1,841 results on '"blood–brain barrier (bbb)"'
Search Results
2. Effect of chlorpyrifos on VEGF gene expression
- Author
-
Li, Wen and Ehrich, Marion
- Published
- 2023
- Full Text
- View/download PDF
3. Application of low-intensity ultrasound by opening blood–brain barrier for enhanced brain-targeted drug delivery
- Author
-
Li, Qian, Tang, Ziyan, Zhang, Yuanyuan, Yuan, Tianyu, Yuan, Bochuan, Du, Lina, and Jin, Yiguang
- Published
- 2023
- Full Text
- View/download PDF
4. A four-channel microfluidic model of the blood–brain and blood–cerebrospinal fluid barriers: fluid dynamics analysis.
- Author
-
Libet, Pavel A., Polynkin, Leonid Y., Saridis, Mikis R., Yakovlev, Egor V., Korsakova, Sofia A., Salmina, Alla B., Averchuk, Anton S., Rozanova, Natalia A., and Yurchenko, Stanislav O.
- Subjects
MICROPHYSIOLOGICAL systems ,COMPUTATIONAL fluid dynamics ,MICROFLUIDIC devices ,SHEARING force ,FLUID dynamics - Abstract
Brain-on-a-chip is an emerging field involving microfluidic devices capable of mimicking the structure and function of the human brain. Existing research often focuses on single barriers, such as the blood–brain barrier or blood–cerebrospinal fluid barrier (BCSFB). However, the brain has both barriers working together, and mimicking this dual system is crucial for better understanding of brain (patho)physiology. In this work, we present a four-channel microfluidic chip model that incorporates both the BBB and BCSFB, to reproduce physiologically correct architecture. Using computer simulations, we demonstrate that this model can mimic both healthy and diseased states by adjusting the shear stress experienced by the barriers, which is a key factor in their function. These findings offer valuable insights for designing future brain-on-a-chip devices with improved accuracy. This improved technology could contribute to wider advancements in tissue engineering and the study of brain function and diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Uncovering blood–brain barrier permeability: a comparative study of machine learning models using molecular fingerprints, and SHAP explainability.
- Author
-
Raveendrakumar, E., Gopichand, B., Bhosale, H., Melethadathil, N., and Valadi, J.
- Subjects
- *
MACHINE learning , *NEURAL development , *DRUG discovery , *DNA fingerprinting , *SUPPORT vector machines , *HUMAN fingerprints - Abstract
This study illustrates the use of chemical fingerprints with machine learning for blood–brain barrier (BBB) permeability prediction. Employing the Blood Brain Barrier Database (B3DB) dataset for BBB permeability prediction, we extracted nine different fingerprints. Support Vector Machine (SVM) and Extreme Gradient Boosting (XGBoost) algorithms were used to develop models for permeability prediction. Random Forest recursive Feature Selection (RF-RFS) method was used for extracting informative attributes. An additional database was employed for the validation phase. The results indicate that all nine datasets achieved good performance in training, test and validation stages. We further took MACC Keys fingerprints, one of the best performing models for explainability analysis. For this purpose, we used SHapley Additive exPlanations (SHAP) analysis on this dataset for the identification of key structural features influencing BBB permeability prediction. These features include aliphatic carbons, methyl groups and oxygen-containing groups. This study highlights the effectiveness of different fingerprint descriptors in predicting BBB permeability. SHAP analysis provides value additions to the simulations. These simulations will be of significant help in drug discovery processes, particularly in developing Central Nervous System (CNS) therapeutics. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. The Therapeutic Effects of SP-8356, a Verbenone Derivative, with Multimodal Cytoprotective Mechanisms in an Ischemic Stroke Rat Model.
- Author
-
Song, Hwa Young, Jin, Sejong, Lee, Sekwang, Jalin, Angela Melinda Anthony, Roh, Kyung-Hye, and Kim, Won-Ki
- Abstract
An ischemic cerebral stroke results from the interruption of blood flow to the brain, triggering rapid and complex cascades of excitotoxicity, oxidative stress, and inflammation. Current reperfusion therapies, including intravenous thrombolysis and mechanical thrombectomy, cause further brain injury due to reperfusion-induced cytotoxicity. To date, novel cytoprotective therapies that could address these challenges have yet to be developed, likely due to the limitations of targeting a single pathologic mechanism. To address these unmet clinical needs, we investigated a synthetic verbenone derivative, SP-8356, as a potential multi-target cytoprotective agent for acute ischemic strokes. In transient middle cerebral artery occlusion (MCAO) rats, SP-8356 significantly reduced brain infarct and edema volumes while improving acute neurological deficits in a dose-dependent manner. Furthermore, SP-8356 improved long-term outcomes, particularly by reducing mortality. These potent cytoprotective effects of SP-8356 were achieved by suppressing the excessive production of free radicals and pro-inflammatory cytokines, reducing the infiltration of inflammatory cells, and mitigating increases in blood–brain barrier permeability. Additional research is needed to determine whether co-administration of SP-8356 can extend the therapeutic time window of reperfusion therapies by mitigating ischemia/reperfusion injury. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Selection of LRP1 ligand phage-displayed single domain antibody that transmigrates BBB.
- Author
-
Manrique-Suárez, Viana, Mangui Catota, Bryan A., Camacho Casanova, Frank, Jara Mendoza, Nery A., Contreras Vera, Maria A., Maura Pérez, Rafael, Reyes López, Fátima, Toledo Alonso, Roberto, Castro Henriquez, Pablo Ignacio, and Sánchez Ramos, Oliberto
- Subjects
- *
CENTRAL nervous system , *ENDOTHELIAL cells , *MACROMOLECULES , *DRUG development , *LIGANDS (Biochemistry) - Abstract
AbstractEffective drug delivery to the central nervous system (CNS) remains a challenge due to the blood–brain barrier (BBB). Macromolecules such as proteins and peptides are unable to cross BBB and have poor therapeutic efficacy due to little or no drug distribution. A promising alternative is the conjugation of a drug to a shuttle molecule that can reach the CNS via receptor-mediated transcytosis (RMT). Several receptors have been described for RMT, such as low-density lipoprotein receptor-related protein 1 (LRP1). We used phage display technology combined with an
in vitro BBB model to identify LRP1 ligands. A single domain antibody (dAb) library was used to enrich for species that selectively bind to immobilised LRP1 ligand. We obtained a novel nanobody, dAb D11, that selectively binds to LRP1 receptor and mediatesin vitro internalisation of phage particles in brain endothelial cells, with a dissociation constant Kd of 183.1 ± 85.8 nM. The high permeability of D11 was demonstrated by anin vivo biodistribution assay in mice. We discovered D11, the first LRP1 binding dAb with BBB permeability. Our findings will contribute to the development of RMT-based drugs for the treatment of CNS diseases. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
8. IP-10 acts early in CV-A16 infection to induce BBB destruction and promote virus entry into the CNS by increasing TNF-α expression.
- Author
-
Hu, Yajie, Hu, Yunguang, Yin, Anguo, Lv, Yaming, Li, Jiang, Fan, Jingyuan, Qian, Baojiang, Song, Jie, and Zhang, Yunhui
- Subjects
HAND, foot & mouth disease ,PATHOLOGICAL physiology ,CENTRAL nervous system ,BLOOD-brain barrier ,VIRUS diseases - Abstract
The mechanisms underlying pathological changes in the central nervous system (CNS) following Coxsackievirus A16 (CV-A16) infection have not yet been elucidated. IFN-γ-inducible protein-10 (IP-10) is often used as a predictive factor to monitor early virus infection. It has also been reported that IP-10 plays a pivotal role in neuroinflammation. In this study, we aimed to explore the role of IP-10 in the neuropathogenesis of CV-A16 infection. We observed that the level of IP-10, as well as the TLR3-TRIF-TRAF3-TBK1-NF-κB and RIG-I/MDA5-MAVS-TRAFS-TBK1-NF-κB pathways, which are the upstream of IP-10, were significantly elevated during the course of CV-A16 infection. This increase was accompanied by an increase in a series of inflammatory cytokines at different time-points during CV-A16 infection. To determine whether IP-10 influences BBB integrity, we examined junctional complexes. Our results revealed that the expression levels of Claudin5, Occludin, ZO-1 and VE-Cadherin were notably decreased in CV-A16-infected HUVECs, but these indicators were restored in CV-A16-infected HUVECs with Eldelumab treatment. Nevertheless, IP-10 is only a chemokine that primarily traffics CXCR3-positive immune cells to inflammatory sites or promotes the production of inflammatory cytokines. Therefore, the interactions between IP-10 and inflammatory cytokines were evaluated. Our data revealed that IP-10 mediated the production of TNF-α, which was also observed to change the junctional complexes. Moreover, in a suckling mouse model, IP-10 and TNF-α treatments exacerbated clinical symptoms, mortality and pathological changes in the brain of CV-A16-infected mice, but Anti-IP-10 and Anti-TNF-α treatments alleviated these changes. Our data also revealed that IP-10 may be detected early in CV-A16 infection, whereas TNF-α was detected late in CV-A16 infection, and the production of TNF-α was also found to be positively correlated with IP-10. In addition, IP-10 and TNF-α were observed to reduce junctional complexes and enhance virus entry into the CNS. Taken together, this study provides the first evidence that CV-A16 activates the IP-10/TNF-α regulatory axis to cause BBB damage and accelerate the formation of neuroinflammation in infected hosts, which not only provides a new understanding of the neuropathogenesis caused by CV-A16, but also offers a promising target for the development of CV-A16 antiviral drugs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. The impacts of tobacco and nicotine on HIV-1 infection, inflammation, and the blood-brain barrier in the central nervous system.
- Author
-
Keane, Aislinn M. and Swartz, Talia H.
- Subjects
HIV ,LATENT infection ,CENTRAL nervous system ,NEUROBEHAVIORAL disorders ,TOBACCO smoke - Abstract
Human immunodeficiency virus (HIV-1) remains a persistent global health crisis. Even while successfully virologically suppressed, people with HIV (PWH) experience a higher risk for inflammatory disorders such as HIV-associated neurocognitive disorder (HAND). Tobacco use puts PWH at higher risk for neurocognitive symptoms resulting from HIV-associated neuroinflammation. The NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome has been implicated as a driver of HIV-associated inflammation, including HAND. Nicotine, the psychoactive component of tobacco smoke, has also been shown to signal through the NLRP3 inflammasome and modulate inflammatory signaling in the CNS. Here, we explore the impacts of nicotine and tobacco on the complex neurobiology of HAND, including effects on cognition, inflammation, viral latency, and blood-brain barrier integrity. We outline nicotine's role in the establishment of active and latent infection in the brain and posit the NLRP3 inflammasome as a common pathway by which HIV-1 and nicotine promote neuroinflammation in PWH. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Elucidating the E‐Cadherin⋯ADTC5 Interactions: A Theoretical Examination of Amino Acid Active Site Binding on the Electronic Level via Density Functional Theory.
- Author
-
Siahaan, Parsaoran, Royhansyah, Moch Freskha Fauzi, Abid, Muhammad Bahrul, Hudiyanti, Dwi, Anam, Khairul, Djunaidi, Muhammad Cholid, Sethio, Daniel, Anugrah, Daru Seto Bagus, and Sasongko, Nurwarrohman Andre
- Subjects
- *
ATOMS in molecules theory , *NATURAL orbitals , *DENSITY functional theory , *PEPTIDES , *HYDROGEN as fuel - Abstract
The presence of E‐cadherin⋯E‐cadherin interactions in the intercellular space (paracellular pathway) poses a notable limitation in drug delivery across the blood‐brain barrier (BBB). ADTC5 peptide has shown promising results in improving drug delivery across the BBB by modulating E‐cadherin⋯E‐cadherin interactions. However, the study of E‐cadherin⋯ADTC5 amino acid interactions using quantum mechanics approach has not been observed clearly. Herein, we observed the interaction between E‐cadherin⋯ADTC5 amino acids using density functional theory (DFT). Natural bond orbital (NBO), quantum theory of atoms in molecules (QTAIM), and reduced density gradient (RDG) analysis are performed to study the non‐covalent interaction in detail. The Arg55⋯Asp2 shows the strongest interaction between E‐cadherin⋯ADTC5 modulation, indicated by the highest stabilization energy (E(2)), and hydrogen bonding energy (EHB) about 22.04 and −6.90 kcal/mol, respectively. This interaction is dominated by hydrogen bonding. Such advancements could lead to significant improvements in therapeutic strategies for neurodegenerative diseases, where effective drug delivery across the BBB remains a major challenge. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Aging Increases Hypoxia-Induced Endothelial Permeability and Blood-Brain Barrier Dysfunction by Upregulating Arginase-II.
- Author
-
Xin Cheng, Potenza, Duilio M., Brenna, Andrea, Ajalbert, Guillaume, Zhihong Yang, and Xiu-Fen Ming
- Subjects
- *
ENDOTHELIAL cells , *BLOOD-brain barrier disorders , *CELL permeability - Abstract
Increased endothelial permeability plays an important role in blood-brain barrier (BBB) dysfunction and is implicated in neuronal injury in many diseased conditions. BBB disruption is primarily determined by dysfunction of endothelial cell-cell junctions. Deprivation of oxygen supply or hypoxia, a common feature of a variety of human diseases, is a major risk factor for BBB disruption. The molecular regulatory mechanisms of hypoxiainduced BBB dysfunction remain incompletely understood. The mitochondrial enzyme, arginase type II (Arg-II), has been shown to promote endothelial dysfunction. However, its role in hypoxia-induced BBB dysfunction has not been explored. In the C57BL/6J mouse model, hypoxia (8% O2, 24 hours) augments vascular Arg-II in the hippocampus, decreases cell-cell junction protein levels of Zonula occludens-1 (ZO-1), occludin, and CD31 in endothelial cells, increases BBB leakage in the brain in old mice (20 to 24 months) but not in young animals (3 to 6 months). These effects of hypoxia in aging are suppressed in arg-ii-/- mice. Moreover, the age-associated vulnerability of endothelial integrity to hypoxia is demonstrated in senescent human brain microvascular endothelial cell (hCMEC/D3) culture model. Further results in the cell culture model show that hypoxia augments Arg-II, decreases ZO-1 and occludin levels, and increases endothelial permeability, which is prevented by arg-ii gene silencing or by inhibition of mitochondrial reactive oxygen species (mtROS) production. Our study demonstrates an essential role of Arg-II in increased endothelial permeability and BBB dysfunction by promoting mtROS generation, resulting in decreased endothelial cell-cell junction protein levels under hypoxic conditions particularly in aging. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Collagen I Microfiber Promotes Brain Capillary Network Formation in Three–Dimensional Blood–Brain Barrier Microphysiological Systems.
- Author
-
Nakayama-Kitamura, Kimiko, Shigemoto-Mogami, Yukari, Piantino, Marie, Naka, Yasuhiro, Yamada, Asuka, Kitano, Shiro, Furihata, Tomomi, Matsusaki, Michiya, and Sato, Kaoru
- Subjects
MICROPHYSIOLOGICAL systems ,TRANSFERRIN receptors ,CENTRAL nervous system ,ANIMAL experimentation ,DRUG development - Abstract
Background: The blood–brain barrier (BBB) strictly regulates the penetration of substances into the brain, which, although important for maintaining brain homeostasis, may delay drug development because of the difficulties in predicting pharmacokinetics/pharmacodynamics (PKPD), toxicokinetics/toxicodynamics (TKTD), toxicity, safety, and efficacy in the central nervous system (CNS). Moreover, BBB functional proteins show species differences; therefore, humanized in vitro BBB models are urgently needed to improve the predictability of preclinical studies. Recently, international trends in the 3Rs in animal experiments and the approval of the FDA Modernization Act 2.0 have accelerated the application of microphysiological systems (MPSs) in preclinical studies, and in vitro BBB models have become synonymous with BBB–MPSs. Recently, we developed an industrialized humanized BBB–MPS, BBB–NET. In our previous report, we reproduced transferrin receptor (TfR)–mediated transcytosis with high efficiency and robustness, using hydrogels including fibrin and collagen I microfibers (CMFs). Methods: We investigated how adding CMFs to the fibrin gel benefits BBB-NETs. Results: We showed that CMFs accelerate capillary network formation and maturation by promoting astrocyte (AC) survival, and clarified that integrin β1 is involved in the mechanism of CMFs. Conclusions: Our data suggest that the quality control (QC) of CMFs is important for ensuring the stable production of BBB–NETs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. A four-channel microfluidic model of the blood–brain and blood–cerebrospinal fluid barriers: fluid dynamics analysis
- Author
-
Pavel A. Libet, Leonid Y. Polynkin, Mikis R. Saridis, Egor V. Yakovlev, Sofia A. Korsakova, Alla B. Salmina, Anton S. Averchuk, Natalia A. Rozanova, and Stanislav O. Yurchenko
- Subjects
Microfluidic ,Blood–brain barrier (BBB) ,Blood–cerebrospinal fluid barrier (BCSFB) ,Fluid dynamics ,Shear stress ,Tissue engineering ,Technology - Abstract
Abstract Brain-on-a-chip is an emerging field involving microfluidic devices capable of mimicking the structure and function of the human brain. Existing research often focuses on single barriers, such as the blood–brain barrier or blood–cerebrospinal fluid barrier (BCSFB). However, the brain has both barriers working together, and mimicking this dual system is crucial for better understanding of brain (patho)physiology. In this work, we present a four-channel microfluidic chip model that incorporates both the BBB and BCSFB, to reproduce physiologically correct architecture. Using computer simulations, we demonstrate that this model can mimic both healthy and diseased states by adjusting the shear stress experienced by the barriers, which is a key factor in their function. These findings offer valuable insights for designing future brain-on-a-chip devices with improved accuracy. This improved technology could contribute to wider advancements in tissue engineering and the study of brain function and diseases.
- Published
- 2024
- Full Text
- View/download PDF
14. Depleting parenchymal border macrophages alleviates cerebral edema and neuroinflammation following status epilepticus
- Author
-
Renbao Lin, Rui Luo, Xinyue Yu, Junjie Zou, Xiaowei Huang, and Yanwu Guo
- Subjects
Parenchymal border macrophage (PBM) ,Cerebral edema ,Cerebrospinal fluid (CSF) ,Blood–brain barrier (BBB) ,Status epilepticus (SE) ,Inflammation ,Medicine - Abstract
Abstract Background Status epilepticus (SE) is a common severe neurological emergency. Cerebral edema caused by SE is unavoidable and may exacerbate epilepsy. Recent studies have identified cerebrospinal fluid (CSF) as a crucial fluid source of initial cerebral edema following ischemic stroke and cardiac arrest. Moreover, synchronized neuronal firings drive CSF influx into interstitial fluid (ISF). Parenchymal border macrophages (PBMs) have been found to play a role in regulating CSF flow dynamics. However, the involvement of CSF and PBMs in cerebral edema during SE remains unclear. Here, we investigated the fluid source of cerebral edema in the initial phase of SE with the role of PBMs involved. Methods Lithium chloride-pilocarpine was used to induce SE in C57BL/6 J mice. Electroencephalogram (EEG) was acquired to assess changes in relative EEG power pre- and post-seizure onset. Apparent diffusion coefficient (ADC) maps reconstructed from diffusion-weighted imaging (DWI) were utilized to evaluate cytotoxic edema. Blood–brain barrier (BBB) permeability was examined using sodium fluorescein (NaFl). CSF tracer influx into the brain was assessed by transcranial imaging and brain slices. PBMs were depleted using clodronate liposomes. Immunohistochemistry was used to evaluate PBM depletion, severity of vasogenic edema, inflammation, and neuronal damage. Results During the initial stage of SE, relative EEG power sharply increased and ADC values significantly decreased. Concurrently, CSF tracer influx into the cortex significantly elevated, though NaFl leakage from blood to brain parenchyma did not evidently alter. Following depletion of PBM, CSF influx declined but AQP4 expression and polarization remained unaffected. Post-PBM depletion, there was no significant alteration in relative EEG power, yet CSF influx decreased substantially during the initial stage of SE. The degree of ADC decline lessened, IgG extravasation after SE decreased, activated microglia and proliferating astrocytes count fell, and neuronal damage post-SE alleviated. Conclusions CSF appeared to contribute to cerebral edema in SE. Depletion of PBM alleviated cytotoxic edema in the initial phase of SE, and subsequent vasogenic edema, inflammatory response and neurological damage were reduced. These findings may provide potential novel strategies for treating cerebral edema following SE.
- Published
- 2024
- Full Text
- View/download PDF
15. Nanotechnological approaches for efficient N2B delivery: from small-molecule drugs to biopharmaceuticals
- Author
-
Selin Akpinar Adscheid, Akif E. Türeli, Nazende Günday-Türeli, and Marc Schneider
- Subjects
antibody delivery ,biopharmaceutical delivery ,blood–brain barrier (bbb) ,cns diseases ,drug delivery ,hybrid nanoparticles ,intranasal delivery ,liposomes ,nanomedicine ,nanostructured lipid carriers (nlcs) ,polymer nanoparticles ,rna delivery ,solid lipid nanoparticles (slns) ,Technology ,Chemical technology ,TP1-1185 ,Science ,Physics ,QC1-999 - Abstract
Central nervous system diseases negatively affect patients and society. Providing successful noninvasive treatments for these diseases is challenging because of the presence of the blood–brain barrier. While protecting the brain’s homeostasis, the barrier limits the passage of almost all large-molecule drugs and most small-molecule drugs. A noninvasive method, nose-to-brain delivery (N2B delivery) has been proposed to overcome this challenge. By exploiting the direct anatomical interaction between the nose and the brain, the drugs can reach the target, the brain. Moreover, the drugs can be encapsulated into various drug delivery systems to enhance physicochemical characteristics and targeting success. Many preclinical data show that this strategy can effectively deliver biopharmaceuticals to the brain. Therefore, this review focuses on N2B delivery while giving examples of different drug delivery systems suitable for the applications. In addition, we emphasize the importance of the effective delivery of monoclonal antibodies and RNA and stress the recent literature tackling this challenge. While giving examples of nanotechnological approaches for the effective delivery of small or large molecules from the current literature, we highlight the preclinical studies and their results to prove the strategies’ success and limitations.
- Published
- 2024
- Full Text
- View/download PDF
16. Truncated mini LRP1 transports cargo from luminal to basolateral side across the blood brain barrier
- Author
-
Laura Fritzen, Katharina Wienken, Lelia Wagner, Magdalena Kurtyka, Katharina Vogel, Jakob Körbelin, Sascha Weggen, Gert Fricker, and Claus U. Pietrzik
- Subjects
Low-density lipoprotein receptor-related protein 1 (LRP1) ,Blood-brain barrier (BBB) ,Drug delivery ,Liposomes ,γ-secretase modulator (GSM) ,Nanocarrier ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background The most crucial area to focus on when thinking of novel pathways for drug delivery into the CNS is the blood brain barrier (BBB). A number of nanoparticulate formulations have been shown in earlier research to target receptors at the BBB and transport therapeutics into the CNS. However, no mechanism for CNS entrance and movement throughout the CNS parenchyma has been proposed yet. Here, the truncated mini low-density lipoprotein receptor-related protein 1 mLRP1_DIV* was presented as blood to brain transport carrier, exemplified by antibodies and immunoliposomes using a systematic approach to screen the receptor and its ligands’ route across endothelial cells in vitro. Methods The use of mLRP1_DIV* as liposomal carrier into the CNS was validated based on internalization and transport assays across an in vitro model of the BBB using hcMEC/D3 and bEnd.3 cells. Trafficking routes of mLRP1_DIV* and corresponding cargo across endothelial cells were analyzed using immunofluorescence. Modulation of γ-secretase activity by immunoliposomes loaded with the γ-secretase modulator BB25 was investigated in co-cultures of bEnd.3 mLRP1_DIV* cells and CHO cells overexpressing human amyloid precursor protein (APP) and presenilin 1 (PSEN1). Results We showed that while expressed in vitro, mLRP1_DIV* transports both, antibodies and functionalized immunoliposomes from luminal to basolateral side across an in vitro model of the BBB, followed by their mLRP1_DIV* dependent release of the cargo. Importantly, functionalized liposomes loaded with the γ-secretase modulator BB25 were demonstrated to effectively reduce toxic Aß42 peptide levels after mLRP1_DIV* mediated transport across a co-cultured endothelial monolayer. Conclusion Together, the data strongly suggest mLRP1_DIV* as a promising tool for drug delivery into the CNS, as it allows a straight transport of cargo from luminal to abluminal side across an endothelial monolayer and it’s release into brain parenchyma in vitro, where it exhibits its intended therapeutic effect.
- Published
- 2024
- Full Text
- View/download PDF
17. Genetic and pathophysiological insights from autopsied patient with primary familial brain calcification: novel MYORG variants and astrocytic implications
- Author
-
Takahiro Hobara, Yujiro Higuchi, Mari Yoshida, Masahito Suehara, Masahiro Ando, Jun-Hui Yuan, Akiko Yoshimura, Fumikazu Kojima, Eiji Matsuura, Yuji Okamoto, Jun Mitsui, Shoji Tsuji, and Hiroshi Takashima
- Subjects
Primary familial brain calcification (PFBC) ,Idiopathic basal ganglia calcification (IBGC) ,MYORG ,Blood-brain barrier (BBB) ,Astrocyte ,Olivary hypertrophy ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Primary familial brain calcification (PFBC) is a genetic neurological disorder characterized by symmetric brain calcifications that manifest with variable neurological symptoms. This study aimed to explore the genetic basis of PFBC and elucidate the underlying pathophysiological mechanisms. Six patients from four pedigrees with brain calcification were enrolled. Whole-exome sequencing identified two novel homozygous variants, c.488G > T (p.W163L) and c.2135G > A (p.W712*), within the myogenesis regulating glycosidase (MYORG) gene. Cerebellar ataxia (n = 5) and pyramidal signs (n = 4) were predominant symptoms, with significant clinical heterogeneity noted even within the same family. An autopsy of one patient revealed extensive brainstem calcifications, sparing the cerebral cortex, and marked by calcifications predominantly in capillaries and arterioles. The pathological study suggested morphological alterations characterized by shortened foot processes within astrocytes in regions with pronounced calcification and decreased immunoreactivity of AQP4. The morphology of astrocytes in regions without calcification remains preserved. Neuronal loss and gliosis were observed in the basal ganglia, thalamus, brainstem, cerebellum, and dentate nucleus. Notably, olivary hypertrophy, a previously undescribed feature in MYORG-PFBC, was discovered. Neuroimaging showed reduced blood flow in the cerebellum, highlighting the extent of cerebellar involvement. Among perivascular cells constituting the blood-brain barrier (BBB) and neurovascular unit, MYORG is most highly expressed in astrocytes. Astrocytes are integral components of the BBB, and their dysfunction can precipitate BBB disruption, potentially leading to brain calcification and subsequent neuronal loss. This study presents two novel homozygous variants in the MYORG gene and highlights the pivotal role of astrocytes in the development of brain calcifications, providing insights into the pathophysiological mechanisms underlying PFBC associated with MYORG variants.
- Published
- 2024
- Full Text
- View/download PDF
18. Therapeutic Effect of Boron Neutron Capture Therapy on Boronophenylalanine Administration via Cerebrospinal Fluid Circulation in Glioma Rat Models.
- Author
-
Kusaka, Sachie, Voulgaris, Nikolaos, Onishi, Kazuki, Ueda, Junpei, Saito, Shigeyoshi, Tamaki, Shingo, Murata, Isao, Takata, Takushi, and Suzuki, Minoru
- Subjects
- *
BORON-neutron capture therapy , *LABORATORY rats , *NEUTRON irradiation , *BRAIN tumors , *RESEARCH reactors - Abstract
In recent years, various drug delivery systems circumventing the blood–brain barrier have emerged for treating brain tumors. This study aimed to improve the efficacy of brain tumor treatment in boron neutron capture therapy (BNCT) using cerebrospinal fluid (CSF) circulation to deliver boronophenylalanine (BPA) to targeted tumors. Previous experiments have demonstrated that boron accumulation in the brain cells of normal rats remains comparable to that after intravenous (IV) administration, despite BPA being administered via CSF at significantly lower doses (approximately 1/90 of IV doses). Based on these findings, BNCT was conducted on glioma model rats at the Kyoto University Research Reactor Institute (KUR), with BPA administered via CSF. This method involved implanting C6 cells into the brains of 8-week-old Wistar rats, followed by administering BPA and neutron irradiation after a 10-day period. In this study, the rats were divided into four groups: one receiving CSF administration, another receiving IV administration, and two control groups without BPA administration, with one subjected to neutron irradiation and the other not. In the CSF administration group, BPA was infused from the cisterna magna at 8 mg/kg/h for 2 h, while in the IV administration group, BPA was intravenously administered at 350 mg/kg via the tail vein over 1.5 h. Thermal neutron irradiation (5 MW) for 20 min, with an average fluence of 3.8 × 1012/cm2, was conducted at KUR's heavy water neutron irradiation facility. Subsequently, all of the rats were monitored under identical conditions for 7 days, with pre- and post-irradiation tumor size assessed through MRI and pathological examination. The results indicate a remarkable therapeutic efficacy in both BPA-administered groups (CSF and IV). Notably, the rats treated with CSF administration exhibited diminished BPA accumulation in normal tissue compared to those treated with IV administration, alongside maintaining excellent overall health. Thus, CSF-based BPA administration holds promise as a novel drug delivery mechanism in BNCT. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. Blood-tumor barrier in focus - investigation of glioblastoma-induced effects on the blood-brain barrier.
- Author
-
Mathew-Schmitt, Sanjana, Peindl, Matthias, Neundorf, Philipp, Dandekar, Gudrun, Metzger, Marco, Nickl, Vera, and Appelt-Menzel, Antje
- Abstract
Purpose: Glioblastoma (GBM) is the most prevalent, malignant, primary brain tumor in adults, characterized by limited treatment options, frequent relapse, and short survival after diagnosis. Until now, none of the existing therapy and treatment approaches have proven to be an effective cure. The availability of predictive human blood-tumor barrier (BTB) test systems that can mimic in-vivo pathophysiology of GBM would be of great interest in preclinical research. Here, we present the establishment of a new BTB in-vitro test system combining GBM spheroids and BBB models derived from human induced pluripotent stem cells (hiPSCs). Methods: We co-cultured hiPSC-derived brain capillary endothelial-like cells (iBCECs) with GBM spheroids derived from U87-MG and U373-MG cell lines in a cell culture insert-based format. Spheroids were monitored over 168 hours (h) of culture, characterized for GBM-specific marker expression and treated with standard chemotherapeutics to distinguish inhibitory effects between 2D mono-culture and 3D spheroids. GBM-induced changes on iBCECs barrier integrity were verified via measurement of transendothelial electrical resistance (TEER), immunocytochemical staining of tight junction (TJ) proteins claudin-5 and occludin as well as the glucose transporter-1 (Glut-1). GBM-induced secretion of vascular endothelial growth factor (VEGF) was additionally quantified. Results: Our hypothesis was validated by reduced expression of TJ proteins, occludin and claudin-5 together with significant barrier breakdown in iBCECs after only 24 h of co-culture, demonstrated by reduction in TEER from 1313 ± 265 Ω*cm
2 to 712 ± 299 Ω*cm2 (iBCECs + U87-MG) and 762 ± 316 Ω*cm2 (iBCECs + U373-MG). Furthermore, 3D spheroids show more resistance to standard GBM chemotherapeutics in-vitro compared to 2D cultures. Conclusions: We demonstrate the establishment of a simplified, robust in-vitro BTB test system, with potential application in preclinical therapeutic screening and in studying GBM-induced pathological changes at the BBB. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
20. Truncated mini LRP1 transports cargo from luminal to basolateral side across the blood brain barrier.
- Author
-
Fritzen, Laura, Wienken, Katharina, Wagner, Lelia, Kurtyka, Magdalena, Vogel, Katharina, Körbelin, Jakob, Weggen, Sascha, Fricker, Gert, and Pietrzik, Claus U.
- Subjects
AMYLOID beta-protein precursor ,CHO cell ,PEPTIDES ,BLOOD-brain barrier ,ENDOTHELIAL cells - Abstract
Background: The most crucial area to focus on when thinking of novel pathways for drug delivery into the CNS is the blood brain barrier (BBB). A number of nanoparticulate formulations have been shown in earlier research to target receptors at the BBB and transport therapeutics into the CNS. However, no mechanism for CNS entrance and movement throughout the CNS parenchyma has been proposed yet. Here, the truncated mini low-density lipoprotein receptor-related protein 1 mLRP1_DIV* was presented as blood to brain transport carrier, exemplified by antibodies and immunoliposomes using a systematic approach to screen the receptor and its ligands' route across endothelial cells in vitro. Methods: The use of mLRP1_DIV* as liposomal carrier into the CNS was validated based on internalization and transport assays across an in vitro model of the BBB using hcMEC/D3 and bEnd.3 cells. Trafficking routes of mLRP1_DIV* and corresponding cargo across endothelial cells were analyzed using immunofluorescence. Modulation of γ-secretase activity by immunoliposomes loaded with the γ-secretase modulator BB25 was investigated in co-cultures of bEnd.3 mLRP1_DIV* cells and CHO cells overexpressing human amyloid precursor protein (APP) and presenilin 1 (PSEN1). Results: We showed that while expressed in vitro, mLRP1_DIV* transports both, antibodies and functionalized immunoliposomes from luminal to basolateral side across an in vitro model of the BBB, followed by their mLRP1_DIV* dependent release of the cargo. Importantly, functionalized liposomes loaded with the γ-secretase modulator BB25 were demonstrated to effectively reduce toxic Aß
42 peptide levels after mLRP1_DIV* mediated transport across a co-cultured endothelial monolayer. Conclusion: Together, the data strongly suggest mLRP1_DIV* as a promising tool for drug delivery into the CNS, as it allows a straight transport of cargo from luminal to abluminal side across an endothelial monolayer and it's release into brain parenchyma in vitro, where it exhibits its intended therapeutic effect. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
21. Assessment of Mannitol-Induced Chronic Blood–Brain Barrier Dysfunction In Vivo Using Magnetic Resonance.
- Author
-
Sampedro-Viana, Ana, Fernández-Rodicio, Sabela, Castillo, José, Hervella, Pablo, Alonso-Alonso, María Luz, and Iglesias-Rey, Ramón
- Subjects
- *
SPRAGUE Dawley rats , *ALZHEIMER'S disease , *MAGNETIC resonance imaging , *MAGNETIC flux leakage ,BRAIN metabolism - Abstract
The blood–brain barrier (BBB) is essential for protection and plays a crucial role in chronic neurological disorders like small-vessel disease and Alzheimer's disease. Its complexity poses significant challenges for effective diagnostics and treatments, highlighting the need for novel animal models and comprehensive BBB dysfunction studies. This study investigates chronic BBB dysfunction induction using osmotic disruption via mannitol in healthy adult male Sprague Dawley rats over 12 weeks. Group 1 received 1 bolus/week (2.0 g/kg), Group 2 received 3 boluses/week (1.5 g/kg), and Group 3 received 3 boluses/week (2.5 g/kg). BBB dysfunction was assessed using gadolinium (Gd) infusion and MRI to evaluate location, severity, evolution, and persistence. MR spectroscopy (MRS) examined the brain metabolism changes due to intravenous mannitol, with T2-weighted MRI assessing brain lesions. Biomarkers of neuroinflammation were analyzed in the highest mannitol dose group. Our data show chronic BBB dysfunction primarily in the cortex, hippocampus, and striatum, but not in the corpus callosum of rats under periodic mannitol dosing in groups 1 and 2. MRS identified a distinctive metabolite signature, including changes in alanine, choline, and N-acetyl aspartate in the striatum of Group 1. No significant differences were found in the serum levels of all pro- and anti-inflammatory cytokines analyzed in the high-dose Group 3. This study underscores the feasibility and implications of using osmotic disruption to model chronic BBB dysfunction, offering insights for future neuroprotection and therapeutic strategies research. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Whole-Genome Omics Elucidates the Role of CCM1 and Progesterone in Cerebral Cavernous Malformations within CmPn Networks.
- Author
-
Croft, Jacob, Grajeda, Brian, Gao, Liyuan, Abou-Fadel, Johnathan, Badr, Ahmed, Sheng, Victor, and Zhang, Jun
- Subjects
- *
INTRACRANIAL hemorrhage , *HEMORRHAGIC stroke , *PROGESTERONE receptors , *SYSTEMS biology , *BRAIN damage , *BLOOD-brain barrier - Abstract
Cerebral cavernous malformations (CCMs) are abnormal expansions of brain capillaries that increase the risk of hemorrhagic strokes, with CCM1 mutations responsible for about 50% of familial cases. The disorder can cause irreversible brain damage by compromising the blood–brain barrier (BBB), leading to fatal brain hemorrhages. Studies show that progesterone and its derivatives significantly impact BBB integrity. The three CCM proteins (CCM1, CCM2, and CCM3) form the CCM signaling complex (CSC), linking classic and non-classic progesterone signaling within the CmPn network, which is crucial for maintaining BBB integrity. This study aimed to explore the relationship between CCM1 and key pathways of the CmPn signaling network using three mouse embryonic fibroblast lines (MEFs) with distinct CCM1 expressions. Omics and systems biology analysis investigated CCM1-mediated signaling within the CmPn network. Our findings reveal that CCM1 is essential for regulating cellular processes within progesterone-mediated CmPn/CmP signaling, playing a crucial role in maintaining microvessel integrity. This regulation occurs partly through gene transcription control. The critical role of CCM1 in these processes suggests it could be a promising therapeutic target for CCMs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. Cerebral malaria pathogenesis: Dissecting the role of CD4+ and CD8+ T-cells as major effectors in disease pathology.
- Author
-
Sharma, Indu, Kataria, Poonam, and Das, Jyoti
- Subjects
- *
T helper cells , *CYTOTOXIC T cells , *REGULATORY T cells , *ERYTHROCYTES , *CEREBRAL malaria - Abstract
Cerebral malaria (CM) is a severe complication of Plasmodium falciparum (P. falciparum) infection, with complex pathogenesis involving multiple factors, including the host's immunological response. T lymphocytes, specifically CD4+ T helper cells and CD8+ cytotoxic T cells, are crucial in controlling parasite growth and activating cells for parasite clearance via cytokine secretion. Contrary to this, reports also suggest the pathogenic nature of T lymphocytes as they are often involved in disease progression and severity. CD8+ cytotoxic T cells migrate to the host's brain vasculature, disrupting the blood-brain barrier and causing neurological manifestations. CD4+ T helper cells on the other hand play a variety of functions as they differentiate into different subtypes which may function as pro-inflammatory or anti-inflammatory. The excessive pro-inflammatory response in CM can lead to multi-organ failure, necessitating a check mechanism to maintain immune homeostasis. This is achieved by regulatory T cells and their characteristic cytokines, which counterbalance the pro-inflammatory immune response. Maintaining a critical balance between pro and anti-inflammatory responses is crucial for determining disease outcomes in CM. A slight change in this balance may contribute to a disease severity owing to an extreme inflammatory response or unrestricted parasite growth, a potential target for designing immunotherapeutic treatment approaches. The review briefly discusses the pathogenesis of CM and various mechanisms responsible for the disruption of the blood-brain barrier. It also highlights the role of different T cell subsets during infection and emphasizes the importance of balance between pro and anti-inflammatory T cells that ultimately decides the outcome of the disease. SUMMARY: CM is potentially fatal complication of P. falciparum infection that presents with high mortality and morbidity. Vaccines are extensively being developed against the Plasmodium parasite but very few of them are effective. Artemisinin Combination Therapy (ACT) is a major treatment for malaria, but its effectiveness is declining due to Plasmodium sp. developing resistance to it, necessitating the need for development of new drugs and treatments. During infection, the parasite is responsible for causing infected red blood cell (RBC) sequestration and cytoadherence in brain vasculature and extreme pro-inflammatory response that ultimately causes endothelial dysfunction and bloodbrain barrier (BBB) disruption. The host initiates a pro-inflammatory response against the parasite which includes activation of cells of both innate and adaptive immune response. These cells control the parasite growth and aid in parasite clearance from host's body. The inflammatory response generally targets foreign pathogens and provides protection against possible infection but can also cause harm to the self when left unchecked. It has been reported that activated immune cells, mainly T-lymphocytes often migrate to brain vasculature and ultimately results in neuronal damage characteristic CM. To counteract the overwhelming pro-inflammatory response, the host immune system deploys an anti-inflammatory response, which often involves regulatory cells and cytokines that help the body maintain immunological homeostasis. The review briefly highlights the necessity of balancing the pro- and anti-inflammatory responses for successful parasite clearance without the deleterious effects to the host that might increase disease severity in CM. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. Genetic and pathophysiological insights from autopsied patient with primary familial brain calcification: novel MYORG variants and astrocytic implications.
- Author
-
Hobara, Takahiro, Higuchi, Yujiro, Yoshida, Mari, Suehara, Masahito, Ando, Masahiro, Yuan, Jun-Hui, Yoshimura, Akiko, Kojima, Fumikazu, Matsuura, Eiji, Okamoto, Yuji, Mitsui, Jun, Tsuji, Shoji, and Takashima, Hiroshi
- Subjects
OLIVARY degeneration ,DENTATE nucleus ,CEREBRAL cortex ,NEUROLOGICAL disorders ,CEREBELLAR ataxia - Abstract
Primary familial brain calcification (PFBC) is a genetic neurological disorder characterized by symmetric brain calcifications that manifest with variable neurological symptoms. This study aimed to explore the genetic basis of PFBC and elucidate the underlying pathophysiological mechanisms. Six patients from four pedigrees with brain calcification were enrolled. Whole-exome sequencing identified two novel homozygous variants, c.488G > T (p.W163L) and c.2135G > A (p.W712*), within the myogenesis regulating glycosidase (MYORG) gene. Cerebellar ataxia (n = 5) and pyramidal signs (n = 4) were predominant symptoms, with significant clinical heterogeneity noted even within the same family. An autopsy of one patient revealed extensive brainstem calcifications, sparing the cerebral cortex, and marked by calcifications predominantly in capillaries and arterioles. The pathological study suggested morphological alterations characterized by shortened foot processes within astrocytes in regions with pronounced calcification and decreased immunoreactivity of AQP4. The morphology of astrocytes in regions without calcification remains preserved. Neuronal loss and gliosis were observed in the basal ganglia, thalamus, brainstem, cerebellum, and dentate nucleus. Notably, olivary hypertrophy, a previously undescribed feature in MYORG-PFBC, was discovered. Neuroimaging showed reduced blood flow in the cerebellum, highlighting the extent of cerebellar involvement. Among perivascular cells constituting the blood-brain barrier (BBB) and neurovascular unit, MYORG is most highly expressed in astrocytes. Astrocytes are integral components of the BBB, and their dysfunction can precipitate BBB disruption, potentially leading to brain calcification and subsequent neuronal loss. This study presents two novel homozygous variants in the MYORG gene and highlights the pivotal role of astrocytes in the development of brain calcifications, providing insights into the pathophysiological mechanisms underlying PFBC associated with MYORG variants. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Poliovirus capsid protein VP3 can penetrate vascular endothelial cells.
- Author
-
Mizutani, Taketoshi and Ishizaka, Aya
- Subjects
- *
VASCULAR endothelial cells , *MONONUCLEAR leukocytes , *CENTRAL nervous system , *PEPTIDES , *ENDOTHELIAL cells , *PERICYTES - Abstract
The poliovirus (PV) enters the central nervous system (CNS) via the bloodstream, suggesting the existence of a mechanism to cross the blood–brain barrier. Here, we report that PV capsid proteins (VP1 and VP3) can penetrate cells, with VP3 being more invasive. Two independent parts of VP3 are responsible for this function. Both peptides can penetrate human umbilical cord vascular endothelial cells, and one peptide of VP3 could also penetrate peripheral blood mononuclear cells. In an in vitro blood–brain barrier model using rat‐derived astrocytes, pericytes, and endothelial cells, both peptides were observed to traverse from the blood side to the brain side at 6 h after administration. These results provide insights into the molecular mechanisms underlying PV invasion into the CNS. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. A novel histone deacetylase inhibitor W2A-16 improves the barrier integrity in brain vascular endothelial cells.
- Author
-
Yasuteru Inoue, Yingxue Ren, Shuwen Zhang, Bamkole, Michael, Islam, Naeyma N., Selvaraj, Manikandan, Wenyan Lu, Caulfield, Thomas R., Yonghe Li, and Takahisa Kanekiyo
- Abstract
The maturation of brain microvascular endothelial cells leads to the formation of a tightly sealed monolayer, known as the blood-brain barrier (BBB). The BBB damage is associated with the pathogenesis of age-related neurodegenerative diseases including vascular cognitive impairment and Alzheimer's disease. Growing knowledge in the field of epigenetics can enhance the understanding of molecular profile of the BBB and has great potential for the development of novel therapeutic strategies or targets to repair a disrupted BBB. Histone deacetylases (HDACs) inhibitors are epigenetic regulators that can induce acetylation of histones and induce open chromatin conformation, promoting gene expression by enhancing the binding of DNA with transcription factors. We investigated how HDAC inhibition influences the barrier integrity using immortalized human endothelial cells (HCMEC/D3) and the human induced pluripotent stem cell (iPSC)-derived brain vascular endothelial cells. The endothelial cells were treated with or without a novel compound named W2A-16. W2A-16 not only activates Wnt/ß-catenin signaling but also functions as a class I HDAC inhibitor. We demonstrated that the administration with W2A-16 sustained barrier properties of the monolayer of endothelial cells, as evidenced by increased trans-endothelial electrical resistance (TEER). The BBB-related genes and protein expression were also increased compared with non-treated controls. Analysis of transcript profiles through RNA-sequencing in hCMEC/D3 cells indicated that W2A-16 potentially enhances BBB integrity by influencing genes associated with the regulation of the extracellular microenvironment. These findings collectively propose that the HDAC inhibition by W2A-16 plays a facilitating role in the formation of the BBB. Pharmacological approaches to inhibit HDAC may be a potential therapeutic strategy to boost and/or restore BBB integrity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Effect of high-fat diet on cerebral pathological changes of cerebral small vessel disease in SHR/SP rats.
- Author
-
Zhang, Yuchi, Sheikh, Abdullah Md., Tabassum, Shatera, Iwasa, Kenichi, Shibly, Abu Zaffar, Zhou, Xiaojing, Wang, Ruochen, Bhuiya, Jubo, Abdullah, Fatema Binte, Yano, Shozo, Aoki, Yoshihito, and Nagai, Atsushi
- Subjects
CEREBRAL small vessel diseases ,HIGH-fat diet ,PATHOLOGICAL physiology ,RATS ,HYPERTENSION risk factors ,CORPUS callosum - Abstract
Cerebral small vessel diseases (CSVD) are neurological disorders associated with microvessels, manifested pathologically as white matter (WM) changes and cortical microbleeds, with hypertension as a risk factor. Additionally, a high-fat diet (HFD) can affect peripheral vessel health. Our study explored how HFD affects cerebral small vessels in normotensive WKY, hypertensive SHR, and SHR/SP rats. The MRI results revealed that HFD specifically increased WM hyperintensity in SHR/SP rats. Pathologically, it increased WM pallor and vacuolation in SHR and SHR/SP rats. Levels of blood–brain barrier (BBB) protein claudin 5 were decreased in SHR and SHR/SP compared to WKY, with HFD having minimal impact on these levels. Conversely, collagen IV levels remained consistent among the rat strains, which were increased by HFD. Consequently, HFD caused vessel leakage in all rat strains, particularly within the corpus callosum of SHR/SP rats. To understand the underlying mechanisms, we assessed the levels of hypoxia-inducible factor-1α (HIF-1α), Gp91-phox, and neuroinflammatory markers astrocytes, and microglia were increased in SHR and SHR/SP compared to WKY and were further elevated by HFD in all rat strains. Gp91-phox was also increased in SHR and SHR/SP compared to WKY, with HFD causing an increase in WKY but little effect in SHR and SHR/SP. In conclusion, our study demonstrates that HFD, in combined with hypertension, intensifies cerebral pathological alterations in CSVD rats. This exacerbation involves increased oxidative stress and HIF-1α in cerebral vessels, triggering neuroinflammation, vascular basement membrane remodeling, IgG leakage, and ultimately WM damage. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. Elevated cerebrospinal fluid IgG index in herpes simplex encephalitis post‐HSV‐1 clearance: A preliminary study.
- Author
-
Asakura, Mao, Mizutani, Yasuaki, Shima, Sayuri, Kawamura, Yoshiki, Ueda, Akihiro, Ito, Mizuki, Mutoh, Tatsuro, Yoshikawa, Tetsushi, and Watanabe, Hirohisa
- Subjects
HUMAN herpesvirus 1 ,HERPES simplex ,HERPES simplex virus ,CEREBROSPINAL fluid ,IMMUNOGLOBULIN G ,CHICKENPOX - Abstract
Herpes simplex encephalitis (HSE) is an acute form of encephalitis that can lead to poor neurological outcomes. Although the exact pathogenesis of HSE remains elusive, recent reports suggest a significant role for postinfectious immune‐inflammatory processes in the central nervous system (CNS). This study aimed to clarify the association between CNS autoimmune responses and clinical presentation in patients with HSE, focusing on cerebrospinal fluid (CSF) characteristics, particularly the IgG index. We retrospectively analyzed 176 consecutive patients suspected of having aseptic meningitis /encephalitis for chronological changes in CSF findings and clinical presentations. These patients underwent PCR screening for herpesviruses (HV) in their CSF. We identified seven patients positive for herpes simplex virus type 1 (HSV‐1), 20 patients positive for varicella‐zoster virus, and 17 patients who met the criteria for aseptic meningitis but were PCR‐negative for HV. Patients in the HSV‐1‐positive group exhibited a significant increase in the IgG index at the time of PCR‐negative conversion compared with on admission (p = 0.0156), while such a change was not observed in the other two groups. Additionally, all patients in the HSV‐1‐positive group tested negative for anti‐neural autoantibodies in CSF and serum samples collected approximately 3 weeks after onset. This study, therefore, highlights that CSF IgG index elevation occurs even after PCR‐confirmed HSV‐1 clearance, which might indicate immunopathogenesis that is independent of antibody‐mediated mechanisms. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. Curcumin encapsulated in PAMAM dendrimers for the therapeutic treatment of ischemic stroke in rats
- Author
-
Justin Stadler, Lucas G. Garmo, David Doyle, Chin-I. Cheng, Garrett Richardson, Zain Waheed, Tim Tofan, Bhairavi Srinageshwar, Ajit Sharma, Robert B. Petersen, Gary L. Dunbar, and Julien Rossignol
- Subjects
blood-brain barrier (BBB) ,curcumin ,ischemic stroke ,neuroinflammation ,PAMAM dendrimers ,Biology (General) ,QH301-705.5 - Abstract
IntroductionIschemic stroke is a devastating neurovascular condition that occurs when cerebral tissue fails to receive an adequate supply of oxygen. Despite being a leading cause of death and disability worldwide, therapeutic interventions are currently limited. Polyamidoamine (PAMAM) dendrimers are nanomolecules commonly used in biomedical applications due to their ability to encapsulate small-molecules and improve their pharmacokinetic properties. Curcumin is known to have anti-inflammatory and antioxidant effects yet suffers from poor solubility and bioavailability. The purpose of this study is to investigate the efficacy of curcumin encapsulated in PAMAM dendrimers as a potential therapeutic treatment for ischemic stroke by studying post-stroke lesion size, astrocyte reactivity, and functional recovery in a rat model of cerebral ischemia.MethodsForty-eight male and female Sprague-Dawley rats (280–380 g) underwent either a 90-min middle cerebral artery occlusion (MCAo) or sham surgery before receiving one of four treatments: (1) Hanks’ balanced salt solution (HBSS) control, (2) empty dendrimer control, (3) curcumin control, or (4) curcumin encapsulated in PAMAM dendrimer. Neurobehavioral outcomes were evaluated at 1-, 3-, 5-, and 7-day post-surgery, after which animals were euthanized on day 8 to assess infarct volume and GFAP immunoreactivity.ResultsAnimals that received formulations containing dendrimers (curcumin encapsulated in dendrimers or empty dendrimers) demonstrated significantly lower levels of GFAP immunoreactivity and improved functional recovery, including weight and neurobehavioral scores, compared to the formulations that did not contain dendrimers (curcumin and HBSS control). Additionally, the dendrimer-curcumin treatment group exhibited a significantly improved paw laterality index over the course of the study compared with the other three treatment groups.ConclusionAlthough the post-stroke administration of curcumin encapsulated in PAMAM dendrimers modulates the astrocytic response and promotes functional recovery following ischemic stroke in rats, its therapeutic benefits may be driven by PAMAM dendrimers as the empty dendrimer treatment group also showed significant improvements post-stroke. Further investigation regarding PAMAM dendrimers in treating neuroinflammatory conditions remains warranted.
- Published
- 2025
- Full Text
- View/download PDF
30. Reducing neonatal Fc receptor binding enhances clearance and brain-to-blood ratio of TfR-delivered bispecific amyloid-β antibody
- Author
-
Eva Schlein, Ken G. Andersson, Tiffany Dallas, Stina Syvänen, and Dag Sehlin
- Subjects
Alzheimer’s disease (AD) ,amyloid-β (aβ) ,bispecific antibody ,blood-brain barrier (BBB) ,neonatal Fc receptor (FcRn) ,receptor mediated transcytosis (RMT) ,Therapeutics. Pharmacology ,RM1-950 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Recent development of amyloid-β (Aβ)-targeted immunotherapies for Alzheimer’s disease (AD) have highlighted the need for accurate diagnostic methods. Antibody-based positron emission tomography (PET) ligands are well suited for this purpose as they can be directed toward the same target as the therapeutic antibody. Bispecific, brain-penetrating antibodies can achieve sufficient brain concentrations, but their slow blood clearance remains a challenge, since it prolongs the time required to achieve a target-specific PET signal. Here, two antibodies were designed based on the Aβ antibody bapineuzumab (Bapi) – one monospecific IgG (Bapi) and one bispecific antibody with an antigen binding fragment (Fab) of the transferrin receptor (TfR) antibody 8D3 fused to one of the heavy chains (Bapi-Fab8D3) for active, TfR-mediated transport into the brain. A variant of each antibody was designed to harbor a mutation to the neonatal Fc receptor (FcRn) binding domain, to increase clearance. Blood and brain pharmacokinetics of radiolabeled antibodies were studied in wildtype (WT) and AD mice (AppNL-G-F). The FcRn mutation substantially reduced blood half-life of both Bapi and Bapi-Fab8D3. Bapi-Fab8D3 showed high brain uptake and the brain-to-blood ratio of its FcRn mutated form was significantly higher in AppNL-G-F mice than in WT mice 12 h after injection and increased further up to 168 h. Ex vivo autoradiography showed specific antibody retention in areas with abundant Aβ pathology. Taken together, these results suggest that reducing FcRn binding of a full-sized bispecific antibody increases the systemic elimination and could thereby drastically reduce the time from injection to in vivo imaging.
- Published
- 2024
- Full Text
- View/download PDF
31. IP-10 acts early in CV-A16 infection to induce BBB destruction and promote virus entry into the CNS by increasing TNF-α expression
- Author
-
Yajie Hu, Yunguang Hu, Anguo Yin, Yaming Lv, Jiang Li, Jingyuan Fan, Baojiang Qian, Jie Song, and Yunhui Zhang
- Subjects
hand foot and mouth disease (HFMD) ,coxsackievirus A16 (CV-A16) ,IFN-γ-inducible protein-10 (IP-10) ,blood-brain barrier (BBB) ,neuroinflammation ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The mechanisms underlying pathological changes in the central nervous system (CNS) following Coxsackievirus A16 (CV-A16) infection have not yet been elucidated. IFN-γ-inducible protein-10 (IP-10) is often used as a predictive factor to monitor early virus infection. It has also been reported that IP-10 plays a pivotal role in neuroinflammation. In this study, we aimed to explore the role of IP-10 in the neuropathogenesis of CV-A16 infection. We observed that the level of IP-10, as well as the TLR3-TRIF-TRAF3-TBK1-NF-κB and RIG-I/MDA5-MAVS-TRAFS-TBK1-NF-κB pathways, which are the upstream of IP-10, were significantly elevated during the course of CV-A16 infection. This increase was accompanied by an increase in a series of inflammatory cytokines at different time-points during CV-A16 infection. To determine whether IP-10 influences BBB integrity, we examined junctional complexes. Our results revealed that the expression levels of Claudin5, Occludin, ZO-1 and VE-Cadherin were notably decreased in CV-A16-infected HUVECs, but these indicators were restored in CV-A16-infected HUVECs with Eldelumab treatment. Nevertheless, IP-10 is only a chemokine that primarily traffics CXCR3-positive immune cells to inflammatory sites or promotes the production of inflammatory cytokines. Therefore, the interactions between IP-10 and inflammatory cytokines were evaluated. Our data revealed that IP-10 mediated the production of TNF-α, which was also observed to change the junctional complexes. Moreover, in a suckling mouse model, IP-10 and TNF-α treatments exacerbated clinical symptoms, mortality and pathological changes in the brain of CV-A16-infected mice, but Anti-IP-10 and Anti-TNF-α treatments alleviated these changes. Our data also revealed that IP-10 may be detected early in CV-A16 infection, whereas TNF-α was detected late in CV-A16 infection, and the production of TNF-α was also found to be positively correlated with IP-10. In addition, IP-10 and TNF-α were observed to reduce junctional complexes and enhance virus entry into the CNS. Taken together, this study provides the first evidence that CV-A16 activates the IP-10/TNF-α regulatory axis to cause BBB damage and accelerate the formation of neuroinflammation in infected hosts, which not only provides a new understanding of the neuropathogenesis caused by CV-A16, but also offers a promising target for the development of CV-A16 antiviral drugs.
- Published
- 2024
- Full Text
- View/download PDF
32. Enhancing Blood–Brain Barrier Integrity in Patients With Acute Ischemic Stroke Via Normobaric Hyperoxia
- Author
-
Weili Li, Wenbo Hu, Shuhua Yuan, Jiahao Chen, Qi Wang, Jiayue Ding, Zhiying Chen, Zhifeng Qi, and Ju Han
- Subjects
blood–brain barrier (BBB) ,contrast extravasation ,hemorrhagic transformation ,occludin ,serum biomarker ,stroke ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
BACKGROUND Recent advancements in animal studies have demonstrated the potential of normobaric hyperoxia (NBO) as a promising intervention for preserving the integrity of the blood–brain barrier (BBB). However, there is still limited understanding of the effects of NBO on BBB function in patients with clinical stroke. Therefore, the objective of this study was to investigate the efficacy of NBO therapy in attenuating BBB damage and reducing brain injury in individuals undergoing endovascular treatment (EVT) for acute stroke. METHODS AND RESULTS This study enrolled patients from the OPENS‐1 (Normobaric Hyperoxia Combined With Reperfusion for Acute Ischemic Stroke) study, with 43 patients receiving NBO combined with EVT and 43 patients receiving EVT alone. The main outcome measures included serum levels of occludin, MMP‐9 (matrix metalloproteinase‐9), NSE (neuron‐specific enolase), and S100b at 24 hours and 7 days, as well as the intracranial extravasation rate at 24 hours. Serum markers were assessed using ELISA, and intracranial contrast extravasation was visualized using dual‐energy computed tomography scan. We analyzed a total of 86 patients and found that the 24‐hour serum markers levels of BBB damage and brain injury were significantly lower in the group receiving NBO therapy combined with EVT compared with the group receiving EVT alone. Similarly, at 7 days, the levels of occludin, MMP‐9, and NSE were lower in the NBO+EVT group. We also found that the 24‐hour serum levels of occludin and MMP‐9 were correlated with intracranial contrast extravasation. Additionally, the incidence of intracranial contrast extravasation was lower in the NBO+EVT group compared with the EVT group (35.9% versus 60.5%, P=0.031). CONCLUSIONS This study offers valuable insights into the positive impact of NBO on maintaining BBB integrity and reducing brain injury in patients with acute stroke undergoing EVT.
- Published
- 2024
- Full Text
- View/download PDF
33. Molecular determinants for brain targeting by peptides: a meta-analysis approach with experimental validation
- Author
-
Marco Cavaco, Patrícia Fraga, Javier Valle, Ruben D. M. Silva, Lurdes Gano, João D. G. Correia, David Andreu, Miguel A. R. B. Castanho, and Vera Neves
- Subjects
Blood–brain barrier (BBB) ,Blood–brain barrier peptide shuttles (BBBpS) ,Brain delivery ,Cell-penetrating peptides (CPPs) ,Neurological disorders ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Blood–brain barrier (BBB) peptide-shuttles (BBBpS) are able to translocate the BBB and reach the brain. Despite the importance of brain targeting in pharmacology, BBBpS are poorly characterized. Currently, their development relies on the empiric assumption that cell-penetrating peptides (CPPs), with proven ability to traverse lipid membranes, will likewise behave as a BBBpS. The relationship between CPPs/BBBpS remains elusive and, to the best of our knowledge, has not hitherto been subject to thorough experimental scrutiny. In this work, we have identified/quantified the main physicochemical properties of BBBpS and then searched for CPPs with these properties, hence potential BBBpS. The specific features found for BBBpS are: (i) small size, (ii) none or few aromatic residues, (iii) hydrophobic, and (iv) slight cationic nature. Then, we selected the 10 scoring best in an ordinary least squares analysis, and tested them in vitro and in vivo. Overall, we identified the molecular determinants for brain targeting by peptides, devised a methodology that can be used to assist in the design of peptides with potential brain penetration from amino acid residue sequences, and found four new BBBpS within the CPP library. Graphical Abstract
- Published
- 2024
- Full Text
- View/download PDF
34. Transporter-mediated drug-drug interactions: regulatory guidelines, <italic>in vitro</italic> and <italic>in vivo</italic> methodologies and translation, special populations, and the blood-brain barrier.
- Author
-
Russell, Laura E., Yadav, Jaydeep, Maldonato, Benjamin J., Chien, Huan-Chieh, Zou, Ling, Vergara, Ana G., and Villavicencio, Erick G.
- Subjects
- *
DRUG interactions , *BLOOD-brain barrier , *GENETIC translation , *DRUG absorption , *DRUG development , *CYTOCHROME P-450 , *PHARMACOKINETICS - Abstract
AbstractThis review, part of a special issue on drug-drug interactions (DDIs) spearheaded by the International Society for the Study of Xenobiotics (ISSX) New Investigators, explores the critical role of drug transporters in absorption, disposition, and clearance in the context of DDIs. Over the past two decades, significant advances have been made in understanding the clinical relevance of these transporters. Current knowledge on key uptake and efflux transporters that affect drug disposition and development is summarized. Regulatory guidelines from the FDA, EMA, and PMDA that inform the evaluation of potential transporter-mediated DDIs are discussed in detail. Methodologies for preclinical and clinical testing to assess potential DDIs are reviewed, with an emphasis on the utility of physiologically based pharmacokinetic (PBPK) modeling. This includes the application of relative abundance and expression factors to predict human pharmacokinetics (PK) using preclinical data, integrating the latest regulatory guidelines. Considerations for assessing transporter-mediated DDIs in special populations, including pediatric, hepatic, and renal impairment groups, are provided. Additionally, the impact of transporters at the blood-brain barrier (BBB) on the disposition of CNS-related drugs is explored. Enhancing the understanding of drug transporters and their role in drug disposition and toxicity can improve efficacy and reduce adverse effects. Continued research is essential to bridge remaining gaps in knowledge, particularly in comparison with cytochrome P450 (CYP) enzymes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
35. Effect of Apolipoprotein E isoforms on the Abundance and Function of P-glycoprotein in Human Brain Microvascular Endothelial Cells.
- Author
-
Kreutzer, Ethan, Short, Jennifer L., and Nicolazzo, Joseph A.
- Subjects
- *
APOLIPOPROTEIN E , *ALZHEIMER'S disease , *APOLIPOPROTEIN E4 , *ENDOTHELIAL cells , *P-glycoprotein , *BLOOD-brain barrier - Abstract
Background: Individuals with Alzheimer's disease (AD) often require many medications; however, these medications are dosed using regimens recommended for individuals without AD. This is despite reduced abundance and function of P-glycoprotein (P-gp) at the blood-brain barrier (BBB) in AD, which can impact brain exposure of drugs. The fundamental mechanisms leading to reduced P-gp abundance in sporadic AD remain unknown; however, it is known that the apolipoprotein E (apoE) gene has the strongest genetic link to sporadic AD development, and apoE isoforms can differentially alter BBB function. The aim of this study was to assess if apoE affects P-gp abundance and function in an isoform-dependent manner using a human cerebral microvascular endothelial cell (hCMEC/D3) model. Methods: This study assessed the impact of apoE isoforms on P-gp abundance (by western blot) and function (by rhodamine 123 (R123) uptake) in hCMEC/D3 cells. Cells were exposed to recombinant apoE3 and apoE4 at 2 – 10 µg/mL over 24 – 72 hours. hCMEC/D3 cells were also exposed for 72 hours to astrocyte-conditioned media (ACM) from astrocytes expressing humanised apoE isoforms. Results: P-gp abundance in hCMEC/D3 cells was not altered by recombinant apoE4 relative to recombinant apoE3, nor did ACM containing human apoE isoforms alter P-gp abundance. R123 accumulation in hCMEC/D3 cells was also unchanged with recombinant apoE isoform treatments, suggesting no change to P-gp function, despite both abundance and function being altered by positive controls SR12813 (5 µM) and PSC 833 (5 µM), respectively. Conclusions: Different apoE isoforms have no direct influence on P-gp abundance or function within this model, and further in vivo studies would be required to address whether P-gp abundance or function are reduced in sporadic AD in an apoE isoform-specific manner. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. Exposure of quantum dots in the nervous system: Central nervous system risks and the blood–brain barrier interface.
- Author
-
Guan, Shujing and Tang, Meng
- Subjects
CENTRAL nervous system ,QUANTUM dots ,BLOOD-brain barrier ,POISONS ,MEDICAL sciences ,EVIDENCE gaps - Abstract
Quantum dots currently possess significant importance in the field of biomedical science. Upon introduction into the body, quantum dots exhibit a tendency to accumulate in diverse tissues including the central nervous system (CNS). Consequently, it becomes imperative to devote specific attention to their potential toxic effects. Moreover, the preservation of optimal CNS function relies heavily on blood–brain barrier (BBB) integrity, thereby necessitating its prioritization in neurotoxicological investigations. A more comprehensive understanding of the BBB and CNS characteristics, along with the underlying mechanisms that may contribute to neurotoxicity, will greatly aid researchers in the development of effective design strategies. This article offers an in‐depth look at the methods used to reduce the harmful effects of quantum dots on the nervous system, alongside the progression of effective treatments for brain‐related conditions. The focal point of this discussion is the BBB and its intricate association with the CNS and neurotoxicology. The discourse commences by recent advancements in the medical application of quantum dots are examined. Subsequently, elucidating the mechanisms through which quantum dots infiltrate the human body and traverse into the brain. Additionally, the discourse delves into the factors that facilitate the passage of quantum dots across the BBB, primarily encompassing the physicochemical properties of quantum dots and the BBB's inherent capacity for self‐permeability alteration. Furthermore, a concluding summary is presented, emphasizing existing research deficiencies and identifying promising avenues for further investigation within this field. This article explores quantum dots' biomedical applications, emphasizing their potential neurotoxicity and impact on the blood‐brain barrier (BBB). It discusses methods to mitigate quantum dots' harmful effects on the nervous system and highlights advancements in brain‐related treatments. The focus is on understanding BBB integrity, quantum dots' mechanisms of entry into the CNS, and factors influencing their passage across the barrier. The conclusion underscores research gaps and suggests future avenues for investigation in this field. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
37. Molecular determinants for brain targeting by peptides: a meta-analysis approach with experimental validation.
- Author
-
Cavaco, Marco, Fraga, Patrícia, Valle, Javier, Silva, Ruben D. M., Gano, Lurdes, Correia, João D. G., Andreu, David, Castanho, Miguel A. R. B., and Neves, Vera
- Subjects
NEUROPEPTIDES ,CELL-penetrating peptides ,AMINO acid residues ,BLOOD-brain barrier ,AMINO acid sequence - Abstract
Blood–brain barrier (BBB) peptide-shuttles (BBBpS) are able to translocate the BBB and reach the brain. Despite the importance of brain targeting in pharmacology, BBBpS are poorly characterized. Currently, their development relies on the empiric assumption that cell-penetrating peptides (CPPs), with proven ability to traverse lipid membranes, will likewise behave as a BBBpS. The relationship between CPPs/BBBpS remains elusive and, to the best of our knowledge, has not hitherto been subject to thorough experimental scrutiny. In this work, we have identified/quantified the main physicochemical properties of BBBpS and then searched for CPPs with these properties, hence potential BBBpS. The specific features found for BBBpS are: (i) small size, (ii) none or few aromatic residues, (iii) hydrophobic, and (iv) slight cationic nature. Then, we selected the 10 scoring best in an ordinary least squares analysis, and tested them in vitro and in vivo. Overall, we identified the molecular determinants for brain targeting by peptides, devised a methodology that can be used to assist in the design of peptides with potential brain penetration from amino acid residue sequences, and found four new BBBpS within the CPP library. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. Current immunotherapeutic approaches to diffuse intrinsic pontine glioma.
- Author
-
Lin, Catherine, Smith, Christian, and Rutka, James
- Subjects
BLOOD-brain barrier ,CHILD death ,GLIOMAS ,BREAST ,BRAIN tumors ,CLINICAL drug trials ,TECHNOLOGICAL innovations ,CHIMERIC antigen receptors - Abstract
Diffuse intrinsic pontine glioma (DIPG) is an aggressive brain tumour that occurs in the pons of the brainstem and accounts for over 80% of all brainstem gliomas. The median age at diagnosis is 6--7 years old, with less than 10% overall survival 2 years after diagnosis and less than 1% after 5 years. DIPGs are surgically inaccessible, and radiation therapy provides only transient benefit, with death ensuing from relentless local tumour infiltration. DIPGs are now the leading cause of brain tumour deaths in children, with a societal cancer burden in years of life lost (YLL) of more than 67 per individual, versus approximately 14 and 16 YLL for lung and breast cancer respectively. More than 95 clinical drug trials have been conducted on children with DIPGs, and all have failed to improve survival. No single or combination chemotherapeutic strategy has been successful to date because of our inability to identify targeted drugs for this disease and to deliver these drugs across an intact blood-brain barrier (BBB). Accordingly, there has been an increased focus on immunotherapy research in DIPG, with explorations into treatments such as chimeric antigen receptor T (CAR-T) cells, immune checkpoint blockades, cancer vaccines, and autologous cell transfer therapy. Here, we review the most recent advances in identifying genetic factors influencing the development of immunotherapy for DIPG. Additionally, we explore emerging technologies such as Magnetic Resonance-guided Focused Ultrasound (MRgFUS) in potential combinatorial approaches to treat DIPG. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
39. Editorial: Immunosuppression mechanisms and immunotherapy strategies in glioblastoma.
- Author
-
Sihan Xiong, Bing Qin, Chuang Liu, and Yuanbo Pan
- Subjects
GLIOBLASTOMA multiforme ,CENTRAL nervous system tumors ,BLOOD-brain barrier ,BEVACIZUMAB ,MACROPHAGE colony-stimulating factor ,IMMUNOTHERAPY ,PROGNOSIS ,T cell receptors - Abstract
This article discusses the challenges and potential strategies for treating glioblastoma (GBM), an aggressive type of brain tumor. It explores the immunosuppressive tumor microenvironment and the blood-brain barrier as barriers to effective treatment. The article discusses various immunotherapy strategies and other approaches for preclinical studies. It also highlights the role of neutrophils and macrophages in promoting tumor growth. The article emphasizes the complexity of GBM and the need for a multidisciplinary approach to improve patient outcomes. The document provides a list of references that offer a comprehensive overview of current research on GBM treatment and the need for further investigation into novel therapeutic approaches. [Extracted from the article]
- Published
- 2024
- Full Text
- View/download PDF
40. Blood–Brain Barrier-Targeting Nanoparticles: Biomaterial Properties and Biomedical Applications in Translational Neuroscience.
- Author
-
Asimakidou, Evridiki, Tan, Justin Kok Soon, Zeng, Jialiu, and Lo, Chih Hung
- Subjects
- *
BIOMOLECULES , *BLOOD-brain barrier , *SURFACE charges , *NANOPARTICLES , *POLYETHYLENE glycol , *NEUROSCIENCES - Abstract
Overcoming the blood–brain barrier (BBB) remains a significant hurdle in effective drug delivery to the brain. While the BBB serves as a crucial protective barrier, it poses challenges in delivering therapeutic agents to their intended targets within the brain parenchyma. To enhance drug delivery for the treatment of neurological diseases, several delivery technologies to circumvent the BBB have been developed in the last few years. Among them, nanoparticles (NPs) are one of the most versatile and promising tools. Here, we summarize the characteristics of NPs that facilitate BBB penetration, including their size, shape, chemical composition, surface charge, and importantly, their conjugation with various biological or synthetic molecules such as glucose, transferrin, insulin, polyethylene glycol, peptides, and aptamers. Additionally, we discuss the coating of NPs with surfactants. A comprehensive overview of the common in vitro and in vivo models of the BBB for NP penetration studies is also provided. The discussion extends to discussing BBB impairment under pathological conditions and leveraging BBB alterations under pathological conditions to enhance drug delivery. Emphasizing the need for future studies to uncover the inherent therapeutic properties of NPs, the review advocates for their role beyond delivery systems and calls for efforts translating NPs to the clinic as therapeutics. Overall, NPs stand out as a highly promising therapeutic strategy for precise BBB targeting and drug delivery in neurological disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
41. The Future Prospects in Effective Management of Glioblastoma Multiforme Using Exosomes: A Comprehensive Review
- Author
-
Bhaskaran, Mahendran, C, Surabhi, Nesamony, Jerry, Chelliah, Selvam, Chidambaram, Saravana Babu, Section editor, and Essa, Musthafa M., editor
- Published
- 2024
- Full Text
- View/download PDF
42. Toxicity Studies of Exosomes and Potential Overcome Approaches
- Author
-
Chandra, Phool, Ali, Zeeshan, Fatma, Nishat, Sachan, Neetu, Mishra, Neeraj, editor, Ashique, Sumel, editor, Garg, Ashish, editor, Chithravel, Vadivalagan, editor, and Anand, Krishnan, editor
- Published
- 2024
- Full Text
- View/download PDF
43. Blood-Brain Barrier (BBB)-Crossing Strategies for Improved Treatment of CNS Disorders
- Author
-
Zhang, Wandong, Michel, Martin C., Editor-in-Chief, Barrett, James E., Editorial Board Member, Centurión, David, Editorial Board Member, Flockerzi, Veit, Editorial Board Member, Meier, Kathryn Elaine, Editorial Board Member, Page, Clive P., Editorial Board Member, Seifert, Roland, Editorial Board Member, Wang, KeWei, Editorial Board Member, Schäfer-Korting, Monika, editor, and Schubert, Ulrich S., editor
- Published
- 2024
- Full Text
- View/download PDF
44. Poly(Butyl Cyanoacrylate) Nanoparticles Deliver β-Nerve Growth Factor to the Brain After Traumatic Brain Injury
- Author
-
Lin, Yong, Turksen, Kursad, Series Editor, Peplow, Philip V., editor, Martinez, Bridget, editor, and Gennarelli, Thomas A., editor
- Published
- 2024
- Full Text
- View/download PDF
45. The impacts of tobacco and nicotine on HIV-1 infection, inflammation, and the blood-brain barrier in the central nervous system
- Author
-
Aislinn M. Keane and Talia H. Swartz
- Subjects
HIV-1 ,NLRP3 inflammasome ,neuroinflammation ,nicotine ,blood-brain barrier (BBB) ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Human immunodeficiency virus (HIV-1) remains a persistent global health crisis. Even while successfully virologically suppressed, people with HIV (PWH) experience a higher risk for inflammatory disorders such as HIV-associated neurocognitive disorder (HAND). Tobacco use puts PWH at higher risk for neurocognitive symptoms resulting from HIV-associated neuroinflammation. The NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome has been implicated as a driver of HIV-associated inflammation, including HAND. Nicotine, the psychoactive component of tobacco smoke, has also been shown to signal through the NLRP3 inflammasome and modulate inflammatory signaling in the CNS. Here, we explore the impacts of nicotine and tobacco on the complex neurobiology of HAND, including effects on cognition, inflammation, viral latency, and blood-brain barrier integrity. We outline nicotine’s role in the establishment of active and latent infection in the brain and posit the NLRP3 inflammasome as a common pathway by which HIV-1 and nicotine promote neuroinflammation in PWH.
- Published
- 2024
- Full Text
- View/download PDF
46. Mesothelin promotes brain metastasis of non-small cell lung cancer by activating MET
- Author
-
Shengkai Xia, Wenzhe Duan, Mingxin Xu, Mengqi Li, Mengyi Tang, Song Wei, Manqing Lin, Encheng Li, Wenwen Liu, and Qi Wang
- Subjects
MSLN ,NSCLC ,Brain metastasis (BM) ,Blood-brain barrier (BBB) ,MET ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Brain metastasis (BM) is common among cases of advanced non-small cell lung cancer (NSCLC) and is the leading cause of death for these patients. Mesothelin (MSLN), a tumor-associated antigen expressed in many solid tumors, has been reported to be involved in the progression of multiple tumors. However, its potential involvement in BM of NSCLC and the underlying mechanism remain unknown. Methods The expression of MSLN was validated in clinical tissue and serum samples using immunohistochemistry and enzyme-linked immunosorbent assay. The ability of NSCLC cells to penetrate the blood-brain barrier (BBB) was examined using an in vitro Transwell model and an ex vivo multi-organ microfluidic bionic chip. Immunofluorescence staining and western blotting were used to detect the disruption of tight junctions. In vivo BBB leakiness assay was performed to assess the barrier integrity. MET expression and activation was detected by western blotting. The therapeutic efficacy of drugs targeting MSLN (anetumab) and MET (crizotinib/capmatinib) on BM was evaluated in animal studies. Results MSLN expression was significantly elevated in both serum and tumor tissue samples from NSCLC patients with BM and correlated with a poor clinical prognosis. MSLN significantly enhanced the brain metastatic abilities of NSCLC cells, especially BBB extravasation. Mechanistically, MSLN facilitated the expression and activation of MET through the c-Jun N-terminal kinase (JNK) signaling pathway, which allowed tumor cells to disrupt tight junctions and the integrity of the BBB and thereby penetrate the barrier. Drugs targeting MSLN (anetumab) and MET (crizotinib/capmatinib) effectively blocked the development of BM and prolonged the survival of mice. Conclusions Our results demonstrate that MSLN plays a critical role in BM of NSCLC by modulating the JNK/MET signaling network and thus, provides a potential novel therapeutic target for preventing BM in NSCLC patients.
- Published
- 2024
- Full Text
- View/download PDF
47. Amyloid β oligomer induces cerebral vasculopathy via pericyte-mediated endothelial dysfunction
- Author
-
Siqi Chen, Daji Guo, Yuanyuan Zhu, Songhua Xiao, Jiatian Xie, Zhan Zhang, Yu Hu, Jialin Huang, Xueying Ma, Zhiyuan Ning, Lin Cao, Jinping Cheng, and Yamei Tang
- Subjects
Alzheimer’s disease ,Aβ oligomer ,Blood–brain barrier (BBB) ,Pericytes ,Tight junction proteins ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Although abnormal accumulation of amyloid beta (Aβ) protein is thought to be the main cause of Alzheimer’s disease (AD), emerging evidence suggests a pivotal vascular contribution to AD. Aberrant amyloid β induces neurovascular dysfunction, leading to changes in the morphology and function of the microvasculature. However, little is known about the underlying mechanisms between Aβ deposition and vascular injuries. Recent studies have revealed that pericytes play a substantial role in the vasculopathy of AD. Additional research is imperative to attain a more comprehensive understanding. Methods Two-photon microscopy and laser speckle imaging were used to examine cerebrovascular dysfunction. Aβ oligomer stereotactic injection model was established to explain the relationship between Aβ and vasculopathy. Immunofluorescence staining, western blot, and real-time PCR were applied to detect the morphological and molecular alternations of pericytes. Primary cultured pericytes and bEnd.3 cells were employed to explore the underlying mechanisms. Results Vasculopathy including BBB damage, hypoperfusion, and low vessel density were found in the cortex of 8 to 10-month-old 5xFAD mice. A similar phenomenon accompanied by pericyte degeneration appeared in an Aβ-injected model, suggesting a direct relationship between Aβ and vascular dysfunction. Pericytes showed impaired features including low PDGFRβ expression and increased pro-inflammatory chemokines secretion under the administration of Aβ in vitro, of which supernatant cultured with bEND.3 cells led to significant endothelial dysfunction characterized by TJ protein deficiency. Conclusions Our results provide new insights into the pathogenic mechanism underlying Aβ-induced vasculopathy. Targeting pericyte therapies are promising to ameliorate vascular dysfunction in AD.
- Published
- 2024
- Full Text
- View/download PDF
48. Transforming brain cancer therapeutics: unlocking the power of blood-brain barrier-targeting strategies for superior treatment outcomes and precision medicine.
- Author
-
Viswanathan, Akshaya, Brahma, Neha, and S, Vimal
- Subjects
- *
INTRANASAL administration , *TARGETED drug delivery , *TREATMENT effectiveness , *BRAIN tumors , *BRAIN cancer - Abstract
The treatment of brain tumors is significantly hindered by the Blood-Brain Barrier (BBB), a selective barrier that restricts the passage of therapeutic agents to the brain. Recent advancements in BBB-targeting therapies offer promising strategies to overcome this challenge, providing new avenues for the effective treatment of brain cancer. This article reviews innovative approaches, including Convection-Enhanced Delivery (CED) and RNA-based therapeutics, which enhance drug delivery directly to tumor sites, bypassing the BBB and reducing systemic toxicity. Additionally, the use of theranostic nanoparticles and CRISPR-Cas9 gene editing presents novel opportunities for real-time monitoring and precision-targeted therapy, respectively. Techniques such as magnetic nanoparticles, intranasal drug administration, and focused ultrasound with microbubbles are also being refined to improve drug penetration across the BBB. Furthermore, peptide-based delivery systems and small molecules designed to mimic endogenous transport pathways are accelerating the discovery of more effective therapies. The exploration of combination therapies that synergize BBB-penetrant drugs with conventional chemotherapeutic agents or immunotherapies holds the potential to enhance treatment efficacy and patient outcomes. Continued research and interdisciplinary collaboration are essential to develop predictive models, personalized treatment strategies, and alternative delivery methods that ensure the long-term safety and effectiveness of these novel therapies. Advancements in BBB-targeting therapeutics are poised to transform the landscape of brain cancer treatment, offering renewed hope for improved survival rates and quality of life for patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
49. Exploring molecular mechanisms of postoperative delirium through multi-omics strategies in plasma exosomes
- Author
-
Yan, Fuhui, Chen, Bowang, Ma, Zhen, Chen, Qirong, Jin, Zhi, Wang, Yujie, Qu, Feng, and Meng, Qiang
- Published
- 2024
- Full Text
- View/download PDF
50. The Progress in Molecular Transport and Therapeutic Development in Human Blood–Brain Barrier Models in Neurological Disorders
- Author
-
Korszun-Karbowniczak, Joanna, Krysiak, Zuzanna Joanna, Saluk, Joanna, Niemcewicz, Marcin, and Zdanowski, Robert
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.