transient middle cerebral artery occlusion in mice. A simple cooling device was used to induce TSBH during cerebral ischemia. This treatment reduced mortality from 31.8% to 0% and improved neurological outcomes for at least 35 days post-injury. TSBH mitigated blood-brain barrier leakage during the hyperacute and acute injury stages (1-23 h post-reperfusion). This early protection of the blood-brain barrier was associated with anti-inflammatory phenotypic polarization of microglia/macrophages, reduced production of pro-inflammatory cytokines, and less brain infiltration of neutrophils and macrophages during the subacute injury stage (three days post-reperfusion). TSBH elicited enduring protective effects on both grey and white matter for at least 35 days post-injury and preserved the long-term electrophysiological function of fiber tracts. In conclusion, TSBH ameliorates ischemia/reperfusion injury in the neurovascular unit from hyperacute to chronic injury stages after experimental stroke.


Asunto(s)
Encéfalo/fisiopatología , Hipotermia Inducida/métodos , Daño por Reperfusión/prevención & control , Daño por Reperfusión/fisiopatología , Animales , Barrera Hematoencefálica/fisiopatología , Encéfalo/irrigación sanguínea , Citocinas/metabolismo , Infarto de la Arteria Cerebral Media , Inflamación/prevención & control , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología , Daño por Reperfusión/etiología , Sustancia Blanca/fisiopatología
11.
Proc Natl Acad Sci U S A ; 115(39): E9230-E9238, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30201709

RESUMEN

Recombinant tissue plasminogen activator (tPA) is a Food and Drug Administration-approved thrombolytic treatment for ischemic stroke. tPA is also naturally expressed in glial and neuronal cells of the brain, where it promotes axon outgrowth and synaptic plasticity. However, there are conflicting reports of harmful versus neuroprotective effects of tPA in acute brain injury models. Furthermore, its impact on white matter integrity in preclinical traumatic brain injury (TBI) has not been thoroughly explored, although white matter disruption is a better predictor of long-term clinical outcomes than focal lesion volumes. Here we show that the absence of endogenous tPA in knockout mice impedes long-term recovery of white matter and neurological function after TBI. tPA-knockout mice exhibited greater asymmetries in forepaw use, poorer sensorimotor balance and coordination, and inferior spatial learning and memory up to 35 d after TBI. White matter damage was also more prominent in tPA knockouts, as shown by diffusion tensor imaging, histological criteria, and electrophysiological assessments of axon conduction properties. Replenishment of tPA through intranasal application of the recombinant protein in tPA-knockout mice enhanced neurological function, the structural and functional integrity of white matter, and postinjury compensatory sprouting in corticofugal projections. tPA also promoted neurite outgrowth in vitro, partly through the epidermal growth factor receptor. Both endogenous and exogenous tPA protected against white matter injury after TBI without increasing intracerebral hemorrhage volumes. These results unveil a previously unappreciated role for tPA in the protection and/or repair of white matter and long-term functional recovery after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Activador de Tejido Plasminógeno/uso terapéutico , Sustancia Blanca/efectos de los fármacos , Animales , Lesiones Traumáticas del Encéfalo/patología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibras Nerviosas/efectos de los fármacos , Red Nerviosa/efectos de los fármacos , Proteínas Recombinantes , Sustancia Blanca/patología
12.
J Cereb Blood Flow Metab ; 38(12): 2073-2091, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30191760

RESUMEN

Recent advances in stroke reperfusion therapies have led to remarkable improvement in clinical outcomes, but many patients remain severely disabled, due in part to the lack of effective neuroprotective strategies. In this review, we show that 95% of published preclinical studies on "neuroprotectants" (1990-2018) reported positive outcomes in animal models of ischemic stroke, while none translated to successful Phase III trials. There are many complex reasons for this failure in translational research, including that the majority of clinical trials did not test early delivery of neuroprotectants in combination with successful reperfusion. In contrast to the clinical trials, >80% of recent preclinical studies examined the neuroprotectant in animal models of transient ischemia with complete reperfusion. Furthermore, only a small fraction of preclinical studies included long-term functional assessments, aged animals of both genders, and models with stroke comorbidities. Recent clinical trials demonstrate that 70%-80% of patients treated with endovascular thrombectomy achieve successful reperfusion. These successes revive the opportunity to retest previously failed approaches, including cocktail drugs that target multiple injury phases and different cell types. It is our hope that neurovascular protectants can be retested in future stroke research studies with specific criteria outlined in this review to increase translational successes.


Asunto(s)
Revascularización Cerebral/métodos , Fármacos Neuroprotectores/farmacología , Reperfusión/métodos , Accidente Cerebrovascular/terapia , Animales , Humanos
13.
Neurobiol Dis ; 91: 37-46, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26921472

RESUMEN

Omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been shown to protect the neonatal brain against hypoxic/ischemic (H/I) injury. However, the mechanism of n-3 PUFA-afforded neuroprotection is not well understood. One major determinant of H/I vulnerability is the permeability of the blood-brain barrier (BBB). Therefore, we examined the effects of n-3 PUFAs on BBB integrity after neonatal H/I. Female rats were fed a diet with or without n-3 PUFA enrichment from day 2 of pregnancy to 14days after parturition. H/I was introduced in 7day-old offspring. We observed relatively rapid BBB penetration of the small molecule cadaverine (640Da) at 4h post-H/I and a delayed penetration of larger dextrans (3kD-40kD) 24-48h after injury. Surprisingly, the neonatal BBB was impermeable to Evans Blue or 70kD dextran leakage for up to 48h post-H/I, despite evidence of IgG extravasation at this time. As expected, n-3 PUFAs ameliorated H/I-induced BBB damage, as shown by reductions in tracer efflux and IgG extravasation, preservation of BBB ultrastructure, and enhanced tight junction protein expression. Furthermore, n-3 PUFAs prevented the elevation in matrix metalloproteinase (MMP) activity in the brain and blood after H/I. Thus, n-3 PUFAs may protect neonates against BBB damage by blunting MMPs activation after H/I.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Ácidos Grasos Omega-3/farmacología , Hipoxia-Isquemia Encefálica/metabolismo , Animales , Animales Recién Nacidos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Ácidos Grasos Omega-3/metabolismo , Femenino , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA