Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
Adv Ther (Weinh) ; 7(2)2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-39071184

RESUMEN

Multiple therapies have been studied to ameliorate the neuroinhibitory cues present after traumatic injury to the central nervous system. Two previous in vitro studies have demonstrated the efficacy of the FDA-approved cardiovascular therapeutic, protamine (PRM), to overcome neuroinhibitory cues presented by chondroitin sulfates; however, the effect of a wide range of PRM concentrations on neuronal and glial cells has not been evaluated. In this study, we investigate the therapeutic efficacy of PRM with primary cortical neurons, hippocampal neurons, mixed glial cultures, and astrocyte cultures. We show the threshold for PRM toxicity to be at or above 10 µg/ml depending on the cell population, that 10 µg/ml PRM enables neurons to overcome the inhibitory cues presented by chondroitin sulfate type A, and that soluble PRM allows neurons to more effectively overcome inhibition compared to a PRM coating. We also assessed changes in gene expression of reactive astrocytes with soluble PRM and determined that PRM does not increase their neurotoxic phenotype and that PRM may reduce brevican production and serpin transcription in cortical and spinal cord astrocytes. This is the first study to thoroughly assess the toxicity threshold of PRM with neural cells and study astrocyte response after acute exposure to PRM in vitro.

2.
J Neural Eng ; 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39029499

RESUMEN

OBJECTIVE: Macrophages and astrocytes play a crucial role in the aftermath of a traumatic spinal cord injury (SCI). Infiltrating macrophages adopt a pro-inflammatory phenotype while resident astrocytes adopt a neurotoxic phenotype at the injury site, both of which contribute to neuronal death and inhibit axonal regeneration. The cytokine interleukin-4 (IL-4) has shown significant promise in preclinical models of SCI by alleviating the macrophage-mediated inflammation and promoting functional recovery. However, its effect on neurotoxic reactive astrocytes remains to be elucidated, which we explored in this study. We also studied the beneficial effects of a sustained release of IL-4 from an injectable biomaterial compared to bolus administration of IL-4. APPROACH: We fabricated a heparin-based coacervate capable of anchoring and releasing bioactive IL-4 and tested its efficacy in vitro and in vivo. MAIN RESULTS: We show that IL-4 coacervate is biocompatible and drives a robust anti-inflammatory macrophage phenotype in culture. We also show that IL-4 and IL-4 coacervate can alleviate the reactive neurotoxic phenotype of astrocytes in culture. Finally, using a murine model of contusion SCI, we show that IL-4 and IL-4 coacervate, injected intraspinally 2 days post-injury, can reduce macrophage-mediated inflammation, and alleviate neurotoxic astrocyte phenotype, acutely and chronically, while also promoting neuroprotection with significant improvements in hindlimb locomotor recovery. We observed that IL-4 coacervate can promote a more robust regenerative macrophage phenotype in vitro, as well as match its efficacy in vivo, compared to bolus IL-4. SIGNIFICANCE: Our work shows the promise of coacervate as a great choice for local and prolonged delivery of cytokines like IL-4. We support this by showing that the coacervate can release bioactive IL-4, which acts on macrophages and astrocytes to promote a pro-regenerative environment following a spinal cord injury leading to robust neuroprotective and functional outcomes.

3.
Adv Biol (Weinh) ; : e2300531, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38935534

RESUMEN

A spinal cord injury (SCI) compresses the spinal cord, killing neurons and glia at the injury site and resulting in prolonged inflammation and scarring that prevents regeneration. Astrocytes, the main glia in the spinal cord, become reactive following SCI and contribute to adverse outcomes. The anti-inflammatory cytokine transforming growth factor beta 3 (TGFß3) has been shown to mitigate astrocyte reactivity; however, the effects of prolonged TGFß3 exposure on reactive astrocyte phenotype have not yet been explored. This study investigates whether magnetic core-shell electrospun fibers can be used to alter the release rate of TGFß3 using externally applied magnetic fields, with the eventual application of tailored drug delivery based on SCI severity. Magnetic core-shell fibers are fabricated by incorporating superparamagnetic iron oxide nanoparticles (SPIONs) into the shell and TGFß3 into the core solution for coaxial electrospinning. Magnetic field stimulation increased the release rate of TGFß3 from the fibers by 25% over 7 days and released TGFß3 reduced gene expression of key astrocyte reactivity markers by at least twofold. This is the first study to magnetically deliver bioactive proteins from magnetic fibers and to assess the effect of sustained release of TGFß3 on reactive astrocyte phenotype.

4.
ACS Biomater Sci Eng ; 10(1): 482-496, 2024 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-38109315

RESUMEN

Clinical use of polymeric scaffolds for tissue engineering often suffers from their inability to promote strong cellular interactions. Functionalization with biomolecules may improve outcomes; however, current functionalization approaches using covalent chemistry or physical adsorption can lead to loss of biomolecule bioactivity. Here, we demonstrate a novel bottom-up approach for enhancing the bioactivity of poly(l-lactic acid) electrospun scaffolds though interfacial coassembly of protein payloads with silk fibroin into nanothin coatings. In our approach, protein payloads are first added into an aqueous solution with Bombyx mori-derived silk fibroin. Phosphate anions are then added to trigger coassembly of the payload and silk fibroin, as well as noncovalent formation of a payload-silk fibroin coating at poly(l-lactic) acid fiber surfaces. Importantly, the coassembly process results in homogeneous distribution of protein payloads, with the loading quantity depending on payload concentration in solution and coating time. This coassembly process yields greater loading capacity than physical adsorption methods, and the payloads can be released over time in physiologically relevant conditions. We also demonstrate that the coating coassembly process can incorporate nerve growth factor and that coassembled coatings lead to significantly more neurite extension than loading via adsorption in a rat dorsal root ganglia explant culture model.


Asunto(s)
Bombyx , Fibroínas , Ratas , Animales , Seda/química , Fibroínas/farmacología , Ingeniería de Tejidos/métodos , Regeneración Nerviosa
5.
Front Cell Neurosci ; 17: 1266019, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37941607

RESUMEN

Central nervous system (CNS) glia, including astrocytes, microglia, and oligodendrocytes, play prominent roles in traumatic injury and degenerative disorders. Due to their importance, active pharmaceutical ingredients (APIs) are being developed to modulate CNS glia in order to improve outcomes in traumatic injury and disease. While many of these APIs show promise in vitro, the majority of APIs that are systemically delivered show little penetration through the blood-brain barrier (BBB) or blood-spinal cord barrier (BSCB) and into the CNS, rendering them ineffective. Novel nanomaterials are being developed to deliver APIs into the CNS to modulate glial responses and improve outcomes in injury and disease. Nanomaterials are attractive options as therapies for central nervous system protection and repair in degenerative disorders and traumatic injury due to their intrinsic capabilities in API delivery. Nanomaterials can improve API accumulation in the CNS by increasing permeation through the BBB of systemically delivered APIs, extending the timeline of API release, and interacting biophysically with CNS cell populations due to their mechanical properties and nanoscale architectures. In this review, we present the recent advances in the fields of both locally implanted nanomaterials and systemically administered nanoparticles developed for the delivery of APIs to the CNS that modulate glial activity as a strategy to improve outcomes in traumatic injury and disease. We identify current research gaps and discuss potential developments in the field that will continue to translate the use of glia-targeting nanomaterials to the clinic.

6.
ACS Appl Bio Mater ; 6(2): 806-818, 2023 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-36749645

RESUMEN

Intracortical microelectrodes are used with brain-computer interfaces to restore lost limb function following nervous system injury. While promising, recording ability of intracortical microelectrodes diminishes over time due, in part, to neuroinflammation. As curcumin has demonstrated neuroprotection through anti-inflammatory activity, we fabricated a 300 nm-thick intracortical microelectrode coating consisting of a polyurethane copolymer of curcumin and polyethylene glycol (PEG), denoted as poly(curcumin-PEG1000 carbamate) (PCPC). The uniform PCPC coating reduced silicon wafer hardness by two orders of magnitude and readily absorbed water within minutes, demonstrating that the coating is soft and hydrophilic in nature. Using an in vitro release model, curcumin eluted from the PCPC coating into the supernatant over 1 week; the majority of the coating was intact after an 8-week incubation in buffer, demonstrating potential for longer term curcumin release and softness. Assessing the efficacy of PCPC within a rat intracortical microelectrode model in vivo, there were no significant differences in tissue inflammation, scarring, neuron viability, and myelin damage between the uncoated and PCPC-coated probes. As the first study to implant nonfunctional probes with a polymerized curcumin coating, we have demonstrated the biocompatibility of a PCPC coating and presented a starting point in the design of poly(pro-curcumin) polymers as coating materials for intracortical electrodes.


Asunto(s)
Curcumina , Ratas , Animales , Microelectrodos , Curcumina/farmacología , Electrodos Implantados , Neuronas , Polímeros
7.
Bioengineering (Basel) ; 10(2)2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36829756

RESUMEN

Curcumin, a bioactive phenol derived from turmeric, is an antioxidant, anti-inflammatory, and antibacterial molecule. Although curcumin exhibits beneficial effects in its innate form, it is highly hydrophobic, which leads to poor water solubility and, consequently, low bioavailability. The lack of bioavailability limits curcumin's effectiveness as a treatment and restricts its use in clinical applications. Furthermore, to achieve beneficial, clinically relevant results, high doses of curcumin are required for systemic administration. Many researchers have utilized biomaterial carriers, including electrospun fibers, nanoparticles, hydrogels, and composite scaffolds, to overcome curcumin's principle therapeutic limitation of low bioavailability. By using biomaterials to deliver curcumin directly to injury sites, researchers have harnessed the beneficial natural properties of curcumin while providing scaffolding to support tissue regeneration. This review will provide an in-depth overview of the literature that utilizes biomaterial delivery of curcumin for tissue regeneration in injury and disease models.

8.
Acta Biomater ; 155: 370-385, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36423820

RESUMEN

Aligned electrospun fibers provide topographical cues and local therapeutic delivery to facilitate robust peripheral nerve regeneration. mRNA delivery enables transient expression of desired proteins that promote axonal regeneration. However, no prior work delivers mRNA from electrospun fibers for peripheral nerve regeneration applications. Here, we developed the first aligned electrospun fibers to deliver pseudouridine-modified (Ψ) neurotrophin-3 (NT-3) mRNA (ΨNT-3mRNA) to primary Schwann cells and assessed NT-3 secretion and bioactivity. We first electrospun aligned poly(L-lactic acid) (PLLA) fibers and coated them with the anionic substrates dextran sulfate sodium salt (DSS) or poly(3,4-dihydroxy-L-phenylalanine) (pDOPA). Cationic lipoplexes containing ΨNT-3mRNA complexed to JetMESSENGER® were then immobilized to the fibers, resulting in detectable ΨNT-3mRNA release for 28 days from all fiber groups investigated (PLLA+mRNA, 0.5DSS4h+mRNA, and 2pDOPA4h+mRNA). The 2pDOPA4h+mRNA group significantly increased Schwann cell secretion of NT-3 for 21 days compared to control PLLA fibers (p < 0.001-0.05) and, on average, increased Schwann cell secretion of NT-3 by ≥ 2-fold compared to bolus mRNA delivery from the 1µgBolus+mRNA and 3µgBolus+mRNA groups. The 2pDOPA4h+mRNA fibers supported Schwann cell secretion of NT-3 at levels that significantly increased dorsal root ganglia (DRG) neurite extension by 44% (p < 0.0001) and neurite area by 64% (p < 0.001) compared to control PLLA fibers. The data show that the 2pDOPA4h+mRNA fibers enhance the ability of Schwann cells to promote neurite growth from DRG, demonstrating this platform's potential capability to improve peripheral nerve regeneration. STATEMENT OF SIGNIFICANCE: Aligned electrospun fibers enhance axonal regeneration by providing structural support and guidance cues, but further therapeutic stimulation is necessary to improve functional outcomes. mRNA delivery enables the transient expression of therapeutic proteins, yet achieving local, sustained delivery remains challenging. Previous work shows that genetic material delivery from electrospun fibers improves regeneration; however, mRNA delivery has not been explored. Here, we examine mRNA delivery from aligned electrospun fibers to enhance neurite outgrowth. We show that immobilization of NT-3mRNA/JetMESSENGER® lipoplexes to aligned electrospun fibers functionalized with pDOPA enables local, sustained NT-3mRNA delivery to Schwann cells, increasing Schwann cell secretion of NT-3 and enhancing DRG neurite outgrowth. This study displays the potential benefits of electrospun fiber-mediated mRNA delivery platforms for neural tissue engineering.


Asunto(s)
Ingeniería de Tejidos , Andamios del Tejido , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Polímeros/química , Ácido Láctico/química , Neuritas/metabolismo , Regeneración Nerviosa/fisiología , Factores de Crecimiento Nervioso/metabolismo
9.
Biomacromolecules ; 24(1): 294-307, 2023 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-36512693

RESUMEN

Curcumin is a natural polyphenol that exhibits remarkable antioxidant and anti-inflammatory activities; however, its clinical application is limited in part by its physiological instability. Here, we report the synthesis of curcumin-derived polyesters that release curcumin upon hydrolytic degradation to improve curcumin stability and solubility in physiological conditions. Curcumin was incorporated in the polymer backbone by a one-pot condensation polymerization in the presence of sebacoyl chloride and polyethylene glycol (PEG, Mn = 1 kDa). The thermal and mechanical properties, surface wettability, self-assembly behavior, and drug-release kinetics all depend sensitively on the mole percentage of curcumin incorporated in these statistical copolymers. Curcumin release was triggered by the hydrolysis of phenolic esters on the polymer backbone, which was confirmed using a PEGylated curcumin model compound, which represented a putative repeating unit within the polymer. The release rate of curcumin was controlled by the hydrophilicity of the polymers. Burst release (2 days) and extended release (>8 weeks) can be achieved from the same polymer depending on curcumin content in the copolymer. The materials can quench free radicals for at least 8 weeks and protect primary neurons from oxidative stress in vitro. Further, these copolymer materials could be processed into both thin films and self-assembled particles, depending on the solvent-based casting conditions. Finally, we envision that these materials may have potential for neural tissue engineering application, where antioxidant release can mitigate oxidative stress and the inflammatory response following neural injury.


Asunto(s)
Curcumina , Curcumina/farmacología , Antioxidantes/farmacología , Portadores de Fármacos , Polímeros , Polietilenglicoles , Poliésteres
10.
Ann Jt ; 8: 27, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38529222

RESUMEN

Glenohumeral bone loss is frequently observed in cases of recurrent anterior and posterior shoulder instability and represents a risk factor for failure of nonoperative treatment. Patients with suspected glenoid or humeral bone loss in the setting of recurrent instability should be evaluated with a thorough history and physical examination, as well as advanced imaging including computed tomography (CT) and/or magnetic resonance imaging (MRI). In cases of both anterior and posterior instability, the magnitude and location of bone loss should be determined, as well as the relationship between the glenoid track (GT) and any humeral defects. While the degree and pattern of osseous deficiency help guide treatment, patient-specific risk factors for recurrent instability must also be considered when determining patient management. Treatment options for subcritical anterior bone loss include labral repair and capsular plication, while more severe deficiency should prompt consideration of bony augmentation including coracoid transfer or free bone block procedures. Concomitant humeral lesions are treated according to the degree of engagement with the glenoid rim and may be addressed with soft tissue remplissage or bony augmentation procedures. While critical and subcritical thresholds of glenoid bone loss guide the management of anterior instability, such thresholds are less defined in the setting of posterior instability. Furthermore, current treatment algorithms are limited by a lack of long-term comparative studies. Future high-quality studies as well as possible modifications in indications and surgical technique are required to elucidate the optimal treatment of anterior, posterior, and bipolar glenohumeral bone loss in the setting of recurrent shoulder instability.

12.
J Neural Eng ; 19(3)2022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35580576

RESUMEN

Objective.Nerve guidance scaffolds containing anisotropic architectures provide topographical cues to direct regenerating axons through an injury site to reconnect the proximal and distal end of an injured nerve or spinal cord. Previousin vitrocultures of individual neurons revealed that fiber characteristics such as fiber diameter and inter-fiber spacing alter neurite morphological features, such as total neurite length, the longest single neurite, branching density, and the number of primary neurites. However, the relationships amongst these four neurite morphological features have never been studied on fibrous topographies using multivariate analysis.Approach.In this study, we cultured dissociated dorsal root ganglia on aligned, fibrous scaffolds and flat, isotropic films and evaluated the univariate and multivariate differences amongst these four neurite morphological features.Main results.Univariate analysis showed that fibrous scaffolds increase the length of the longest neurite and decrease branching density compared to film controls. Further, multivariate analysis revealed that, regardless of scaffold type, overall neurite length increases due to a compromise between the longest extending neurite, branching density, and the number of primary neurites. Additionally, multivariate analysis indicated that neurite branching is more independent of the other neurite features when neurons were cultured on films but that branching is strongly related to the other neurite features when cultured on fibers.Significance.These findings are significant as they are the first evidence that aligned topographies affect the relationships between neurite morphological features. This study provides a foundation for analyzing how individual neurite morphology may relate to neural regeneration on a macroscopic scale and provide information that may be used to optimize nerve guidance scaffolds.


Asunto(s)
Ganglios Espinales , Neuritas , Células Cultivadas , Ganglios Espinales/fisiología , Análisis Multivariante , Regeneración Nerviosa/fisiología , Neuritas/fisiología , Neuronas/fisiología , Poliésteres , Andamios del Tejido
13.
Cancers (Basel) ; 14(9)2022 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-35565350

RESUMEN

18F-fluorodeoxyglucose (FDG) is a glucose analog that acts as a marker for glucose uptake and metabolism. FDG PET scans are used in monitoring pediatric cancers. The handheld PET probe localization of FDG-avid lesions is an emerging modality for radio-guided surgery (RGS). We sought to assess the utility of PET probe in localizing occult FDG-avid tumors in pediatric patients. PET probe functionality was evaluated by using a PET/CT scan calibration phantom. The PET probe was able to detect FDG photon emission from simulated tumors with an expected decay of the radioisotope over time. Specificity for simulated tumor detection was lower in a model that included background FDG. In a clinical model, eight pediatric patients with FDG-avid primary, recurrent or metastatic cancer underwent a tumor excision, utilizing IV FDG and PET probe survey. Adequate tissue for diagnosis was present in 16 of 17 resected specimens, and pathology was positive for malignancy in 12 of the 17 FDG-avid lesions. PET probe gamma counts per second were higher in tumors compared with adjacent benign tissue in all operations. The median ex vivo tumor-to-background ratio (TBR) was 4.0 (range 0.9-12). The PET probe confirmed the excision of occult FDG-avid tumors in eight pediatric patients.

14.
Adv Drug Deliv Rev ; 183: 114161, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35183657

RESUMEN

Electrospun fibers are versatile biomaterial platforms with great potential to support regeneration. Electrospun fiber characteristics such as fiber diameter, degree of alignment, rate of degradation, and surface chemistry enable the creation of unique, tunable scaffolds for various drug or gene delivery applications. The delivery of genetic material and genome editing tools via viral and non-viral vectors are approaches to control cellular protein production. However, immunogenicity, off-target effects, and low delivery efficiencies slow the progression of gene delivery strategies to clinical settings. The delivery of genetic material from electrospun fibers overcomes such limitations by allowing for localized, tunable delivery of genetic material. However, the process of electrospinning is harsh, and care must be taken to retain genetic material bioactivity. This review presents an up-to-date summary of strategies to incorporate genetic material onto or within electrospun fiber platforms to improve delivery efficiency and enhance the regenerative potential of electrospun fibers for various tissue engineering applications.


Asunto(s)
Nanofibras , Materiales Biocompatibles , Edición Génica , Técnicas de Transferencia de Gen , Humanos , Ingeniería de Tejidos , Andamios del Tejido
16.
Adv Healthc Mater ; 10(22): e2101329, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34494398

RESUMEN

Neurological and functional recovery is limited following central nervous system injury and severe injury to the peripheral nervous system. Extracellular matrix (ECM)-mimetic hydrogels are of particular interest as regenerative scaffolds for the injured nervous system as they provide 3D bioactive interfaces that modulate cellular response to the injury environment and provide naturally degradable scaffolding for effective tissue remodeling. In this review, three unique ECM-mimetic hydrogels used in models of neural injury are reviewed: fibrin hydrogels, which rely on a naturally occurring enzymatic gelation, hyaluronic acid hydrogels, which require chemical modification prior to chemical crosslinking, and elastin-like polypeptide (ELP) hydrogels, which exhibit a temperature-sensitive gelation. The hydrogels are reviewed by summarizing their unique biological properties, their use as drug depots, and their combination with other biomaterials, such as electrospun fibers and nanoparticles. This review is the first to focus on these three ECM-mimetic hydrogels for their use in neural tissue engineering. Additionally, this is the first review to summarize the use of ELP hydrogels for nervous system applications. ECM-mimetic hydrogels have shown great promise in preclinical models of neural injury and future advancements in their design and use can likely lead to viable treatments for patients with neural injury.


Asunto(s)
Elastina , Ácido Hialurónico , Matriz Extracelular , Fibrina , Humanos , Hidrogeles , Péptidos , Ingeniería de Tejidos
17.
Acta Biomater ; 131: 302-313, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34271170

RESUMEN

Magnetic fiber composites combining superparamagnetic iron oxide nanoparticles (SPIONs) and electrospun fibers have shown promise in tissue engineering fields. Controlled grafting of SPIONs to the fibers post-electrospinning generates biocompatible magnetic composites without altering desired fiber morphology. Here, for the first time, we assess the potential of SPION-grafted scaffolds combined with magnetic fields to promote neurite outgrowth by providing contact guidance from the aligned fibers and mechanical stimulation from the SPIONs in the magnetic field. Neurite outgrowth from primary rat dorsal root ganglia (DRG) was assessed from explants cultured on aligned control and SPION-grafted electrospun fibers as well as on non-grafted fibers with SPIONs dispersed in the culture media. To determine the optimal magnetic field stimulation to promote neurite outgrowth, we generated a static, alternating, and linearly moving magnet and simulated the magnetic flux density at different areas of the scaffold over time. The alternating magnetic field increased neurite length by 40% on control fibers compared to a static magnetic field. Additionally, stimulation with an alternating magnetic field resulted in a 30% increase in neurite length and 62% increase in neurite area on SPION-grafted fibers compared to DRG cultured on PLLA fibers with untethered SPIONs added to the culture media. These findings demonstrate that SPION-grafted fiber composites in combination with magnetic fields are more beneficial for stimulating neurite outgrowth on electrospun fibers than dispersed SPIONs. STATEMENT OF SIGNIFICANCE: Aligned electrospun fibers improve axonal regeneration by acting as a passive guidance cue but do not actively interact with cells, while magnetic nanoparticles can be remotely manipulated to interact with neurons and elicit neurite outgrowth. Here, for the first time, we examine the combination of magnetic fields, magnetic nanoparticles, and aligned electrospun fibers to enhance neurite outgrowth. We show an alternating magnetic field alone increases neurite outgrowth on aligned electrospun fibers. However, combining the alternating field with magnetic nanoparticle-grafted fibers does not affect neurite outgrowth compared to control fibers but improves outgrowth compared to freely dispersed magnetic nanoparticles. This study provides the groundwork for utilizing magnetic electrospun fibers and magnetic fields as a method for promoting axonal growth.


Asunto(s)
Ganglios Espinales , Andamios del Tejido , Animales , Campos Magnéticos , Nanopartículas Magnéticas de Óxido de Hierro , Neuritas , Proyección Neuronal , Ratas
18.
Sci Transl Med ; 13(591)2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33910979

RESUMEN

Treatment of solid cancers with chimeric antigen receptor (CAR) T cells is plagued by the lack of ideal target antigens that are both absolutely tumor specific and homogeneously expressed. We show that multi-antigen prime-and-kill recognition circuits provide flexibility and precision to overcome these challenges in the context of glioblastoma. A synNotch receptor that recognizes a specific priming antigen, such as the heterogeneous but tumor-specific glioblastoma neoantigen epidermal growth factor receptor splice variant III (EGFRvIII) or the central nervous system (CNS) tissue-specific antigen myelin oligodendrocyte glycoprotein (MOG), can be used to locally induce expression of a CAR. This enables thorough but controlled tumor cell killing by targeting antigens that are homogeneous but not absolutely tumor specific. Moreover, synNotch-regulated CAR expression averts tonic signaling and exhaustion, maintaining a higher fraction of the T cells in a naïve/stem cell memory state. In immunodeficient mice bearing intracerebral patient-derived xenografts (PDXs) with heterogeneous expression of EGFRvIII, a single intravenous infusion of EGFRvIII synNotch-CAR T cells demonstrated higher antitumor efficacy and T cell durability than conventional constitutively expressed CAR T cells, without off-tumor killing. T cells transduced with a synNotch-CAR circuit primed by the CNS-specific antigen MOG also exhibited precise and potent control of intracerebral PDX without evidence of priming outside of the brain. In summary, by using circuits that integrate recognition of multiple imperfect but complementary antigens, we improve the specificity, completeness, and persistence of T cells directed against glioblastoma, providing a general recognition strategy applicable to other solid tumors.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Animales , Encéfalo/metabolismo , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Glioblastoma/terapia , Inmunoterapia Adoptiva , Ratones , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
ACS Chem Neurosci ; 12(6): 959-965, 2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33635633

RESUMEN

17ß-Estradiol (E2) confers neuroprotection in preclinical models of spinal cord injury when administered systemically. The goal of this study was to apply E2 locally to the injured spinal cord for a sustained duration using poly(pro-E2) film biomaterials. Following contusive spinal cord injury in adult male mice, poly(pro-E2) films were implanted subdurally and neuroprotection was assessed using immunohistochemistry 7 days after injury and implantation. In these studies, poly(pro-E2) films modestly improved neuroprotection without affecting the inflammatory response when compared to the injured controls. To increase the E2 dose released, bolus-releasing poly(pro-E2) films were fabricated by incorporating unbound E2 into the poly(pro-E2) films. However, compared to the injured controls, bolus-releasing poly(pro-E2) films did not significantly enhance neuroprotection or limit inflammation at either 7 or 21 days post-injury. Future work will focus on developing poly(pro-E2) biomaterials capable of more precisely releasing therapeutic doses of E2.


Asunto(s)
Contusiones , Fármacos Neuroprotectores , Traumatismos de la Médula Espinal , Animales , Estradiol , Masculino , Ratones , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/tratamiento farmacológico
20.
J Neurosci Res ; 99(3): 806-826, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33295039

RESUMEN

Astrocytes are responsible for a wide variety of essential functions throughout the central nervous system. The protein markers glial fibrillary acidic protein (GFAP), glutamate aspartate transporter (GLAST), glutamate transporter-1 (GLT-1), glutamine synthetase (GS), 10-formyltetrahydrofolate dehydrogenase (ALDH1L1), and the transcription factor SOX9 are routinely used to label astrocytes in primary rodent cultures. However, GLAST, GLT-1, GS, and SOX9 are also produced by microglia and oligodendrocytes and GFAP, GLAST, GLT-1, and GS production levels are affected by astrocyte phenotypic changes associated with reactive astrogliosis. No group has performed a comprehensive immunocytochemical evaluation to quantify the percentage of cells labeled by these markers in vitro, nor compared changes in staining between cortex- and spinal cord-derived cells in naïve and stimulated cultures. Here, we quantified the percentage of cells positively stained for these six markers in astrocyte, microglia, and oligodendrocyte cultures isolated from neonatal rat cortices and spinal cords. Additionally, we incubated the astrocytes with transforming growth factor (TGF)-ß1 or TGF-ß3 to determine if the labeling of these markers is altered by these stimuli. We found that only SOX9 in cortical cultures and ALDH1L1 in spinal cord cultures labeled more than 75% of the cells in naïve and stimulated astrocyte cultures and stained less than 5% of the cells in microglia and oligodendrocyte cultures. Furthermore, significantly more cortical than spinal cord astrocytes stained for GFAP, GLAST, and ALDH1L1 in naïve cultures, whereas significantly more spinal cord than cortical astrocytes stained for GLAST and GS in TGF-ß1-treated cultures. These findings are important as variability in marker staining may lead to misinterpretation of the astrocyte response in cocultures, migration assays, or engineered disease models.


Asunto(s)
Astrocitos/metabolismo , Corteza Cerebelosa/metabolismo , Médula Espinal/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Factor de Crecimiento Transformador beta3/farmacología , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutamato-Amoníaco Ligasa/metabolismo , Microglía/metabolismo , Neuroglía/metabolismo , Oligodendroglía/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Factor de Transcripción SOX9/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA