Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (129)

Search Parameters:
Keywords = brain biodistribution

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
29 pages, 3008 KiB  
Review
Small Extracellular Vesicles in Neurodegenerative Disease: Emerging Roles in Pathogenesis, Biomarker Discovery, and Therapy
by Mousumi Ghosh, Amir-Hossein Bayat and Damien D. Pearse
Int. J. Mol. Sci. 2025, 26(15), 7246; https://doi.org/10.3390/ijms26157246 - 26 Jul 2025
Viewed by 298
Abstract
Neurodegenerative diseases (NDDs) such as Alzheimer’s, Parkinson’s, ALS, and Huntington’s pose a growing global challenge due to their complex pathobiology and aging demographics. Once considered as cellular debris, small extracellular vesicles (sEVs) are now recognized as active mediators of intercellular signaling in NDD [...] Read more.
Neurodegenerative diseases (NDDs) such as Alzheimer’s, Parkinson’s, ALS, and Huntington’s pose a growing global challenge due to their complex pathobiology and aging demographics. Once considered as cellular debris, small extracellular vesicles (sEVs) are now recognized as active mediators of intercellular signaling in NDD progression. These nanovesicles (~30–150 nm), capable of crossing the blood–brain barrier, carry pathological proteins, RNAs, and lipids, facilitating the spread of toxic species like Aβ, tau, TDP-43, and α-synuclein. sEVs are increasingly recognized as valuable diagnostic tools, outperforming traditional CSF biomarkers in early detection and disease monitoring. On the therapeutic front, engineered sEVs offer a promising platform for CNS-targeted delivery of siRNAs, CRISPR tools, and neuroprotective agents, demonstrating efficacy in preclinical models. However, translational hurdles persist, including standardization, scalability, and regulatory alignment. Promising solutions are emerging, such as CRISPR-based barcoding, which enables high-resolution tracking of vesicle biodistribution; AI-guided analytics to enhance quality control; and coordinated regulatory efforts by the FDA, EMA, and ISEV aimed at unifying identity and purity criteria under forthcoming Minimal Information for Studies of Extracellular Vesicles (MISEV) guidelines. This review critically examines the mechanistic roles, diagnostic potential, and therapeutic applications of sEVs in NDDs, and outlines key strategies for clinical translation. Full article
(This article belongs to the Special Issue Molecular Advances in Neurologic and Neurodegenerative Disorders)
Show Figures

Graphical abstract

18 pages, 741 KiB  
Review
Cardiovascular Toxicity of Metal-Based Nanoparticles
by Eun-Hye Kim, Sehyeon Park and Ok-Nam Bae
Int. J. Mol. Sci. 2025, 26(12), 5816; https://doi.org/10.3390/ijms26125816 - 17 Jun 2025
Viewed by 788
Abstract
The rapid development of nanotechnology has led to increased human exposure to metal-based nanoparticles (MNPs) through inhalation, ingestion, and dermal contact, raising growing concerns on their potential health effects. Due to their nanoscale size and unique physicochemical properties, the MNPs can translocate from [...] Read more.
The rapid development of nanotechnology has led to increased human exposure to metal-based nanoparticles (MNPs) through inhalation, ingestion, and dermal contact, raising growing concerns on their potential health effects. Due to their nanoscale size and unique physicochemical properties, the MNPs can translocate from the initial exposure sites to the circulatory system and accumulate in the body. This review focuses on MNP-induced cardiovascular toxicity, highlighting its biodistribution, cytotoxic mechanisms, and pathological impact associated with various cardiovascular diseases. MNPs disrupt endothelial function, promote oxidative stress, and induce apoptosis and ferroptosis in cardiovascular cells. Furthermore, MNPs increase endothelial permeability, impair blood–brain barrier integrity, and enhance procoagulant activity, thereby contributing to vascular and cardiac dysfunction. The particles and their released metal ions play a synergistic role in mediating these toxic effects. Here, we focused on the effects of nano-sized particles while incorporating recent in vitro and in vivo studies that address the cardiovascular impacts and mechanisms of MNP-induced toxicity. This comprehensive review will help understand and explain the potentially toxic effects of MNPs on the cardiovascular system. Full article
Show Figures

Figure 1

19 pages, 3804 KiB  
Article
Peptide-Engineered Seliciclib Nanomedicine for Brain-Targeted Delivery and Neuroprotection
by Guan Zhen He and Wen Jen Lin
Int. J. Mol. Sci. 2025, 26(12), 5768; https://doi.org/10.3390/ijms26125768 - 16 Jun 2025
Viewed by 326
Abstract
Seliciclib, a cyclin-dependent kinase 5 (CDK5) inhibitor, has demonstrated neuroprotective potential. However, its therapeutic application is limited by poor permeability across the blood–brain barrier (BBB). In this study, polymeric nanoparticles (NPs) modified with a BBB-targeting peptide ligand (His-Ala-Ile-Tyr-Pro-Arg-His) were employed to encapsulate seliciclib. [...] Read more.
Seliciclib, a cyclin-dependent kinase 5 (CDK5) inhibitor, has demonstrated neuroprotective potential. However, its therapeutic application is limited by poor permeability across the blood–brain barrier (BBB). In this study, polymeric nanoparticles (NPs) modified with a BBB-targeting peptide ligand (His-Ala-Ile-Tyr-Pro-Arg-His) were employed to encapsulate seliciclib. In vitro transport studies showed that the peptide-modified NPs exhibited significantly greater translocation across a bEnd.3 cell monolayer compared to unmodified NPs. Furthermore, in vivo biodistribution analysis revealed that the brain accumulation of peptide-modified NPs was 3.38-fold higher than that of unmodified NPs. Notably, the peptide-conjugated, seliciclib-loaded NPs demonstrated a significant neuroprotective effect against the neurotoxin 1-methyl-4-phenylpyridinium (MPP⁺) in differentiated SH-SY5Y cells. Full article
(This article belongs to the Special Issue Multifunctional Nanocomposites for Bioapplications)
Show Figures

Figure 1

15 pages, 4089 KiB  
Article
Increased [18F]DPA-714 Uptake in the Skeletal Muscle of SOD1G93A Mice: A New Potential of Translocator Protein 18 kDa Imaging in Amyotrophic Lateral Sclerosis
by Cecilia Marini, Mattia Riondato, Edoardo Dighero, Alessia Democrito, Serena Losacco, Laura Emionite, Lucilla Nobbio, Irene Di Patrizi, Mattia Camera, Chiara Ghersi, Maddalena Ghelardoni, Francesco Lanfranchi, Francesca Vitale, Sonia Carta, Sabrina Chiesa, Carola Torazza, Marco Milanese, Matteo Bauckneht, Mehrnaz Hamedani, Federico Zaottini, Angelo Schenone, Carlo Martinoli, Federica Grillo and Gianmario Sambucetiadd Show full author list remove Hide full author list
Biomolecules 2025, 15(6), 799; https://doi.org/10.3390/biom15060799 - 31 May 2025
Viewed by 583
Abstract
Purpose: The skeletal muscle has been proposed to contribute to the progressive loss of motor neurons typical of amyotrophic lateral sclerosis (ALS). However, this mechanism has not yet been clarified due to the lack of suitable imaging tools. Here, we aimed to verify [...] Read more.
Purpose: The skeletal muscle has been proposed to contribute to the progressive loss of motor neurons typical of amyotrophic lateral sclerosis (ALS). However, this mechanism has not yet been clarified due to the lack of suitable imaging tools. Here, we aimed to verify whether PET imaging of the translocator protein 18 kDa (TSPO) can detect a muscular abnormality in an experimental model of ALS. Methods: In vivo biodistribution and kinetics of [18F]DPA-714 were analyzed in skeletal muscle and brain of SOD1G93A transgenic mice and in wildtype (WT) littermates. Both cohorts were divided into three groups (n = 6 each) to be studied at 60, 90 and 120 days. After microPET imaging, animals were sacrificed to evaluate inflammatory infiltrates by hematoxylin/eosin staining and TSPO expression by immunohistochemistry and Western blot in both quadriceps and brain. Results: [18F]DPA-714 uptake was higher in the skeletal muscles of SOD1G93A than in WT mice in the preclinical phase (60 and 90 days) and further increased up to the symptomatic late stage (120 days). Inflammatory cells were absent in the quadriceps of SOD1G93A mice whose myocytes, instead, showed a progressive increase in TSPO expression with advancing age. By contrast, brain tracer uptake and TSPO expression were comparably low in both groups, regardless of age and genotype. Conclusion: Upregulation of TSPO expression is characteristic of skeletal muscle, but not the brain, in the experimental SOD1G93A mouse model of ALS. Tracers targeting this pathway have been mostly proposed for the evaluation of inflammatory processes within the central nervous system. Nevertheless, the ubiquitous nature of TSPO expression and its responsiveness to various signals may broaden the diagnostic potential of these tracers to include disease conditions beyond inflammation. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Graphical abstract

19 pages, 7094 KiB  
Article
Biodistribution of Polyaldehydedextran Nanoparticle-Encapsulated Epirubicin in Ovarian Tumor-Bearing Mice via Optical Imaging
by Wioletta Kośnik, Hanna Sikorska, Adam Kiciak and Tomasz Ciach
Int. J. Mol. Sci. 2025, 26(3), 970; https://doi.org/10.3390/ijms26030970 - 24 Jan 2025
Viewed by 1199
Abstract
This study investigates the biodistribution of polysaccharide-based nanoparticles loaded with epirubicin (POLEPI) compared to epirubicin hydrochloride (EPI) in naïve female nude mice following a single intravenous dose. The inherent fluorescence of epirubicin was tracked using Newton 7 animal imager and Varioskan. Initial whole-animal [...] Read more.
This study investigates the biodistribution of polysaccharide-based nanoparticles loaded with epirubicin (POLEPI) compared to epirubicin hydrochloride (EPI) in naïve female nude mice following a single intravenous dose. The inherent fluorescence of epirubicin was tracked using Newton 7 animal imager and Varioskan. Initial whole-animal optical imaging failed to reliably detect epirubicin distribution, necessitating ex vivo imaging of key tissues harvested at intervals between 10 min and 48 h post-injection. Optimal imaging conditions were established using a 5 s exposure time with excitation (Ex)/emission (Em) at 480 nm/550 nm. The biodistribution of POLEPI was further evaluated in both naïve mice and immunocompromised mice bearing patient-derived ovarian tumors. Unlike epirubicin, POLEPI exhibited notable tissue distribution within 3 h post-injection. By 48 h, fluorescence signals were undetectable in both models, although non-tumored animals exhibited persistent signals. In both models, the liver was the primary organ for POLEPI accumulation, with lower levels in tumored mice. Interestingly, brain fluorescence was higher in POLEPI-treated mice compared to those receiving epirubicin. Neither POLEPI nor epirubicin accumulated in the spleen or bone marrow. In tumors, POLEPI fluorescence peaked at 24 h, with levels 2.1 times higher than in the epirubicin-treated group over a 48 h period. Furthermore, POLEPI uptake in tumors exceeded that in healthy ovaries, with the most significant tumor-to-healthy-ovary ratio observed between 6 and 24 h post-injection. These findings demonstrate that POLEPI, a novel polyaldehydedextran nanoparticle formulation, exhibits enhanced accumulation and retention in tumor tissue compared to epirubicin, with preferential distribution to the orthotopic tumor-bearing ovary over healthy ovarian tissue. The inherent fluorescence of epirubicin provided a rapid and cost-effective means of estimating biodistribution, although the limitations of this method—particularly, the inability to differentiate between the parent drug and its metabolites—were acknowledged. Full article
Show Figures

Figure 1

15 pages, 4504 KiB  
Article
Optimization of Transcardiac Perfusion for More Accurately Evaluating Biodistribution of Large Molecules
by Zuoxu Xie, Annie Guo and Ekta Kadakia
Int. J. Mol. Sci. 2024, 25(22), 12180; https://doi.org/10.3390/ijms252212180 - 13 Nov 2024
Cited by 1 | Viewed by 1913
Abstract
The accurate assessment of drug concentrations in biodistribution studies is crucial for evaluating the efficacy and toxicity of compounds in drug development. As the concentration of biologics in plasma can be higher than in tissue due to their potentially low volume of distribution, [...] Read more.
The accurate assessment of drug concentrations in biodistribution studies is crucial for evaluating the efficacy and toxicity of compounds in drug development. As the concentration of biologics in plasma can be higher than in tissue due to their potentially low volume of distribution, transcardiac perfusion is commonly employed to reduce the influence of excess drugs in residual blood. However, there is a lack of consistency in the literature on the conditions and methods of perfusion. To enhance blood removal during transcardiac perfusion, sodium nitrite (NaNO2), a vasodilator, has been widely used with concentrations up to 5% in publications. However, we found that such high NaNO2 could disrupt the BBB during perfusion, which should be avoided in experiments. In this study, we examined the impact of various vasodilators on blood–brain barrier integrity and vascular permeability using the ratio of FITC-Dextran to Texas Red-Dextran (FITC/Texas Red). Additionally, we optimized perfusion conditions—including euthanasia method and perfusion flow rate—based on hemoglobin levels and the FITC/Texas Red ratio in tissues. Despite the superiority of NaNO2 in terms of solubility and cost over other vasodilators, we found that 2% NaNO2 disrupted blood–brain barrier integrity, significantly altering the FITC/Texas Red ratio. In contrast, 100 mM NaNO2 did not significantly affect this ratio. Moreover, under Ketamine/Xylazine (Ket/Xyl) anesthesia, which reduced blood clot formation compared to CO2 euthanasia, 100 mM NaNO2 achieved the lowest hemoglobin levels in the brain. Compared to other vasodilators and the PBS control group, 100 mM NaNO2 decreased the tissue/plasma ratio (Kp,t) but not brain/plasma ratio (Kp,b) of hIgG1 and human transferrin. We have developed a method to efficiently evaluate blood–brain barrier integrity during transcardiac perfusion. The combination of Ket/Xyl anesthesia and 100 mM NaNO2 effectively removes residual blood from tissues without significantly affecting blood vessel permeability. Full article
(This article belongs to the Section Macromolecules)
Show Figures

Figure 1

24 pages, 519 KiB  
Article
Acute Biodistribution Comparison of Fentanyl and Morphine
by Rosamond Goodson, Justin Poklis, Harrison J. Elder, D. Matthew Walentiny, William Dewey and Matthew Halquist
Psychoactives 2024, 3(4), 437-460; https://doi.org/10.3390/psychoactives3040027 - 26 Sep 2024
Viewed by 3132
Abstract
Synthetic opioids such as fentanyl are key drivers of the opioid crisis, contributing to approximately 68% of the nearly 108,000 deaths linked to drug overdose in 2022 (CDC). Though fentanyl is a μ opioid receptor agonist, it demonstrates enhanced lipophilicity, heightened potency to [...] Read more.
Synthetic opioids such as fentanyl are key drivers of the opioid crisis, contributing to approximately 68% of the nearly 108,000 deaths linked to drug overdose in 2022 (CDC). Though fentanyl is a μ opioid receptor agonist, it demonstrates enhanced lipophilicity, heightened potency to induce respiratory depression, and more rapid central nervous system entry compared to certain other opioids, i.e., morphine. However, there are relatively few biodistribution comparison studies of fentanyl and classical opioids like morphine in mice, despite the use of mice as preclinical models of opioid effects, i.e., respiratory depression. Therefore, the current study compared acute fentanyl (0.3 mg/kg) and morphine (30 mg/kg) biodistribution in blood and 12 tissues at doses causing respiratory depression in male Swiss Webster mice. Whole-body plethysmography was used to select fentanyl and morphine doses producing comparable respiratory depression, and an LC/MS-MS protocol was developed to quantify fentanyl, morphine, and metabolites in diverse tissue samples. Drug distribution time courses varied by tissue, with fentanyl and morphine displaying similar time courses in the lung, stomach, and small intestine, but differing in the brain and spleen. Fentanyl exhibited greater distribution out of the blood and into the brain, liver, lung, and heart than morphine early after administration and out of the blood into fat at later time points after administration. The ratios of total drug distribution (area under the curve) in tissue–blood over time suggest that fentanyl accumulation in tissue relative to blood in several areas, such as lung, heart, kidney, spleen, fat, and small intestine, is greater than morphine. These findings indicate that fentanyl administration may affect several organs to a larger degree than morphine. Full article
Show Figures

Figure 1

28 pages, 8501 KiB  
Article
The Bifunctional Dimer Caffeine-Indan Attenuates α-Synuclein Misfolding, Neurodegeneration and Behavioral Deficits after Chronic Stimulation of Adenosine A1 Receptors
by Elisabet Jakova, Omozojie P. Aigbogun, Mohamed Taha Moutaoufik, Kevin J. H. Allen, Omer Munir, Devin Brown, Changiz Taghibiglou, Mohan Babu, Chris P. Phenix, Ed S. Krol and Francisco S. Cayabyab
Int. J. Mol. Sci. 2024, 25(17), 9386; https://doi.org/10.3390/ijms25179386 - 29 Aug 2024
Cited by 2 | Viewed by 1933
Abstract
We previously found that chronic adenosine A1 receptor stimulation with N6-Cyclopentyladenosine increased α-synuclein misfolding and neurodegeneration in a novel α-synucleinopathy model, a hallmark of Parkinson’s disease. Here, we aimed to synthesize a dimer caffeine-indan linked by a 6-carbon chain to cross [...] Read more.
We previously found that chronic adenosine A1 receptor stimulation with N6-Cyclopentyladenosine increased α-synuclein misfolding and neurodegeneration in a novel α-synucleinopathy model, a hallmark of Parkinson’s disease. Here, we aimed to synthesize a dimer caffeine-indan linked by a 6-carbon chain to cross the blood–brain barrier and tested its ability to bind α-synuclein, reducing misfolding, behavioral abnormalities, and neurodegeneration in our rodent model. Behavioral tests and histological stains assessed neuroprotective effects of the dimer compound. A rapid synthesis of the 18F-labeled analogue enabled Positron Emission Tomography and Computed Tomography imaging for biodistribution measurement. Molecular docking analysis showed that the dimer binds to α-synuclein N- and C-termini and the non-amyloid-β-component (NAC) domain, similar to 1-aminoindan, and this binding promotes a neuroprotective α-synuclein “loop” conformation. The dimer also binds to the orthosteric binding site for adenosine within the adenosine A1 receptor. Immunohistochemistry and confocal imaging showed the dimer abolished α-synuclein upregulation and aggregation in the substantia nigra and hippocampus, and the dimer mitigated cognitive deficits, anxiety, despair, and motor abnormalities. The 18F-labeled dimer remained stable post-injection and distributed in various organs, notably in the brain, suggesting its potential as a Positron Emission Tomography tracer for α-synuclein and adenosine A1 receptor in Parkinson’s disease therapy. Full article
Show Figures

Figure 1

23 pages, 7257 KiB  
Article
Green Solid Lipid Nanoparticles by Fatty Acid Coacervation: An Innovative Nasal Delivery Tool for Drugs Targeting Cerebrovascular and Neurological Diseases
by Annalisa Bozza, Valentina Bordano, Arianna Marengo, Elisabetta Muntoni, Elisabetta Marini, Loretta Lazzarato, Chiara Dianzani, Chiara Monge, Arianna Carolina Rosa, Luigi Cangemi, Maria Carmen Valsania, Barbara Colitti, Ezio Camisassa and Luigi Battaglia
Pharmaceutics 2024, 16(8), 1051; https://doi.org/10.3390/pharmaceutics16081051 - 8 Aug 2024
Cited by 2 | Viewed by 2066
Abstract
Cerebrovascular and neurological diseases are characterized by neuroinflammation, which alters the neurovascular unit, whose interaction with the choroid plexus is critical for maintaining brain homeostasis and producing cerebrospinal fluid. Dysfunctions in such process can lead to conditions such as idiopathic normal pressure hydrocephalus, [...] Read more.
Cerebrovascular and neurological diseases are characterized by neuroinflammation, which alters the neurovascular unit, whose interaction with the choroid plexus is critical for maintaining brain homeostasis and producing cerebrospinal fluid. Dysfunctions in such process can lead to conditions such as idiopathic normal pressure hydrocephalus, a common disease in older adults. Potential pharmacological treatments, based upon intranasal administration, are worthy of investigation because they might improve symptoms and avoid surgery by overcoming the blood–brain barrier and avoiding hepatic metabolism. Nasal lipid nanocarriers, such as solid lipid nanoparticles, may increase the nasal retention and permeation of drugs. To this aim, green solid lipid nanoparticles, obtained by coacervation from natural soaps, are promising vehicles due to their specific lipid matrix composition and the unsaponifiable fraction, endowed with antioxidant and anti-inflammatory properties, and thus suitable for restoring the neurovascular unit function. In this experimental work, such green solid lipid nanoparticles, fully characterized from a physico-chemical standpoint, were loaded with a drug combination suitable for reverting hydrocephalus symptoms, allowing us to obtain a non-toxic formulation, a reduction in the production of the cerebrospinal fluid in vitro, and a vasoprotective effect on an isolated vessel model. The pharmacokinetics and biodistribution of fluorescently labelled nanoparticles were also tested in animal models. Full article
(This article belongs to the Special Issue Advances in Nanotechnology-Based Drug Delivery Systems)
Show Figures

Figure 1

12 pages, 4240 KiB  
Article
Labeling of Bruton’s Tyrosine Kinase (BTK) Inhibitor [11C]BIO-2008846 in Three Different Positions and Measurement in NHP Using PET
by Sangram Nag, Prodip Datta, Anton Forsberg Morén, Yasir Khani, Laurent Martarello, Maciej Kaliszczak and Christer Halldin
Int. J. Mol. Sci. 2024, 25(14), 7870; https://doi.org/10.3390/ijms25147870 - 18 Jul 2024
Viewed by 1334
Abstract
Bruton’s tyrosine kinase (BTK) is pivotal in B-cell signaling and a target for potential anti-cancer and immunological disorder therapies. Improved selective reversible BTK inhibitors are in demand due to the absence of direct BTK engagement measurement tools. Promisingly, PET imaging can non-invasively evaluate [...] Read more.
Bruton’s tyrosine kinase (BTK) is pivotal in B-cell signaling and a target for potential anti-cancer and immunological disorder therapies. Improved selective reversible BTK inhibitors are in demand due to the absence of direct BTK engagement measurement tools. Promisingly, PET imaging can non-invasively evaluate BTK expression. In this study, radiolabeled BIO-2008846 ([11C]BIO-2008846-A), a BTK inhibitor, was used for PET imaging in NHPs to track brain biodistribution. Radiolabeling BIO-2008846 with carbon-11, alongside four PET scans on two NHPs each, showed a homogeneous distribution of [11C]BIO-2008846-A in NHP brains. Brain uptake ranged from 1.8% ID at baseline to a maximum of 3.2% post-pretreatment. The study found no significant decrease in regional VT values post-dose, implying minimal specific binding of [11C]BIO-2008846-A compared to free and non-specific components in the brain. Radiometabolite analysis revealed polar metabolites with 10% unchanged radioligand after 30 min. The research highlighted strong brain uptake despite minor distribution variability, confirming passive diffusion kinetics dominated by free and non-specific binding. Full article
(This article belongs to the Special Issue Diagnostic Tools for Neuropsychological Disorders)
Show Figures

Figure 1

20 pages, 2378 KiB  
Article
Numerical Study towards In Vivo Tracking of Micro-/Nanoplastic Based on X-ray Fluorescence Imaging
by Carolin von der Osten-Sacken, Theresa Staufer, Kai Rothkamm, Robert Kuhrwahl and Florian Grüner
Biomedicines 2024, 12(7), 1500; https://doi.org/10.3390/biomedicines12071500 - 5 Jul 2024
Cited by 2 | Viewed by 1669
Abstract
There is a rising awareness of the toxicity of micro- and nanoplastics (MNPs); however, fundamental precise information on MNP-biodistribution in organisms is currently not available. X-ray fluorescence imaging (XFI) is introduced as a promising imaging modality to elucidate the effective MNP bioavailability and [...] Read more.
There is a rising awareness of the toxicity of micro- and nanoplastics (MNPs); however, fundamental precise information on MNP-biodistribution in organisms is currently not available. X-ray fluorescence imaging (XFI) is introduced as a promising imaging modality to elucidate the effective MNP bioavailability and is expected to enable exact measurements on the uptake over the physical barriers of the organism and bioaccumulation in different organs. This is possible because of the ability of XFI to perform quantitative studies with a high spatial resolution and the possibility to conduct longitudinal studies. The focus of this work is a numerical study on the detection limits for a selected XFI-marker, here, palladium, to facilitate the design of future preclinical in vivo studies. Based on Monte Carlo simulations using a 3D voxel mouse model, the palladium detection thresholds in different organs under in vivo conditions in a mouse are estimated. The minimal Pd-mass in the scanning position at a reasonable significance level is determined to be <20 ng/mm2 for abdominal organs and <16 μg/mm2 for the brain. MNPs labelled with Pd and homogeneously distributed in the organ would be detectable down to a concentration of <1 μg/mL to <2.5 mg/mL in vivo. Long-term studies with a chronic MNP exposure in low concentrations are therefore possible such that XFI measurements could, in the future, contribute to MNP health risk assessment in small animals and humans. Full article
(This article belongs to the Section Biomedical Engineering and Materials)
Show Figures

Figure 1

15 pages, 3500 KiB  
Article
Magnet-Guided Temozolomide and Ferucarbotran Loaded Nanoparticles to Enhance Therapeutic Efficacy in Glioma Model
by Reju George Thomas, Subin Kim, Thi-Anh-Thuy Tran, Young Hee Kim, Raveena Nagareddy, Tae-Young Jung, Seul Kee Kim and Yong Yeon Jeong
Nanomaterials 2024, 14(11), 939; https://doi.org/10.3390/nano14110939 - 27 May 2024
Cited by 4 | Viewed by 1757
Abstract
Background. The aim of the study was to synthesize liposomal nanoparticles loaded with temozolomide and ferucarbotran (LTF) and to evaluate the theranostic effect of LTF in the glioma model. Methods. We synthesized an LTF that could pass through the Blood Brain Barrier (BBB) [...] Read more.
Background. The aim of the study was to synthesize liposomal nanoparticles loaded with temozolomide and ferucarbotran (LTF) and to evaluate the theranostic effect of LTF in the glioma model. Methods. We synthesized an LTF that could pass through the Blood Brain Barrier (BBB) and localize in brain tumor tissue with the help of magnet guidance. We examined the chemical characteristics. Cellular uptake and cytotoxicity studies were conducted in vitro. A biodistribution and tumor inhibition study was conduted using an in vivo glioma model. Results. The particle size and surface charge of LTF show 108 nm and −38 mV, respectively. Additionally, the presence of ferucarbotran significantly increased the contrast agent effect of glioma compared to the control group in MR imaging. Magnet-guided LTF significantly reduced the tumor size compared to control and other groups. Furthermore, compared to the control group, our results demonstrate a significant inhibition in brain tumor size and an increase in lifespan. Conclusions. These findings suggest that the LTF with magnetic guidance represents a novel approach to address current obstacles, such as BBB penetration of nanoparticles and drug resistance. Magnet-guided LTF is able to enhance therapeutic efficacy in mouse brain glioma. Full article
Show Figures

Graphical abstract

15 pages, 3048 KiB  
Article
PET Imaging of Neurofibromatosis Type 1 with a Fluorine-18 Labeled Tryptophan Radiotracer
by Xuyi Yue, Erik Stauff, Shriya Boyapati, Sigrid A. Langhans, Wenqi Xu, Sokratis Makrogiannis, Uchenna J. Okorie, Azubuike M. Okorie, Vinay V. R. Kandula, Heidi H. Kecskemethy, Rahul M. Nikam, Lauren W. Averill and Thomas H. Shaffer
Pharmaceuticals 2024, 17(6), 685; https://doi.org/10.3390/ph17060685 - 27 May 2024
Cited by 1 | Viewed by 2064
Abstract
Neurofibromatosis type 1 (NF1) is a neurocutaneous disorder. Plexiform neurofibromas (PNFs) are benign tumors commonly formed in patients with NF1. PNFs have a high incidence of developing into malignant peripheral nerve sheath tumors (MPNSTs) with a 5-year survival rate of only 30%. Therefore, [...] Read more.
Neurofibromatosis type 1 (NF1) is a neurocutaneous disorder. Plexiform neurofibromas (PNFs) are benign tumors commonly formed in patients with NF1. PNFs have a high incidence of developing into malignant peripheral nerve sheath tumors (MPNSTs) with a 5-year survival rate of only 30%. Therefore, the accurate diagnosis and differentiation of MPNSTs from benign PNFs are critical to patient management. We studied a fluorine-18 labeled tryptophan positron emission tomography (PET) radiotracer, 1-(2-[18F]fluoroethyl)-L-tryptophan (L-[18F]FETrp), to detect NF1-associated tumors in an animal model. An ex vivo biodistribution study of L-[18F]FETrp showed a similar tracer distribution and kinetics between the wild-type and triple mutant mice with the highest uptake in the pancreas. Bone uptake was stable. Brain uptake was low during the 90-min uptake period. Static PET imaging at 60 min post-injection showed L-[18F]FETrp had a comparable tumor uptake with [1⁸F]fluorodeoxyglucose (FDG). However, L-[18F]FETrp showed a significantly higher tumor-to-brain ratio than FDG (n = 4, p < 0.05). Sixty-minute-long dynamic PET scans using the two radiotracers showed similar kidney, liver, and lung kinetics. A dysregulated tryptophan metabolism in NF1 mice was further confirmed using immunohistostaining. L-[18F]FETrp is warranted to further investigate differentiating malignant NF1 tumors from benign PNFs. The study may reveal the tryptophan–kynurenine pathway as a therapeutic target for treating NF1. Full article
(This article belongs to the Section Radiopharmaceutical Sciences)
Show Figures

Figure 1

13 pages, 3401 KiB  
Article
Oral Microbially-Induced Small Extracellular Vesicles Cross the Blood–Brain Barrier
by Mahmoud Elashiry, Angelica Carroll, Jessie Yuan, Yutao Liu, Mark Hamrick, Christopher W. Cutler, Qin Wang and Ranya Elsayed
Int. J. Mol. Sci. 2024, 25(8), 4509; https://doi.org/10.3390/ijms25084509 - 20 Apr 2024
Cited by 7 | Viewed by 3426
Abstract
Porphyromonas gingivalis (Pg) and its gingipain proteases contribute to Alzheimer’s disease (AD) pathogenesis through yet unclear mechanisms. Cellular secretion of small extracellular vesicles or exosomes (EXO) increases with aging as part of the senescence-associated secretory phenotype (SASP). We have shown that EXO isolated [...] Read more.
Porphyromonas gingivalis (Pg) and its gingipain proteases contribute to Alzheimer’s disease (AD) pathogenesis through yet unclear mechanisms. Cellular secretion of small extracellular vesicles or exosomes (EXO) increases with aging as part of the senescence-associated secretory phenotype (SASP). We have shown that EXO isolated from Pg-infected dendritic cells contain gingipains and other Pg antigens and transmit senescence to bystander gingival cells, inducing alveolar bone loss in mice in vivo. Here, EXO were isolated from the gingiva of mice and humans with/without periodontitis (PD) to determine their ability to penetrate the blood–brain barrier (BBB) in vitro and in vivo. PD was induced by Pg oral gavage for 6 weeks in C57B6 mice. EXO isolated from the gingiva or brain of donor Pg-infected (PD EXO) or control animals (Con EXO) were characterized by NTA, Western blot, and TEM. Gingival PD EXO or Con EXO were labeled and injected into the gingiva of uninfected WT mouse model. EXO biodistribution in brains was tracked by an in vivo imaging system (IVIS) and confocal microscopy. The effect of human PD EXO on BBB integrity and permeability was examined using TEER and FITC dextran assays in a human in vitro 3D model of the BBB. Pg antigens (RGP and Mfa-1) were detected in EXO derived from gingival and brain tissues of donor Pg-infected mice. Orally injected PD EXO from donor mice penetrated the brains of recipient uninfected mice and colocalized with hippocampal microglial cells. IL-1β and IL-6 were expressed in human PD EXO and not in Con EXO. Human PD EXO promoted BBB permeability and penetrated the BBB in vitro. This is the first demonstration that microbial-induced EXO in the oral cavity can disseminate, cross the BBB, and may contribute to AD pathogenesis. Full article
Show Figures

Figure 1

16 pages, 7285 KiB  
Article
Improved Chemical and Radiochemical Synthesis of Neuropeptide Y Y2 Receptor Antagonist N-Methyl-JNJ-31020028 and Preclinical Positron Emission Tomography Studies
by Inês C. F. Fonseca, Mariana Lapo Pais, Fábio M. S. Rodrigues, José Sereno, Miguel Castelo-Branco, Cláudia Cavadas, Mariette M. Pereira and Antero J. Abrunhosa
Pharmaceuticals 2024, 17(4), 474; https://doi.org/10.3390/ph17040474 - 8 Apr 2024
Viewed by 2331
Abstract
Neuropeptide Y (NPY) is one of the most abundant peptides in the central nervous system of mammals and is involved in several physiological processes through NPY Y1, Y2, Y4 and Y5 receptors. Of those, the Y2 [...] Read more.
Neuropeptide Y (NPY) is one of the most abundant peptides in the central nervous system of mammals and is involved in several physiological processes through NPY Y1, Y2, Y4 and Y5 receptors. Of those, the Y2 receptor has particular relevance for its autoreceptor role in inhibiting the release of NPY and other neurotransmitters and for its involvement in relevant mechanisms such as feeding behaviour, cognitive processes, emotion regulation, circadian rhythms and disorders such as epilepsy and cancer. PET imaging of the Y2 receptor can provide a valuable platform to understand this receptor’s functional role and evaluate its potential as a therapeutic target. In this work, we set out to refine the chemical and radiochemical synthesis of the Y2 receptor antagonist N-[11C]Me-JNJ31020028 for in vivo PET imaging studies. The non-radioactive reference compound, N-Me-JNJ-31020028, was synthesised through batch synthesis and continuous flow methodology, with 43% and 92% yields, respectively. N-[11C]Me-JNJ-31020028 was obtained with a radiochemical purity > 99%, RCY of 31% and molar activity of 156 GBq/μmol. PET imaging clearly showed the tracer’s biodistribution in several areas of the mouse brain and gut where Y2 receptors are known to be expressed. Full article
(This article belongs to the Special Issue Next-Generation Contrast Agents for Medical Imaging)
Show Figures

Graphical abstract

Back to TopTop