Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Control Release ; 327: 667-675, 2020 11 10.
Article in English | MEDLINE | ID: mdl-32918963

ABSTRACT

The blood-brain barrier (BBB) is a dynamic diffusional barrier regulating the molecular and chemical flux between the blood and brain, thereby preserving cerebral homeostasis. Endothelial cells form the core anatomical component of the BBB based on properties such as specialized junctional complexes between cells, which restricts paracellular transport, and extremely low levels of vesicular transport, restricting transcytosis. In performing its protective function, the BBB also constrains the entry of therapeutics into the brain, hampering the treatment of various neurological disorders. Focused ultrasound is a novel therapeutic modality that has shown efficacy in transiently and non-invasively opening the BBB for the targeted delivery of therapeutics to the brain. Although the ability of ultrasound to disrupt the junctional assembly of endothelial cells has been partially investigated, its effect on the transcellular mode of transport has been largely neglected. In this study, we found that ultrasound induces a pronounced increase in the levels of the vesicle-forming protein caveolin-1. In order to investigate the role of vesicle-mediated transcytoplasmic transport, we compared the leakage of various cargo sizes between a mouse model that lacks caveolin-1 and wild-type mice following sonication of the hippocampus. The absence of caveolin-1 did not lead to overt abnormalities in the cerebral vasculature in the mice. We found that caveolin-1 has a critical role specifically in the transport of large (500 kDa), but not smaller (3 and 70 kDa) cargoes. Our findings indicate differential effects of therapeutic ultrasound on cellular transport mechanisms, with implications for therapeutic interventions.


Subject(s)
Caveolin 1 , Endothelial Cells , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Endothelial Cells/metabolism , Mice , Transcytosis
2.
Stem Cell Reports ; 14(5): 924-939, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32275861

ABSTRACT

The blood-brain barrier (BBB) presents a barrier for circulating factors, but simultaneously challenges drug delivery. How the BBB is altered in Alzheimer disease (AD) is not fully understood. To facilitate this analysis, we derived brain endothelial cells (iBECs) from human induced pluripotent stem cells (hiPSCs) of several patients carrying the familial AD PSEN1 mutation. We demonstrate that, compared with isogenic PSEN1 corrected and control iBECs, AD-iBECs exhibit altered tight and adherens junction protein expression as well as efflux properties. Furthermore, by applying focused ultrasound (FUS) that transiently opens the BBB and achieves multiple therapeutic effects in AD mouse models, we found an altered permeability to 3-5 kDa dextran as a model cargo and the amyloid-ß (Aß) peptide in AD-iBECs compared with control iBECs. This presents human-derived in vitro models of the BBB as a valuable tool to understand its role and properties in a disease context, with possible implications for drug delivery.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Alzheimer Disease/therapy , Animals , Blood-Brain Barrier/cytology , Cell Line , Cells, Cultured , Connexins/metabolism , Dextrans/pharmacokinetics , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Phenotype , Presenilin-1/genetics , Ultrasonic Therapy
3.
Adv Drug Deliv Rev ; 165-166: 1-14, 2020.
Article in English | MEDLINE | ID: mdl-31790711

ABSTRACT

The blood-brain barrier (BBB) is a dynamic structure that functions as a gatekeeper, reflecting the unique requirements of the brain. In this review, following a brief historical overview of how the concepts of the BBB and the neurovascular unit (NVU) developed, we describe its physiology and architecture, which pose a particular challenge to therapeutic intervention. We then discuss how the restrictive nature of this barrier can be overcome for the delivery of therapeutic agents. Alterations to drug formulation offer one option, in part by utilizing distinct transport modes; another is invasive or non-invasive strategies to bypass the BBB. An emerging non-invasive technology for targeted drug delivery is focused ultrasound that allows for the safe and reversible disruption of the BBB. We discuss the underlying mechanisms and provide an outlook, emphasizing the need for more research into the NVU and investment in innovative technologies to overcome the BBB for drug delivery.


Subject(s)
Blood-Brain Barrier/physiology , Drug Delivery Systems/methods , Ultrasonic Therapy/methods , ATP-Binding Cassette Transporters/metabolism , Biological Transport/physiology , Drug Administration Routes , Endothelial Cells/metabolism , Exosomes/metabolism , Humans , Tight Junctions/metabolism
4.
Theranostics ; 9(13): 3754-3767, 2019.
Article in English | MEDLINE | ID: mdl-31281511

ABSTRACT

Intracellular deposits of pathological tau are the hallmark of a broad spectrum of neurodegenerative disorders collectively known as tauopathies, with Alzheimer's disease, a secondary tauopathy, being further characterized by extracellular amyloid plaques. A major obstacle in developing effective treatments for tauopathies is the presence of the blood-brain barrier, which restricts the access of therapeutic agents to the brain. An emerging technology to overcome this limitation is the application of low-intensity ultrasound which, together with intravenously injected microbubbles, transiently opens the blood-brain barrier, thereby facilitating the delivery of therapeutic agents into the brain. Interestingly, even in the absence of therapeutic agents, ultrasound has previously been shown to reduce amyloid plaques and improve cognitive functions in amyloid-depositing mice through microglial clearance. Ultrasound has also been shown to facilitate the delivery of antibody fragments against pathological tau in P301L tau transgenic mice; however, the effect of ultrasound alone has not been thoroughly investigated in a tauopathy mouse model. Methods: Here, we performed repeated scanning ultrasound treatments over a period of 15 weeks in K369I tau transgenic mice with an early-onset tau-related motor and memory phenotype. We used immunohistochemical and biochemical methods to analyze the effect of ultrasound on the mice and determine the underlying mechanism of action, together with an analysis of their motor and memory functions following repeated ultrasound treatments. Results: Repeated ultrasound treatments significantly reduced tau pathology in the absence of histological damage. Associated impaired motor functions showed improvement towards the end of the treatment regime, with memory functions showing a trend towards improvement. In assessing potential clearance mechanisms, we ruled out a role for ubiquitination of tau, a prerequisite for proteasomal clearance. However, the treatment regime induced the autophagy pathway in neurons as reflected by an increase in the autophagosome membrane marker LC3II and a reduction in the autophagic flux marker p62, along with a decrease of mTOR activity and an increase in beclin 1 levels. Moreover, there was a significant increase in the interaction of tau and p62 in the ultrasound-treated mice, suggesting removal of tau by autophagosomes. Conclusions: Our findings indicate that a neuronal protein aggregate clearance mechanism induced by ultrasound-mediated blood-brain barrier opening operates for tau, further supporting the potential of low-intensity ultrasound to treat neurodegenerative disorders.


Subject(s)
Autophagy , Behavior, Animal , Neurons/metabolism , Ultrasonography , tau Proteins/metabolism , Animals , Memory , Mice, Transgenic , Motor Activity , Neurofibrillary Tangles/metabolism , Phosphorylation
5.
ACS Chem Neurosci ; 10(5): 2498-2509, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30763064

ABSTRACT

While the roles of intrinsically disordered protein domains in driving interprotein interactions are increasingly well-appreciated, the mechanism of toxicity of disease-causing disordered proteins remains poorly understood. A prime example is Alzheimer's disease (AD) associated amyloid beta (Aß). Aß oligomers are highly toxic partially structured peptide assemblies with a distinct ordered region (residues ∼10-40) and a shorter disordered region (residues ∼1-9). Here, we investigate the role of this disordered domain and its relation to the ordered domain in the manifestation of toxicity through a set of Aß fragments and stereoisomers designed for this purpose. We measure their effects on lipid membranes and cultured neurons, probing their toxicity, intracellular distributions, and specific molecular interactions using the techniques of confocal imaging, lattice light sheet imaging, fluorescence lifetime imaging, and fluorescence correlation spectroscopy. Remarkably, we find that neither part-Aß10-40 or Aß1-9, is toxic by itself. The ordered part (Aß10-40) is the major determinant of how Aß attaches to lipid bilayers, enters neuronal cells, and localizes primarily in the late endosomal compartments. However, once Aß enters the cell, it is the disordered part (only when it is connected to the rest of the peptide) that has a strong and stereospecific interaction with an unknown cellular component, as demonstrated by distinct changes in the fluorescence lifetime of a fluorophore attached to the N-terminal. This interaction appears to commit Aß to the toxic pathway. Our findings correlate well with Aß sites of familial AD mutations, a significant fraction of which cluster in the disordered region. We conclude that, while the ordered region dictates attachment and cellular entry, the key to toxicity lies in the ordered part presenting the disordered part for a specific cellular interaction.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/physiology , Membrane Lipids/metabolism , Neurons/physiology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Animals , Cells, Cultured , Female , Isomerism , Rats, Wistar
6.
Theranostics ; 8(22): 6233-6247, 2018.
Article in English | MEDLINE | ID: mdl-30613294

ABSTRACT

The blood-brain barrier presents a major challenge for the delivery of therapeutic agents to the brain; however, it can be transiently opened by combining low intensity ultrasound with microbubble infusion. Studies evaluating this technology have largely been performed in rodents, including models of neurological conditions. However, despite promising outcomes in terms of drug delivery and the amelioration of neurological impairments, the potential for long-term adverse effects presents a major concern in the context of clinical applications. Methods: To fill this gap, we repeatedly treated 12-month-old wild-type mice with ultrasound, followed by a multimodal analysis for up to 18 months of age. Results: We found that spatial memory in these aged mice was not adversely affected as assessed in the active place avoidance test. Sholl analysis of Golgi impregnations in the dentate gyrus of the hippocampus did not reveal any changes to the neuronal cytoarchitecture. Long-term potentiation, a cellular correlate of memory, was still achievable, magnetic resonance spectroscopy revealed no major changes in metabolites, and diffusion tensor imaging revealed normal microstructure and tissue integrity in the hippocampus. More specifically, all measures of diffusion appeared to support a neuroprotective effect of ultrasound treatment on the brain. Conclusion: This multimodal analysis indicates that therapeutic ultrasound for blood-brain barrier opening is safe and potentially protective in the long-term, underscoring its validity as a potential treatment modality for diseases of the brain.


Subject(s)
Aging/radiation effects , Brain/radiation effects , Aging/physiology , Animals , Brain/diagnostic imaging , Brain/physiology , Female , Hippocampus/diagnostic imaging , Hippocampus/physiology , Hippocampus/radiation effects , Mice , Mice, Inbred C57BL , Multimodal Imaging/adverse effects , Spatial Memory/radiation effects , Time Factors , Ultrasonic Therapy/adverse effects , Ultrasonography
7.
ACS Chem Neurosci ; 6(8): 1290-5, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-25951510

ABSTRACT

Small hydrophobic oligomers of aggregation-prone proteins are thought to be generically toxic. Here we examine this view by perturbing an early folding contact between Phe19 and Leu34 formed during the aggregation of Alzheimer's amyloid-ß (Aß40) peptide. We find that even conservative single mutations altering this interaction can abolish Aß40 toxicity. Significantly, the mutants are not distinguishable either by the oligomers size or by the end-state fibrillar structure from the wild type Aß40. We trace the change in their toxicity to a drastic lowering of membrane affinity. Therefore, nonlocal folding contacts play a key role in steering the oligomeric intermediates through specific conformations with very different properties and toxicity levels. Our results suggest that engineering the folding energy landscape may provide an alternative route to Alzheimer therapeutics.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Amyloid beta-Peptides/toxicity , Animals , Cell Survival , Cells, Cultured , Cerebral Cortex/physiology , Membranes, Artificial , Mutation , Neurons/physiology , Peptide Fragments/toxicity , Phosphatidylcholines/chemistry , Phosphatidylglycerols/chemistry , Protein Folding , Rats, Wistar , Unilamellar Liposomes/chemistry
8.
Philos Trans R Soc Lond B Biol Sci ; 370(1672)2015 Jul 05.
Article in English | MEDLINE | ID: mdl-26009766

ABSTRACT

Certain neurodegenerative diseases are thought to be initiated by the aggregation of amyloidogenic proteins. However, the mechanism underlying toxicity remains obscure. Most of the suggested mechanisms are generic in nature and do not directly explain the neuron-type specific lesions observed in many of these diseases. Some recent reports suggest that the toxic aggregates impair the synaptic vesicular machinery. This may lead to an understanding of the neuron-type specificity observed in these diseases. A disruption of the vesicular machinery can also be deleterious for extra-synaptic, especially somatic, neurotransmission (common in serotonergic and dopaminergic systems which are specifically affected in Alzheimer's disease (AD) and Parkinson's disease (PD), respectively), though this relationship has remained unexplored. In this review, we discuss amyloid-induced damage to the neurotransmitter vesicular machinery, with an eye on the possible implications for somatic exocytosis. We argue that the larger size of the system, and the availability of multi-photon microscopy techniques for directly visualizing monoamines, make the somatic exocytosis machinery a more tractable model for understanding the effect of amyloids on all types of vesicular neurotransmission. Indeed, exploring this neglected connection may not just be important, it may be a more fruitful route for understanding AD and PD.


Subject(s)
Amyloid/toxicity , Cell Body/metabolism , Neurodegenerative Diseases/physiopathology , Neurons/metabolism , Synaptic Transmission/physiology , Synaptic Vesicles/drug effects , Exocytosis/physiology , Humans , Neurons/cytology , Synaptic Transmission/drug effects
9.
J Ethnopharmacol ; 128(2): 462-6, 2010 Mar 24.
Article in English | MEDLINE | ID: mdl-20080167

ABSTRACT

AIMS OF STUDY: In Indian traditional system of medicine, Ficus religiosa (Family Moraceae) is prescribed for the treatment of diabetes mellitus. In the present study, the antidiabetic effect of aqueous extract of Ficus religiosa bark (FRAE) was investigated in normal, glucose-loaded hyperglycemic and streptozotocin (STZ)-induced diabetic rats. MATERIALS AND METHODS: Oral administration of FRAE at the doses of 25, 50 and 100mg/kg was studied in normal, glucose-loaded and STZ-diabetic rats. RESULTS: The three doses caused significant reduction in blood glucose levels in all the models. The effect was more pronounced in 50 and 100mg/kg than 25mg/kg. FRAE also showed significant increase in serum insulin, body weight and glycogen content in liver and skeletal muscle of STZ-induced diabetic rats while there was significant reduction in the levels of serum triglyceride and total cholesterol. FRAE also showed significant antilipidperoxidative effect in the pancreas of STZ-induced diabetic rats. The antidiabetic effect of Ficus religiosa was compared with glibenclamide, a well-known hypoglycemic drug. CONCLUSION: The results indicate that aqueous extract of Ficus religiosa bark possesses significant antidiabetic activity.


Subject(s)
Diabetes Mellitus/drug therapy , Ficus , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Streptozocin/adverse effects , Administration, Oral , Animals , Body Weight/drug effects , Cholesterol/pharmacology , Glyburide/pharmacology , Glycogen/pharmacology , Hypoglycemic Agents/administration & dosage , Insulin/adverse effects , Liver/drug effects , Male , Pancreas/drug effects , Rats , Rats, Wistar , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...