Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Adv Funct Mater ; 33(28)2023 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-37873031

RESUMEN

Current screening and diagnostic tools for traumatic brain injury (TBI) have limitations in sensitivity and prognostication. Aberrant protease activity is a central process that drives disease progression in TBI and is associated with worsened prognosis; thus direct measurements of protease activity could provide more diagnostic information. In this study, a nanosensor is engineered to release a measurable signal into the blood and urine in response to activity from the TBI-associated protease calpain. Readouts from the nanosensor were designed to be compatible with ELISA and lateral flow assays, clinically-relevant assay modalities. In a mouse model of TBI, the nanosensor sensitivity is enhanced when ligands that target hyaluronic acid are added. In evaluation of mice with mild or severe injuries, the nanosensor identifies mild TBI with a higher sensitivity than the biomarker GFAP. This nanosensor technology allows for measurement of TBI-associated proteases without the need to directly access brain tissue, and has the potential to complement existing TBI diagnostic tools.

2.
Bioconjug Chem ; 33(9): 1685-1697, 2022 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-36017941

RESUMEN

Traumatic brain injury (TBI) is a major cause of disability and death among children and young adults in the United States, yet there are currently no treatments that improve the long-term brain health of patients. One promising therapeutic for TBI is brain-derived neurotrophic factor (BDNF), a protein that promotes neurogenesis and neuron survival. However, outstanding challenges to the systemic delivery of BDNF are its instability in blood, poor transport into the brain, and short half-life in circulation and brain tissue. Here, BDNF is encapsulated into an engineered, biodegradable porous silicon nanoparticle (pSiNP) in order to deliver bioactive BDNF to injured brain tissue after TBI. The pSiNP carrier is modified with the targeting ligand CAQK, a peptide that binds to extracellular matrix components upregulated after TBI. The protein cargo retains bioactivity after release from the pSiNP carrier, and systemic administration of the CAQK-modified pSiNPs results in effective delivery of the protein cargo to injured brain regions in a mouse model of TBI. When administered after injury, the CAQK-targeted pSiNP delivery system for BDNF reduces lesion volumes compared to free BDNF, supporting the hypothesis that pSiNPs mediate therapeutic protein delivery after systemic administration to improve outcomes in TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Nanopartículas , Animales , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Matriz Extracelular , Ligandos , Ratones , Péptidos/uso terapéutico , Porosidad , Silicio
3.
Mol Pharm ; 18(2): 522-538, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32584042

RESUMEN

Acute brain injuries such as traumatic brain injury and stroke affect 85 million people a year worldwide, and many survivors suffer from long-term physical, cognitive, or psychosocial impairments. There are few FDA-approved therapies that are effective at preventing, halting, or ameliorating the state of disease in the brain after acute brain injury. To address this unmet need, one potential strategy is to leverage the unique physical and biological properties of nanomaterials. Decades of cancer nanomedicine research can serve as a blueprint for innovation in brain injury nanomedicines, both to emulate the successes and also to avoid potential pitfalls. In this review, we discuss how shared disease physiology between cancer and acute brain injuries can inform the design of novel nanomedicines for acute brain injuries. These disease hallmarks include dysregulated vasculature, an altered microenvironment, and changes in the immune system. We discuss several nanomaterial strategies that can be engineered to exploit these disease hallmarks, for example, passive accumulation, active targeting of disease-associated signals, bioresponsive designs that are "smart", and immune interactions.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Portadores de Fármacos/química , Nanopartículas/química , Fármacos Neuroprotectores/administración & dosificación , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Disponibilidad Biológica , Barrera Hematoencefálica/metabolismo , Encéfalo/inmunología , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Modelos Animales de Enfermedad , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Fármacos Neuroprotectores/farmacocinética , Permeabilidad , Accidente Cerebrovascular/patología , Distribución Tisular , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
4.
Mol Pharm ; 17(9): 3633-3642, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32786959

RESUMEN

Therapeutic nucleic acids hold great promise for the treatment of genetic diseases, yet the delivery of this highly charged macromolecular drug remains a challenge in the field. Peptides are promising agents to mediate nucleic acid delivery because they can encode a biological function to overcome the trafficking barriers. Electrostatic nanocomplexes of nucleic acid and peptides can achieve effective delivery, but the balance between their stability and biological function must be finely tuned. In this work, we explore two peptide building blocks that have been studied in the literature: targeting ligands and intracellular trafficking peptides. We grafted these peptides on a polyethylene glycol (PEG) backbone with eight sites for substitution to create so-called "peptide spiders". These conjugates achieve stability via the well-known hydrophilic shielding effect of PEG. In addition, the coordination of peptide building blocks into multimers may create new biological properties, such as the well-known phenomena of increased binding avidity with multivalent ligands. In this work, we linked two trafficking peptides to the PEG backbone using either nonreducible or reducible chemistries and investigated the ability of these materials to carry silencing RNAs into mammalian cells. We then investigated these nanomaterials for their pharmacokinetic properties and silencing of undruggable targets in a mouse model of cancer. While reducible linkages were more potent at silencing in vitro, this effect was reversed when applied in the context of living animals. This work offers an insight into peptide-based delivery materials and investigates peptide-polymer linkages.


Asunto(s)
Ácidos Nucleicos/química , Péptidos/química , Polímeros/química , Animales , Línea Celular Tumoral , Femenino , Humanos , Ligandos , Sustancias Macromoleculares/química , Ratones , Ratones Desnudos , Nanopartículas/química , Neoplasias/metabolismo , Polietilenglicoles/química , ARN Interferente Pequeño/química , Células U937
5.
Proc Natl Acad Sci U S A ; 112(47): 14460-6, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26598694

RESUMEN

Nanoparticle technologies intended for human administration must be designed to interact with, and ideally leverage, a living host environment. Here, we describe smart nanosystems classified in two categories: (i) those that sense the host environment and respond and (ii) those that first prime the host environment to interact with engineered nanoparticles. Smart nanosystems have the potential to produce personalized diagnostic and therapeutic schema by using the local environment to drive material behavior and ultimately improve human health.


Asunto(s)
Nanomedicina , Nanotecnología , Homeostasis , Humanos , Concentración de Iones de Hidrógeno , Nanopartículas , Oxidación-Reducción
6.
Bioconjug Chem ; 27(10): 2323-2331, 2016 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-27583545

RESUMEN

Nanoparticulate systems have shown great promise in overcoming the considerable trafficking barriers associated with systemic nucleic acid delivery, which must be addressed to unlock the full potential of technologies such as RNAi and gene editing in vivo. In addition to mediating the cytoplasmic delivery of nucleic cargo and shielding it from nuclease degradation and immunostimulation, nucleic-acid-containing nanomaterials delivered intravenously must also be stable in the bloodstream after administration to avoid toxicity and off-target delivery. To this end, the hydrophilic molecule polyethylene glycol (PEG) has been deployed in many different nanoparticle systems to prevent aggregation and recognition by the reticuloendothelial system. However, the optimal strategy for incorporating PEG into self-assembled nucleic acid delivery systems to obtain nanoparticle stability while retaining important functions such as receptor targeting and cargo activity remains unclear. In this work, we develop substantially improved formulations of tumor-penetrating nanocomplexes (TPNs), targeted self-assembled nanoparticles formulated with peptide carriers and siRNA that have been shown to mitigate tumor burden in an orthotopic model of ovarian cancer. We specifically sought to tailor TPNs for intravenous delivery by systematically comparing formulations with three different classes of modular PEG incorporation (namely PEG graft polymers, PEG lipids, and PEGylated peptide), each synthesized using straightforward bioconjugation techniques. We found that the addition of PEG lipids or PEGylated peptide carriers led to the formation of small and stable nanoparticles, but only nanoparticles formulated with PEGylated peptide carriers retained substantial activity in a gene silencing assay. In vivo, this formulation significantly decreased accumulation in off-target organs and improved initial availability in circulation compared to results from the original non-PEGylated particles. Thus, from among a set of candidate strategies, we identified TPNs with admixed PEGylated peptide carriers as the optimal formulation for systemic administration of siRNA on the basis of their performance in a battery of physicochemical and biological assays. Moreover, this optimized formulation confers pharmacologic advantages that may enable further translational development of tumor-penetrating nanocomplexes, highlighting the preclinical value of comparing formulation strategies and the relevance of this systematic approach for the development of other self-assembled nanomaterials.


Asunto(s)
Técnicas de Silenciamiento del Gen/métodos , Nanopartículas/química , Péptidos/química , Polietilenglicoles/química , ARN Interferente Pequeño/química , Animales , Sistemas de Liberación de Medicamentos , Dispersión Dinámica de Luz , Femenino , Silenciador del Gen , Humanos , Ratones , Ratones Desnudos , Microscopía Electrónica de Transmisión , Nanopartículas/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/farmacocinética , Distribución Tisular
7.
Adv Mater ; : e2301738, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38780012

RESUMEN

Traumatic brain injury (TBI) is a critical public health concern, yet there are no therapeutics available to improve long-term outcomes. Drug delivery to TBI remains a challenge due to the blood-brain barrier and increased intracranial pressure. In this work, a chemical targeting approach to improve delivery of materials to the injured brain, is developed. It is hypothesized that the provisional fibrin matrix can be harnessed as an injury-specific scaffold that can be targeted by materials via click chemistry. To accomplish this, the brain clot is engineered in situ by delivering fibrinogen modified with strained cyclooctyne (SCO) moieties, which incorporated into the injury lesion and is retained there for days. Improved intra-injury capture and retention of diverse, clickable azide-materials including a small molecule azide-dye, 40 kDa azide-PEG nanomaterial, and a therapeutic azide-protein in multiple dosing regimens is subsequently observed. To demonstrate therapeutic translation of this approach, a reduction in reactive oxygen species levels in the injured brain after delivery of the antioxidant catalase, is achieved. Further, colocalization between azide and SCO-fibrinogen is specific to the brain over off-target organs. Taken together, a chemical targeting strategy leveraging endogenous clot formation is established which can be applied to improve therapeutic delivery after TBI.

8.
ACS Biomater Sci Eng ; 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38870483

RESUMEN

After traumatic brain injury, the brain extracellular matrix undergoes structural rearrangement due to changes in matrix composition, activation of proteases, and deposition of chondroitin sulfate proteoglycans by reactive astrocytes to produce the glial scar. These changes lead to a softening of the tissue, where the stiffness of the contusion "core" and peripheral "pericontusional" regions becomes softer than that of healthy tissue. Pioneering mechanotransduction studies have shown that soft substrates upregulate intermediate filament proteins in reactive astrocytes; however, many other aspects of astrocyte biology remain unclear. Here, we developed a platform for the culture of cortical astrocytes using polyacrylamide (PA) gels of varying stiffness (measured in Pascal; Pa) to mimic injury-related regions in order to investigate the effects of tissue stiffness on astrocyte reactivity and morphology. Our results show that substrate stiffness influences astrocyte phenotype; soft 300 Pa substrates led to increased GFAP immunoreactivity, proliferation, and complexity of processes. Intermediate 800 Pa substrates increased Aggrecan+, Brevican+, and Neurocan+ astrocytes. The stiffest 1 kPa substrates led to astrocytes with basal morphologies, similar to a physiological state. These results advance our understanding of astrocyte mechanotransduction processes and provide evidence of how substrates with engineered stiffness can mimic the injury microenvironment.

9.
Nat Biotechnol ; 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38168984

RESUMEN

RNA-binding proteins (RBPs) modulate alternative splicing outcomes to determine isoform expression and cellular survival. To identify RBPs that directly drive alternative exon inclusion, we developed tethered function luciferase-based splicing reporters that provide rapid, scalable and robust readouts of exon inclusion changes and used these to evaluate 718 human RBPs. We performed enhanced cross-linking immunoprecipitation, RNA sequencing and affinity purification-mass spectrometry to investigate a subset of candidates with no prior association with splicing. Integrative analysis of these assays indicates surprising roles for TRNAU1AP, SCAF8 and RTCA in the modulation of hundreds of endogenous splicing events. We also leveraged our tethering assays and top candidates to identify potent and compact exon inclusion activation domains for splicing modulation applications. Using these identified domains, we engineered programmable fusion proteins that outperform current artificial splicing factors at manipulating inclusion of reporter and endogenous exons. This tethering approach characterizes the ability of RBPs to induce exon inclusion and yields new molecular parts for programmable splicing control.

10.
Learn Mem ; 19(9): 359-68, 2012 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-22904366

RESUMEN

MicroRNAs (miRNAs) represent a class of small regulatory noncoding RNAs ∼22 bp in length that mediate post-transcriptional silencing of gene expression via the recognition of specific sequences in target messenger (m)RNAs. The current body of literature suggests that miRNAs are fine-tuning regulators of gene expression profiles in a wide range of biological processes, from development to cancer. Many miRNAs are highly expressed in the adult nervous system in a spatially and temporally controlled manner in normal physiology, as well as in certain pathological conditions. These findings emphasize that gene regulation networks based on miRNA activities may be particularly important to brain function, and that perturbation of these networks may result in abnormal brain function. Indeed, miRNAs have been implicated in various aspects of dendrite remodeling and synaptic plasticity, as well as in experience-dependent adaptive changes of neural circuits in the postnatal developmental and adult brain. Recent advances in methods of next-generation sequencing, such as RNA-seq, offer the means to quantitatively evaluate the functions of miRNAs in a genome-wide manner in large cohorts of samples. These new technologies have already yielded valuable information and are expanding our understanding of miRNA-based mechanisms in higher-order brain processing, including learning and memory and cognition, as well as in neuropsychiatric disorders.


Asunto(s)
Encéfalo/metabolismo , Aprendizaje/fisiología , MicroARNs/genética , MicroARNs/metabolismo , Enfermedades del Sistema Nervioso/genética , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/genética , Humanos , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/genética , Trastornos de la Memoria/etiología , Trastornos de la Memoria/genética , Enfermedades del Sistema Nervioso/complicaciones , Enfermedades del Sistema Nervioso/metabolismo , Plasticidad Neuronal/fisiología
11.
Biomater Sci ; 11(12): 4238-4253, 2023 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-36987922

RESUMEN

Traumatic brain injury (TBI) affects millions of people worldwide, yet there are currently no therapeutics that address the long-term impairments that develop in a large portion of survivors. Lipid nanoparticles (LNPs) are a promising therapeutic strategy that may address the molecular basis of TBI pathophysiology. LNPs are the only non-viral gene delivery platform to achieve clinical success, but systemically administered formulations have only been established for targets in the liver. In this work, we evaluated the pharmacokinetics and activity of LNPs formulated with polyethylene glycol (PEG)-lipids of different anchor lengths when systemically administered to a mouse model of TBI. We observed an increase in LNP accumulation and activity in the injured brain hemisphere compared to the uninjured contralateral brain hemisphere. Interestingly, transgene expression mediated by LNPs was more durable in injured brain tissue compared to off-target organs when compared between 4 and 24 hours. The PEG-lipid is an important component of LNP formulation necessary for the stable formation and storage of LNPs, but the PEG-lipid structure and content also has an impact on LNP function. LNP formulations containing various ratios of PEG-lipid with C18 (DSPE-PEG) and C14 (DMG-PEG) anchors displayed similar physicochemical properties, independent of the PEG-lipid compositions. As the proportion of DSPE-PEG was increased in formulations, blood circulation times of LNPs increased and the duration of expression increased. We also evaluated diffusion of LNPs after convection enhanced delivery (CED) in healthy brains and found LNPs distributed >1 mm away from the injection site. Understanding LNP pharmacokinetics and activity in TBI models and the impact of PEG-lipid anchor length informs the design of LNP-based therapies for TBI after systemic administration.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Nanopartículas , Ratones , Animales , Polietilenglicoles/química , Liposomas , Nanopartículas/química , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lípidos/química , ARN Interferente Pequeño/genética
12.
Adv Drug Deliv Rev ; 197: 114820, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37054953

RESUMEN

The blood-brain barrier (BBB) is a highly regulated physical and functional boundarythat tightly controls the transport of materials between the blood and the brain. There is an increasing recognition that the BBB is dysfunctional in a wide range of neurological disorders; this dysfunction can be symptomatic of the disease but can also play a role in disease etiology. BBB dysfunction can be exploited for the delivery of therapeutic nanomaterials. Forexample, there can be a transient, physical disruption of the BBB in diseases such as brain injury and stroke, which allows temporary access of nanomaterials into the brain. Physicaldisruption of the BBB through external energy sources is now being clinically pursued toincrease therapeutic delivery into the brain. In other diseases, the BBB takes on new properties that can beleveraged by delivery carriers. For instance, neuroinflammation induces the expression ofreceptors on the BBB that can be targeted by ligand-modified nanomaterials, and theendogenous homing of immune cells into the diseased brain can be hijacked for the delivery ofnanomaterials. Lastly, BBB transport pathways can be altered to increase nanomaterial transport. In this review, we will describe changes that can occur in the BBB in disease, and how these changes have been exploited by engineered nanomaterials forincreased transport into the brain.


Asunto(s)
Nanoestructuras , Accidente Cerebrovascular , Humanos , Barrera Hematoencefálica/metabolismo , Encéfalo , Transporte Biológico , Accidente Cerebrovascular/metabolismo
13.
Adv Healthc Mater ; 12(25): e2300782, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37390094

RESUMEN

Traumatic brain injury (TBI) affects millions of people each year and, in many cases, results in long-term disabilities. Once a TBI has occurred, there is a significant breakdown of the blood-brain barrier resulting in increased vascular permeability and progression of the injury. In this study, the use of an infusible extracellular matrix-derived biomaterial (iECM) for its ability to reduce vascular permeability and modulate gene expression in the injured brain is investigated. First, the pharmacokinetics of iECM administration in a mouse model of TBI is characterized, and the robust accumulation of iECM at the site of injury is demonstrated. Next, it is shown that iECM administration after injury can reduce the extravasation of molecules into the brain, and in vitro, iECM increases trans-endothelial electrical resistance across a monolayer of TNFα-stimulated endothelial cells. In gene expression analysis of brain tissue, iECM induces changes that are indicative of downregulation of the proinflammatory response 1-day post-injury/treatment and neuroprotection at 5 days post-injury/treatment. Therefore, iECM shows potential as a treatment for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Humanos , Ratones , Animales , Células Endoteliales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Barrera Hematoencefálica/metabolismo , Modelos Animales de Enfermedad
14.
Nat Biomed Eng ; 7(2): 94-109, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36581694

RESUMEN

Decellularized extracellular matrix in the form of patches and locally injected hydrogels has long been used as therapies in animal models of disease. Here we report the safety and feasibility of an intravascularly infused extracellular matrix as a biomaterial for the repair of tissue in animal models of acute myocardial infarction, traumatic brain injury and pulmonary arterial hypertension. The biomaterial consists of decellularized, enzymatically digested and fractionated ventricular myocardium, localizes to injured tissues by binding to leaky microvasculature, and is largely degraded in about 3 d. In rats and pigs with induced acute myocardial infarction followed by intracoronary infusion of the biomaterial, we observed substantially reduced left ventricular volumes and improved wall-motion scores, as well as differential expression of genes associated with tissue repair and inflammation. Delivering pro-healing extracellular matrix by intravascular infusion post injury may provide translational advantages for the healing of inflamed tissues 'from the inside out'.


Asunto(s)
Materiales Biocompatibles , Infarto del Miocardio , Ratas , Porcinos , Animales , Miocardio/metabolismo , Infarto del Miocardio/terapia , Hidrogeles , Matriz Extracelular/metabolismo
15.
Neurosci Insights ; 17: 26331055221129641, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36274925

RESUMEN

Mild traumatic brain injury (mTBI), a condition in which brain function is transiently disrupted by a mechanical force, is a major risk factor for developing Alzheimer's disease (AD) and other neurodegenerative conditions. In this commentary, we summarize recent findings in human neurons derived from induced pluripotent stem cells, detailing early neuronal events following mild injury that may seed future neurodegeneration. In particular, we discuss interlinked relationships between mTBI and several biological pathways hypothesized to underlie AD progression, including amyloidogenic cleavage of amyloid precursor protein (APP), impairment of axonal transport, and the development of APP-associated axonal swellings. We also describe the implications of these findings for future mechanistic and translational studies.

16.
ACS Nano ; 15(12): 20504-20516, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34870408

RESUMEN

Traumatic brain injury (TBI) is a critical public health concern and major contributor to death and long-term disability. After the initial trauma, a sustained secondary injury involving a complex continuum of pathophysiology unfolds, ultimately leading to the destruction of nervous tissue. One disease hallmark of TBI is ectopic protease activity, which can mediate cell death, extracellular matrix breakdown, and inflammation. We previously engineered a fluorogenic activity-based nanosensor for TBI (TBI-ABN) that passively accumulates in the injured brain across the disrupted vasculature and generates fluorescent signal in response to calpain-1 cleavage, thus enabling in situ visualization of TBI-associated calpain-1 protease activity. In this work, we hypothesized that actively targeting the extracellular matrix (ECM) of the injured brain would improve nanosensor accumulation in the injured brain beyond passive delivery alone and lead to increased nanosensor activation. We evaluated several peptides that bind exposed/enriched ECM constituents in the brain and discovered that nanomaterials modified with peptides that target hyaluronic acid (HA) displayed widespread distribution across the injury lesion, in particular colocalizing with perilesional and hippocampal neurons. Modifying TBI-ABN with HA-targeting peptide led to increases in activation in a ligand-valency-dependent manner, up to 6.6-fold in the injured cortex compared to a nontargeted nanosensor. This robust nanosensor activation enabled 3D visualization of injury-specific protease activity in a cleared and intact brain. In our work, we establish that targeting brain ECM with peptide ligands can be leveraged to improve the distribution and function of a bioresponsive imaging nanomaterial.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Encéfalo/metabolismo , Calpaína/metabolismo , Matriz Extracelular/metabolismo , Humanos
17.
AAPS J ; 23(5): 100, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34401968

RESUMEN

Peptides are used to control the pharmacokinetic profiles of nanoparticles due to their ability to influence tissue accumulation and cellular interactions. However, beyond the study of specific peptides, there is a lack of understanding of how peptide physicochemical properties affect nanoparticle pharmacokinetics, particularly in the context of traumatic brain injury (TBI). We engineered nanoparticle surfaces with peptides that possess a range of physicochemical properties and evaluated their distribution after two routes of administration: direct injection into a healthy mouse brain and systemic delivery in a mouse model of TBI. In both administration routes, we found that peptide-modified nanoparticle pharmacokinetics were influenced by the charge characteristics of the peptide. When peptide-modified nanoparticles are delivered directly into the brain, nanoparticles modified with positively charged peptides displayed restricted distribution from the injection site compared to nanoparticles modified with neutral, zwitterionic, or negatively charged peptides. After intravenous administration in a TBI mouse model, positively charged peptide-modified nanoparticles accumulated more in off-target organs, including the heart, lung, and kidneys, than zwitterionic, neutral, or negatively charged peptide-modified nanoparticles. The increase in off-target organ accumulation of positively charged peptide-modified nanoparticles was concomitant with a relative decrease in accumulation in the injured brain compared to zwitterionic, neutral, or negatively charged peptide-modified nanoparticles. Understanding how nanoparticle pharmacokinetics are influenced by the physicochemical properties of peptides presented on the nanoparticle surface is relevant to the development of nanoparticle-based TBI therapeutics and broadly applicable to nanotherapeutic design, including synthetic nanoparticles and viruses.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Sistema de Administración de Fármacos con Nanopartículas/farmacocinética , Fármacos Neuroprotectores/administración & dosificación , Péptidos/química , Animales , Disponibilidad Biológica , Lesiones Traumáticas del Encéfalo/patología , Ingeniería Química , Química Farmacéutica , Modelos Animales de Enfermedad , Femenino , Semivida , Humanos , Ratones , Sistema de Administración de Fármacos con Nanopartículas/química , Péptidos/farmacocinética , Distribución Tisular
18.
ACS Infect Dis ; 7(4): 721-732, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33689277

RESUMEN

The drug-impermeable bacterial membrane in Gram-negative pathogens limits antibiotic access to intracellular drug targets. To expand our rapidly waning antibiotic arsenal, one approach is to improve the intracellular delivery of drugs with historically poor accumulation in Gram-negative bacteria. To do so, we engineered macromolecular potentiators to permeabilize the Gram-negative membrane to facilitate drug influx. Potentiators, known as WD40, were synthesized by grafting multiple copies of a cationic α-helical antimicrobial peptide, WLBU2, onto a dextran polymer scaffold. WD40 enabled drug uptake in the model pathogen P. aeruginosa, a capability that was not observed with unmodified WLBU2 peptide. WD40 was able to reduce minimum inhibitory concentrations of a drug panel by up to 3 orders of magnitude. Hydrophobic and highly three-dimensional antibiotics exhibited the greatest potentiation. Antibiotic activity was potentiated in several clinical strains and resulted in sensitization of drug-resistant strains to rifampin, a drug not previously used for Gram-negative infections.


Asunto(s)
Antibacterianos , Bacterias Gramnegativas , Antibacterianos/farmacología , Pruebas de Sensibilidad Microbiana , Rifampin
19.
Mol Pharm ; 7(4): 1260-5, 2010 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-20476763

RESUMEN

HGP is a 24-amino acid peptide derived from HIV gp41 that increases vesicular escape when incorporated into gene delivery vehicles. The typical yield of HGP from solid phase peptide synthesis is low due to its length and hydrophobicity. The goal of this work was to investigate truncated sequences that maintained activity in order to improve the ease and yield of synthesis. A shortened, 15-amino acid sequence retained comparable lytic activity and the ability to interact with lipids when compared to the full length peptide. A scrambled peptide showed poor lytic activity, confirming that the activity of these endosomal escape peptides is sequence specific. Peptides were covalently attached to polyethylenimine (PEI) and used to condense plasmid DNA to form nanoparticulate carriers. When delivery efficiencies of PEI-peptide conjugates were compared in vitro, PEI modified with the truncated HGP sequence increased transgene expression over unmodified PEI and full length HGP.


Asunto(s)
Técnicas de Transferencia de Gen , Péptidos/química , Células HeLa , Humanos , Liposomas/química , Péptidos/síntesis química , Polietileneimina/química
20.
ACS Sens ; 5(3): 686-692, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32100994

RESUMEN

Currently, traumatic brain injury (TBI) is detected by medical imaging; however, medical imaging requires expensive capital equipment, is time- and resource-intensive, and is poor at predicting patient prognosis. To date, direct measurement of elevated protease activity has yet to be utilized to detect TBI. In this work, we engineered an activity-based nanosensor for TBI (TBI-ABN) that responds to increased protease activity initiated after brain injury. We establish that a calcium-sensitive protease, calpain-1, is active in the injured brain hours within injury. We then optimize the molecular weight of a nanoscale polymeric carrier to infiltrate into the injured brain tissue with minimal renal filtration. A calpain-1 substrate that generates a fluorescent signal upon cleavage was attached to this nanoscale polymeric carrier to generate an engineered TBI-ABN. When applied intravenously to a mouse model of TBI, our engineered sensor is observed to locally activate in the injured brain tissue. This TBI-ABN is the first demonstration of a sensor that responds to protease activity to detect TBI.


Asunto(s)
Técnicas Biosensibles , Lesiones Traumáticas del Encéfalo/enzimología , Encéfalo/enzimología , Calpaína/metabolismo , Animales , Calpaína/química , Femenino , Ratones Endogámicos C57BL , Nanopartículas/química , Polímeros/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA