Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
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
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (3,200)

Search Parameters:
Keywords = blood–brain barrier

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 436 KiB  
Review
Mitochondrial Dysfunction: Effects and Therapeutic Implications in Cerebral Gliomas
by Gerardo Caruso, Roberta Laera, Rosamaria Ferrarotto, Cristofer Gonzalo Garcia Moreira, Rajiv Kumar, Tamara Ius, Giuseppe Lombardi and Maria Caffo
Medicina 2024, 60(11), 1888; https://doi.org/10.3390/medicina60111888 - 18 Nov 2024
Viewed by 258
Abstract
Gliomas are the most common primary brain tumors, representing approximately 28% of all central nervous system tumors. These tumors are characterized by rapid progression and show a median survival of approximately 18 months. The therapeutic options consist of surgical resection followed by radiotherapy [...] Read more.
Gliomas are the most common primary brain tumors, representing approximately 28% of all central nervous system tumors. These tumors are characterized by rapid progression and show a median survival of approximately 18 months. The therapeutic options consist of surgical resection followed by radiotherapy and chemotherapy. Despite the multidisciplinary approach and the biomolecular role of targeted therapies, the median progression-free survival is approximately 6–8 months. The incomplete tumor compliance with treatment is due to several factors such as the presence of the blood–brain barrier, the numerous pathways involved in tumor transformation, and the presence of intra-tumoral mutations. Among these, the interaction between the mutations of genes involved in tumor bio-energetic metabolism and the functional response of the tumor has become the protagonist of numerous studies. In this scenario, the main role is played by mitochondria, cellular organelles delimited by a double membrane and containing their own DNA (mtDNA), which participates in numerous cellular processes such as the regulation of cellular metabolism, cellular proliferation, and apoptosis and is also the main source of cellular energy production. Therefore, it is understood that the mitochondrion, specifically its functional alteration, is a leading figure in tumor transformation, including brain tumors. The acquisition of mutations in the mitochondrial DNA of tumor cells and the subsequent identification of the so-called mitochondria-related genes (MRGs), both functional (mutation of Complex I) and structural (mutations of Complex III/IV), have been seen to play an important role in metabolic reprogramming with increased proliferation, resistance to apoptosis, and the progression of tumorigenesis. This demonstrates that these mitochondrial alterations could have a role not only in the intrinsic tumor biology but also in the extrinsic one associated with the therapeutic response. We aim to summarize the main mitochondrial dysfunction interactions present in gliomas and how they might impact prognosis. Full article
(This article belongs to the Section Neurology)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of mitochondrial structures and molecular pathways involved in ATP production.</p>
Full article ">
20 pages, 1005 KiB  
Review
The Involvement of Glial Cells in Blood–Brain Barrier Damage in Neuroimmune Diseases
by Satoshi Nagata and Ryo Yamasaki
Int. J. Mol. Sci. 2024, 25(22), 12323; https://doi.org/10.3390/ijms252212323 - 17 Nov 2024
Viewed by 450
Abstract
The blood–brain barrier and glial cells, particularly astrocytes, interact with each other in neuroimmune diseases. In the inflammatory environment typical of these diseases, alterations in vascular endothelial cell surface molecules and weakened cell connections allow immune cells and autoantibodies to enter the central [...] Read more.
The blood–brain barrier and glial cells, particularly astrocytes, interact with each other in neuroimmune diseases. In the inflammatory environment typical of these diseases, alterations in vascular endothelial cell surface molecules and weakened cell connections allow immune cells and autoantibodies to enter the central nervous system. Glial cells influence the adhesion of endothelial cells by changing their morphology and releasing various signaling molecules. Multiple sclerosis has been the most studied disease in relation to vascular endothelial and glial cell interactions, but these cells also significantly affect the onset and severity of other neuroimmune conditions, including demyelinating and inflammatory diseases. In this context, we present an overview of these interactions and highlight how they vary across different neuroimmune diseases. Full article
Show Figures

Figure 1

Figure 1
<p>Interactions between the BBB and glial cells. Glial cells have either protective or disruptive effects, or both, on the BBB. Abbreviations: bFGF, basic fibroblast growth factor; ApoE, apolipoprotein E; S1P, sphingosine 1-phosphate; Slc4a4, solute carrier family 4 member 4; CCL, CC chemokine ligand; IL, interleukin; Shh, sonic hedgehog; RA, retinoic acid; PRDX, peroxiredoxin; Cx, connexin; HMGB, high mobility group box; VEGF, vascular endothelial growth factor; TYMP, thymidine phosphorylase; ATP, adenosine triphosphate; EphA4, ephrin A4; TNF, tumor necrosis factor; ROS, reactive oxygen species; TREM, triggering receptor of myeloid cells; Ang, angiotensin; MIF, migration inhibitory factor; ECs, endothelial cells.</p>
Full article ">
23 pages, 6586 KiB  
Article
Studies Regarding Antimicrobial Properties of Some Microbial Polyketides Derived from Monascus Strains
by Daniela Albisoru, Nicoleta Radu, Lucia Camelia Pirvu, Amalia Stefaniu, Narcisa Băbeanu, Rusandica Stoica and Dragos Paul Mihai
Antibiotics 2024, 13(11), 1092; https://doi.org/10.3390/antibiotics13111092 - 16 Nov 2024
Viewed by 464
Abstract
Finding new molecules to prevent the growth of antimicrobial resistance is a hot topic for scientists worldwide. It has been reported that some raw bioproducts containing Monascus polyketides have antimicrobial activities, but extensive studies on this effect have not been conducted. In this [...] Read more.
Finding new molecules to prevent the growth of antimicrobial resistance is a hot topic for scientists worldwide. It has been reported that some raw bioproducts containing Monascus polyketides have antimicrobial activities, but extensive studies on this effect have not been conducted. In this context, our studies aimed to evaluate the antimicrobial properties of six raw bioproducts containing three classes of microbial polyketides biosynthesized by three Monascus strains through solid-state biosynthesis. As a methodology, we performed in silico predictions using programs such as PyMOL v3.0.4 and employed ESI-MS techniques to provide evidence of the presence of the six studied compounds in our bioproducts. The results obtained in silico were validated through in vitro studies using the Kirby-Bauer diffusion method on bacteria and fungi. The test performed in silico showed that Monascorubramine has the highest affinity for both Gram-positive and Gram-negative bacteria, followed by yellow polyketides such as Ankaflavin and Monascin. The estimated pharmacokinetic parameters indicated high gastrointestinal absorption and the potential to cross the blood-brain barrier for all studied compounds. However, the compounds also inhibit most enzymes involved in drug metabolism, presenting some level of toxicity. The best in vitro results were obtained for S. aureus, with an extract containing yellow Monascus polyketides. Predictions made for E. coli were validated in vitro for P. aeruginosa, S. enterica, and S. marcescens, as well as for fungi. Significant antibacterial properties were observed during this study for C. albicans, S. aureus, and fungal dermatophytes for crude bioproducts containing Monascus polyketides. In conclusion, the antimicrobial properties of Monascus polyketides were validated both in silico and in vitro. However, due to their potential toxicity, these bioproducts would be safer to use as topical formulations. Full article
Show Figures

Figure 1

Figure 1
<p>Experimental study design.</p>
Full article ">Figure 2
<p>Molecular docking validation—superposition of predicted poses (pink) of co-crystallized inhibitors on initial conformations (green): (<b>a</b>) trimethoprim in saDHFR binding site (PDB ID: 2w9s, RMSD 0.6535 Å); (<b>b</b>) trimethoprim in ecDHFR binding site (PDB ID: 7mym, RMSD 0.3521 Å); (<b>c</b>) UCP11E in caDHFR binding site (PDB ID: 4hoe, RMSD 0.4389 Å); (<b>d</b>) trimethoprim in hDHFR binding site (PDB ID: 2w3a, RMSD 0.9559 Å).</p>
Full article ">Figure 3
<p>Predicted binding poses of Monascorubramine in DHFR active sites. (<b>a</b>) saDHFR; (<b>b</b>) ecDHFR; (<b>c</b>) caDHFR; (<b>d</b>) hDHFR.</p>
Full article ">Figure 4
<p>2D diagrams of predicted molecular interactions between Monascorubramine and active sites of DHFR homologues. (<b>a</b>) saDHFR; (<b>b</b>) ecDHFR; (<b>c</b>) caDHFR; (<b>d</b>) hDHFR.</p>
Full article ">Figure 5
<p>“Boiled egg” diagram illustrating the distribution of the investigated compounds in the chemical space of molecules that are absorbed in the gastrointestinal (GI) tract or passively permeate the blood–brain barrier (BBB) based on calculated WlogP (octanol/water partition coefficient) and TPSA (topological polar surface area) values. Molecules located in the “egg yolk” are predicted to passively permeate through the BBB. Molecules located in the white area are predicted to be passively absorbed in the GI tract.</p>
Full article ">Figure 6
<p>ESI-MS analysis of a total alcoholic extract of the following: (<b>a</b>) <span class="html-italic">Monascus purpureus</span>; (<b>b</b>) <span class="html-italic">Monascus ruber</span>; (<b>c</b>) <span class="html-italic">Monascus</span> sp. 3 <span class="html-italic">(Monascus ruber</span>; highly productive).</p>
Full article ">Figure 7
<p>Antibacterial properties of polyketides obtained from Monascus-derived bioproducts: (<b>a</b>) antibacterial properties for <span class="html-italic">S. aureus</span> (yellow polyketides exhibit the best activities); (<b>b</b>) antibacterial properties for <span class="html-italic">S. aureus</span> MRSA (yellow polyketides exhibit moderate activities); (<b>c</b>) antibacterial properties for <span class="html-italic">S. marcescens</span> (red polyketides exhibit the best activities); (<b>d</b>) antibacterial properties for <span class="html-italic">P. aeruginosa</span> (red polyketides exhibit moderate antimicrobial activities); (<b>e</b>) antibacterial properties for <span class="html-italic">S. enterica</span> (red polyketides exhibit local-moderate antimicrobial activities).</p>
Full article ">Figure 7 Cont.
<p>Antibacterial properties of polyketides obtained from Monascus-derived bioproducts: (<b>a</b>) antibacterial properties for <span class="html-italic">S. aureus</span> (yellow polyketides exhibit the best activities); (<b>b</b>) antibacterial properties for <span class="html-italic">S. aureus</span> MRSA (yellow polyketides exhibit moderate activities); (<b>c</b>) antibacterial properties for <span class="html-italic">S. marcescens</span> (red polyketides exhibit the best activities); (<b>d</b>) antibacterial properties for <span class="html-italic">P. aeruginosa</span> (red polyketides exhibit moderate antimicrobial activities); (<b>e</b>) antibacterial properties for <span class="html-italic">S. enterica</span> (red polyketides exhibit local-moderate antimicrobial activities).</p>
Full article ">Figure 8
<p>Antifungal properties of polyketides obtained from Monascus-derived bioproducts for the following: (<b>a</b>) <span class="html-italic">Candida albicans</span>; (<b>b</b>) <span class="html-italic">S. brevicaulis</span>, (<b>c</b>) <span class="html-italic">M. gypseum</span>; (<b>d</b>) <span class="html-italic">T. mentagrophytes</span>.</p>
Full article ">Figure 9
<p>Flow diagram used to obtain enhanced extracts of yellow, orange, and red polyketides: (<b>a</b>) Solid-state biosynthesis of <span class="html-italic">Monascus</span> bioproducts (RYR); (<b>b</b>) Sample preparation of <span class="html-italic">Monascus</span> bioproducts for analysis; (<b>c</b>) Obtaining <span class="html-italic">Monascus</span> extract with yellow polyketides; (<b>d</b>) Obtaining <span class="html-italic">Monascus</span> extract with orange polyketides; (<b>e</b>) Obtaining <span class="html-italic">Monascus</span> extract with red polyketides.</p>
Full article ">Figure 9 Cont.
<p>Flow diagram used to obtain enhanced extracts of yellow, orange, and red polyketides: (<b>a</b>) Solid-state biosynthesis of <span class="html-italic">Monascus</span> bioproducts (RYR); (<b>b</b>) Sample preparation of <span class="html-italic">Monascus</span> bioproducts for analysis; (<b>c</b>) Obtaining <span class="html-italic">Monascus</span> extract with yellow polyketides; (<b>d</b>) Obtaining <span class="html-italic">Monascus</span> extract with orange polyketides; (<b>e</b>) Obtaining <span class="html-italic">Monascus</span> extract with red polyketides.</p>
Full article ">
12 pages, 1594 KiB  
Review
Exploring the Connection Between Nanomaterials and Neurodegenerative Disorders
by Sitansu Sekhar Nanda and Dong Kee Yi
Micromachines 2024, 15(11), 1382; https://doi.org/10.3390/mi15111382 - 15 Nov 2024
Viewed by 414
Abstract
Drug delivery, tissue engineering, and cell promotion in biomedical fields heavily rely on the use of nanomaterials (NMs). When they penetrate cells, NPs undergo degradation and initiate the generation of reactive oxygen species (ROS) by causing changes in the structures of organelles linked [...] Read more.
Drug delivery, tissue engineering, and cell promotion in biomedical fields heavily rely on the use of nanomaterials (NMs). When they penetrate cells, NPs undergo degradation and initiate the generation of reactive oxygen species (ROS) by causing changes in the structures of organelles linked to mitochondria. Inside the cell, the excess production of ROS can initiate a chain reaction, along with the autophagy process that helps maintain ROS balance by discarding unnecessary materials. At present, there is no effective treatment for Alzheimer’s disease (AD), a progressive neurodegenerative disease. The use of NMs for siRNA delivery could become a promising treatment for AD and other CNS disorders. Recent research demonstrates that the use of combined NPs can induce autophagy in cells. This article emphasizes the importance of the shape of siRNA-encapsulated NMs in determining their efficiency in delivering and suppressing gene activity in the central nervous system. Because of its strict selectivity against foreign substances, the blood–brain barrier (BBB) significantly hinders the delivery of therapeutic agents to the brain. Conventional chemotherapeutic drugs are significantly less effective against brain cancers due to this limitation. As a result, NMs have become a promising approach for targeted drug delivery, as they can be modified to carry specific ligands that direct them to their intended targets. This review thoroughly examines the latest breakthroughs in using NMs to deliver bioactive compounds across the BBB, focusing on their use in cancer treatments. The review starts by examining the structure and functions of the BBB and BBTB, and then emphasizes the benefits that NMs offer. Full article
(This article belongs to the Section B3: Nanoparticles in Biomedicine)
Show Figures

Figure 1

Figure 1
<p>A summary of how induced pluripotent stem cells are used to model the blood–brain barrier. This figure was adapted from reference [<a href="#B5-micromachines-15-01382" class="html-bibr">5</a>] with permission.</p>
Full article ">Figure 2
<p>(<b>A</b>) Models employed to examine nanocarriers’ passage across the BBB. (<b>B</b>) Steps to construct in vivo BBB models. This figure was adapted from reference [<a href="#B9-micromachines-15-01382" class="html-bibr">9</a>] with permission.</p>
Full article ">Figure 3
<p>The Human Protein Atlas provides information on the tissue-level protein expression of APP, BACE1, presenilin-1, and tau, indicating potential for Aβ production. This figure was adapted from reference [<a href="#B20-micromachines-15-01382" class="html-bibr">20</a>] with permission.</p>
Full article ">Figure 4
<p>The biological composition of (<b>A</b>) the blood–brain barrier (BBB) and (<b>B</b>) the blood–brain tumor barrier (BBTB). This figure was adapted from reference [<a href="#B69-micromachines-15-01382" class="html-bibr">69</a>] with permission.</p>
Full article ">
17 pages, 666 KiB  
Review
Propionic Acid Impact on Multiple Sclerosis: Evidence and Challenges
by Lorena Lorefice and Magdalena Zoledziewska
Nutrients 2024, 16(22), 3887; https://doi.org/10.3390/nu16223887 - 14 Nov 2024
Viewed by 447
Abstract
Accumulating evidence suggests that multiple sclerosis (MS) is an environmentally influenced disorder with contributions from life-time exposure to factors including Epstein–Barr virus infection or shifts in microbiome, diet and lifestyle. One suggested factor is a deficiency in propionic acid, a short-chain fatty acid [...] Read more.
Accumulating evidence suggests that multiple sclerosis (MS) is an environmentally influenced disorder with contributions from life-time exposure to factors including Epstein–Barr virus infection or shifts in microbiome, diet and lifestyle. One suggested factor is a deficiency in propionic acid, a short-chain fatty acid produced by gut bacteria that may contribute to the disease pathology both in animal models and in human cases of MS. Propionate appears to exert beneficial effects on the immune, peripheral and central nervous systems of people with MS (pwMS), showing immunoregulatory, neuroprotective and neurogenerative effects. These functions are crucial, given that MS is characterized by immune-mediated damage of myelin in the central nervous system. Accordingly, propionate supplementation or a modulated increase in its levels through the microbiome and diet may help counteract the pro-inflammatory state in MS by directly regulating immune system and/or by decreasing permeability of gut barrier and blood–brain barrier. This could potentially improve outcomes when used with immune-modulating therapy. However, while its broad effects are promising, further large clinical trials are necessary to evaluate its efficacy and safety in pwMS and clarify its role as a complementary therapeutic strategy. This review provides a comprehensive analysis of the evidence, challenges and limitations concerning propionic acid supplementation in MS. Full article
(This article belongs to the Section Nutritional Immunology)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of propionate’s functions, effects and supplementation in MS.</p>
Full article ">
17 pages, 6143 KiB  
Article
Derivatives of Amodiaquine as Potent Human Cholinesterases Inhibitors: Implication for Treatment of Alzheimer’s Disease
by Ana Matošević, Dejan M. Opsenica, Marija Bartolić, Nikola Maraković, Andriana Stoilković, Katarina Komatović, Antonio Zandona, Suzana Žunec and Anita Bosak
Molecules 2024, 29(22), 5357; https://doi.org/10.3390/molecules29225357 - 14 Nov 2024
Viewed by 275
Abstract
As some previously reported studies have proven that amodiaquine, in addition to its primary antimalarial activity, also has potential for new applications such as the inhibition of cholinesterases, in our study we focused on the evaluation of the influence of different substituents in [...] Read more.
As some previously reported studies have proven that amodiaquine, in addition to its primary antimalarial activity, also has potential for new applications such as the inhibition of cholinesterases, in our study we focused on the evaluation of the influence of different substituents in the aminoquinoline part of the amodiaquine structure on the inhibition of human acetylcholinesterase and butyrylcholinesterase to investigate the possibility for their use as drugs for the treatment of AD. We synthesized a series of amodiaquine derivatives bearing H-, F-, CF3-, NO2-, CN-, CO2H- or CH3O- groups on the aminoquinoline ring, and determined that all of the tested derivatives were very potent inhibitors of both cholinesterases, with inhibition constants (Ki) in the nM and low μM range and with prominent selectivity (up to 300 times) for the inhibition of acetylcholinesterase. All compounds displayed an ability to chelate biometal ions Fe2+, Zn2+ and Cu2+ and an antioxidant power comparable to that of standard antioxidants. Most of the compounds were estimated to be able to cross the blood–brain barrier by passive transport and were nontoxic toward cells that represent the models of individual organs. Considering all these beneficial features, our study has singled out compound 5, the most potent AChE inhibitor with a CH3O- on C(7) position, followed by 6 and 14, compounds without substituent or hydroxyl groups in the C(17) position, respectively, as the most promising compounds from the series which could be considered as potential multi-target drugs for the treatment of AD. Full article
Show Figures

Figure 1

Figure 1
<p>The general structure of amodiaquine derivatives. Numbers in brackets refer to the numeration of carbon atoms in the molecule.</p>
Full article ">Figure 2
<p>Superposition of modelled complexes of AChE and AMQ (carbons and interactions in blue) and compound <b>1</b> (carbons and interactions in red) (<b>A</b>), AMQ (carbons and interactions in blue) and compound <b>5</b> (carbons and interactions in red) (<b>B</b>), compound <b>2</b> (carbons and interactions in blue) and compound <b>11</b> (carbons and interactions in red) (<b>C</b>), and AMQ (carbons and interactions in blue) and compound <b>14</b> (carbons and interactions in magenta) (<b>D</b>).</p>
Full article ">Figure 3
<p>Superposition of modelled complexes of BChE and <b>10</b> (carbons and interactions in magenta) and compound <b>11</b> (carbos and interactions in blue).</p>
Full article ">Figure 4
<p>Determined FRAP values. Columns represent the FRAP values for a 10 μM concentration of each amodiaquine derivative.</p>
Full article ">Figure 5
<p>(<b>A</b>) The UV-VIS spectra of compound <b>13</b> (purple) and after mixing with Cu<sup>2+</sup> (green), Zn<sup>2+</sup> (blue) and Fe<sup>2+</sup> (red). (<b>B</b>) The differential spectra of compound <b>13</b>–metal complex for Cu<sup>2+</sup> (green), Zn<sup>2+</sup> (blue) and Fe<sup>2+</sup> complexes (red).</p>
Full article ">Figure 6
<p>Radar plot of physicochemical properties of amodiaquine derivatives. M<sub>w</sub> stands for molecular weight, logP for hydrophobicity, HBD for number of hydrogen bond donors, HBA for hydrogen bond acceptors, RB for number of rotatable bonds and PSA for polar surface area.</p>
Full article ">Figure 7
<p>Schematic presentation of comprehensive analysis of the results.</p>
Full article ">Scheme 1
<p>Hypothesized mechanism of the transformation of CN to CO<sub>2</sub>H in the quinoline system under mild conditions.</p>
Full article ">Scheme 2
<p>Synthesis of amodiaquine derivatives <b>1</b>–<b>14</b>.</p>
Full article ">
25 pages, 2482 KiB  
Review
The Immune Escape Strategy of Rabies Virus and Its Pathogenicity Mechanisms
by Abraha Bahlbi Kiflu
Viruses 2024, 16(11), 1774; https://doi.org/10.3390/v16111774 - 14 Nov 2024
Viewed by 837
Abstract
In contrast to most other rhabdoviruses, which spread by insect vectors, the rabies virus (RABV) is a very unusual member of the Rhabdoviridae family, since it has evolved to be fully adapted to warm-blooded hosts and spread directly between them. There are differences [...] Read more.
In contrast to most other rhabdoviruses, which spread by insect vectors, the rabies virus (RABV) is a very unusual member of the Rhabdoviridae family, since it has evolved to be fully adapted to warm-blooded hosts and spread directly between them. There are differences in the immune responses to laboratory-attenuated RABV and wild-type rabies virus infections. Various investigations showed that whilst laboratory-attenuated RABV elicits an innate immune response, wild-type RABV evades detection. Pathogenic RABV infection bypasses immune response by antagonizing interferon induction, which prevents downstream signal activation and impairs antiviral proteins and inflammatory cytokines production that could eliminate the virus. On the contrary, non-pathogenic RABV infection leads to immune activation and suppresses the disease. Apart from that, through recruiting leukocytes into the central nervous system (CNS) and enhancing the blood–brain barrier (BBB) permeability, which are vital factors for viral clearance and protection, cytokines/chemokines released during RABV infection play a critical role in suppressing the disease. Furthermore, early apoptosis of neural cells limit replication and spread of avirulent RABV infection, but street RABV strains infection cause delayed apoptosis that help them spread further to healthy cells and circumvent early immune exposure. Similarly, a cellular regulation mechanism called autophagy eliminates unused or damaged cytoplasmic materials and destroy microbes by delivering them to the lysosomes as part of a nonspecific immune defense mechanism. Infection with laboratory fixed RABV strains lead to complete autophagy and the viruses are eliminated. But incomplete autophagy during pathogenic RABV infection failed to destroy the viruses and might aid the virus in dodging detection by antigen-presenting cells, which could otherwise elicit adaptive immune activation. Pathogenic RABV P and M proteins, as well as high concentration of nitric oxide, which is produced during rabies virus infection, inhibits activities of mitochondrial proteins, which triggers the generation of reactive oxygen species, resulting in oxidative stress, contributing to mitochondrial malfunction and, finally, neuron process degeneration. Full article
(This article belongs to the Special Issue Viral Infections and Immune Dysregulation 2024)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>). RABV virion. Rabies virus is enveloped, bullet-shaped, with a body size of about 180 × 75 nm. (<b>B</b>). RABV genome. The RNA genome of the RABV is single-stranded and non-segmented, with a genome size of about 12 kb. It contains a leader and trailer region at the 3′ and 5′ ends, along with five structural proteins (N, P, M, G, and L) and four intergenic non-coding sequences. Multiple P proteins (P (P1), P2, P3, P4 and P5) are produced through alternative initiation from in-frame AUG start codons due to a leaky scanning mechanism. (Original source of the image; Philippe Le Mercier, SIB Swiss Institute of Bioinformatics) [<a href="#B9-viruses-16-01774" class="html-bibr">9</a>].</p>
Full article ">Figure 2
<p>The various interactions between RABV proteins and interferon signaling pathway. The IFN-signaling pathway is activated by RABV infection, whereas RABV utilizes its own proteins to suppress the IFN signaling pathway by interacting with key factors in IFN pathways. RABV P interacts directly with tyrosine-phosphorylated STAT (pY-STAT) to inhibit the expression of ISGs (interferon-stimulated genes) by affecting its localization and reducing its ability to bind to ISG promoters. P protein is also able to interact directly with TBK-1 (TANK-binding kinase-1) in a dose-dependent manner, inhibiting phosphorylation of the IRF3 (interferon regulatory factor 3). Moreover, the interaction between P and PML (promyelocytic leukemia) alters PML protein localization and the structure of PML-NBs (nuclear bodies), thereby regulating IFN-induced apoptosis. The encapsulation of RABV RNA by N protein protects viral RNA from getting recognized by RIG-I (retinoic acid–inducible gene I), which subsequently prevents RIG-I-mediated activation of the downstream IRF-3 pathway. Type-I IFN stimulation causes the M to shift toward activated pSTAT1 interaction, which improves the ability of P protein to engage with JAK1 to prevent pSTAT1 from activating and with pSTAT1 to restrain it in the cytoplasm [<a href="#B43-viruses-16-01774" class="html-bibr">43</a>].</p>
Full article ">Figure 3
<p>The method by which neural production of CXCL10 caused by laboratory-attenuated rabies virus (RABV) enhances the blood–brain barrier (BBB) permeability. 1. Chemokine CXCL10 is secreted by laboratory-attenuated RABV infected neurons; 2. CXCL10 mediates CD4<sup>+</sup> T cell recruitment into the CNS; 3. CXCL10 mediates CD4<sup>+</sup> T cell differentiation into Th1 and Th17 cells; 4a, IFN-γ secreting Th1 cells could further boost the induction of CXCL10 through positive feedback; 4b, IL-17 secreting Th17 cells alters the TJ (tight junction) proteins leading to Breakdown of the blood–brain barrier [<a href="#B67-viruses-16-01774" class="html-bibr">67</a>].</p>
Full article ">Figure 4
<p>Mechanisms of mitochondrial dysfunction associated with rabies virus infection. In mitochondria of rabies virus-infected neurons, the activity of electron transport system (ETS) complexes I and IV are increased either due to direct (e.g., interaction with a rabies virus protein) and/or indirect (e.g., high NADH/NAD<sup>+</sup> ratio and up-regulation of sirtuin activity) viral effect. The increased proton pumping across mitochondrial membranes generates a high mitochondrial membrane potential (MMP). Electron leakage during both forward and reverse ETS leads to superoxide (O<sub>2</sub><sup>−</sup>) formation that is dismutated by mitochondrial superoxide dismutase (SOD<sub>2</sub>) to hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>). Both O<sub>2</sub><sup>−</sup> and H<sub>2</sub>O<sub>2</sub> may lower the intracellular ATP levels by increasing hydrolysis of ATP molecules. Overproduction and accumulation of O<sub>2</sub><sup>−</sup> and H<sub>2</sub>O<sub>2</sub> induces oxidative stress and leads to degeneration of neuronal processes (solid lines findings, dashed lines hypothesis) [<a href="#B110-viruses-16-01774" class="html-bibr">110</a>].</p>
Full article ">Figure 5
<p>Intramitochondrial metabolism of nitric oxide and superoxide radicals. The steady-state concentrations are indicated. Oxygen and nitrogen-reactive species are kept in biological systems at steady-state concentrations that can be estimated by using the steady-state approach with the assumption that the rate of production is equal to the rate of utilization. The primary production of O<sub>2</sub><sup>−</sup>, cytosolic Cu-Zn-SOD and mitochondrial Mn-SOD keep steady-state concentrations of 10<sup>−10</sup> M in the mitochondrial matrix and 10<sup>−11</sup> M in the cytosol. The cytosolic steady-state concentration of H<sub>2</sub>O<sub>2</sub> estimated from the rate of H<sub>2</sub>O<sub>2</sub> generation by subcellular sources and its removal by catalase and glutathione peroxidase is about 10<sup>−7</sup>–10<sup>−8</sup> M. considering the rate of H<sub>2</sub>O<sub>2</sub> production, its removal by intra-mitochondrial glutathione peroxidase and its diffusion to the cytosolic space, H<sub>2</sub>O<sub>2</sub> steady-state concentration in the mitochondrial matrix results in approximately 10<sup>−8</sup> M. The balance between NO production and its utilization by the reactions with the components of the respiratory chain and with O<sub>2</sub><sup>−</sup> regulates the intra-mitochondrial steady-state concentration of NO at about 50 nM, which in turn regulates mitochondrial oxygen uptake and energy supply [<a href="#B137-viruses-16-01774" class="html-bibr">137</a>].</p>
Full article ">Figure 6
<p>Modes of mitochondrial operation that lead to O<sub>2</sub><sup>• −</sup> production. There are three modes of mitochondrial operation that are associated with O<sub>2</sub><sup>• −</sup> production. In mode 1, the NADH pool is reduced, for example by a damage to the respiratory chain, loss of cytochrome c during apoptosis or low ATP demand. This leads to a rate of O<sub>2</sub><sup>• −</sup> formation at the FMN of complex I that is determined by the extent of FMN reduction which is in turn set by the NADH/NAD<sup>+</sup> ratio. In mode 2, there is no ATP production and there is a high proton motive force and a reduced CoQ (Ubiquinone) pool which leads to reverse electron transport (RET) through complex I, producing large amounts of O<sub>2</sub><sup>• −</sup>. In mode 3, mitochondria are actively making ATP and consequently have a lower change in membrane proton motive (MPM) force than in mode 2 and a more oxidized NADH pool than in mode 1. Under these conditions, the flux of O<sub>2</sub><sup>• −</sup> within mitochondria is far lower than in modes 1 and 2, and the O<sub>2</sub><sup>• −</sup> sources are unclear [<a href="#B143-viruses-16-01774" class="html-bibr">143</a>].</p>
Full article ">
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
Viewed by 356
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

Figure 1
<p>Schematic diagram of study design and rationales by Figdraw. (<b>A</b>) Rationale for using fluorescent dextran to determine BBB integrity. The situations of single fluorescent (FITC) dextran perfusion and dual fluorescent (FITC &amp; Texas Red) dextran perfusion are shown on the left and right side of the dotted line, respectively. The numbers in the vessels represent the FITC concentration (left side of dotted line) and the FITC-Dextran to Texas Red-Dextran ratio (right side). (<b>B</b>) The timeline of the in vivo experiment.</p>
Full article ">Figure 2
<p>Performance of 2% NaNO<sub>2</sub> perfusion (<span class="html-italic">n</span> = 3). (<b>A</b>) Hemoglobin concentration of mice perfused with PBS or 2% sodium nitrite after CO<sub>2</sub> euthanasia. Hemoglobin concentration in each group was normalized to that of the no perfusion group. (<b>B</b>) K<sub>p,b</sub> of human IgG1, (<b>C</b>) FITC-Dextran concentration in the brain tissue, and (<b>D</b>) the ration of FITC-Dextran to Texas Red-Dextran after PBS and 2% NaNO<sub>2</sub> perfusion. * Represents significant differences (<span class="html-italic">p</span> &lt; 0.05); ** is <span class="html-italic">p</span> &lt; 0.01, *** is <span class="html-italic">p</span> &lt; 0.001; and **** is <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Evaluation of the performance of vasodilators’ perfusion (<span class="html-italic">n</span> = 3). (<b>A</b>) Hemoglobin concentration, (<b>B</b>) FITC-Dextran concentration, (<b>C</b>) Texas Red-Dextran concentration, and (<b>D</b>) the FITC/Texas Red ratio in the brain tissue were measured and analyzed for each perfusion group as mice were euthanized by CO<sub>2</sub>. The superscripted ** in (<b>A</b>) represent significant differences (<span class="html-italic">p</span> &lt; 0.01) compared with the PBS group (one-way ANOVA with post hoc Dunnett’s test). Except for the 2% NaNO<sub>2</sub> group, no significant difference in the FITC/Texas Red ratio was observed between PBS and other groups (one-way ANOVA with post hoc Dunnett’s test <span class="html-italic">t</span>). To verify the impact of NaNO<sub>2</sub> perfusion on BBB integrity, mice were perfused by the Evans Blue in 1% BSA solution followed by 2-min PBS perfusion. (<b>E</b>) The photograph of brain tissue was captured immediately after perfusion. **** represents significant differences (<span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 4
<p>Optimization of perfusion procedure including (<b>A</b>,<b>B</b>) induction of unconsciousness, (<b>C</b>) perfusion flow rate, and (<b>D</b>) osmotic pressure. (<b>A</b>,<b>B</b>) Before PBS perfusion, compared to CO<sub>2</sub> euthanasia, using Ket/Xyl to anesthetize the mouse significantly reduced hemoglobin concentration in the brain without changing the FITC/Texas Red ratio (unpaired Student’s <span class="html-italic">t</span>-test). (<b>C</b>) The PBS perfusion decreased hemoglobin concentration to the lowest level when perfusion rate was 10 mL/min, although there was no significant difference in the hemoglobin concentration among groups (one-way ANOVA). (<b>D</b>) The 100 mM NaNO2 in PBS solution (hypertonic, 463 mOsmol/L) removed more hemoglobin from the brain vessels (unpaired Student’s <span class="html-italic">t</span>-test). * Represents significant differences (<span class="html-italic">p</span> &lt; 0.05). The “ns” is an abbreviation for “not significant”.</p>
Full article ">Figure 5
<p>Hemoglobin concentration in different tissues (<span class="html-italic">n</span> = 3), including (<b>A</b>) brain, (<b>B</b>) liver, (<b>C</b>) kidney, and (<b>D</b>) spleen. Mice were anesthetized by Ket/Xyl and perfused by different solutions at 10 mL/min in the experiment. (<b>A</b>–<b>C</b>) The 100 mM NaNO<sub>2</sub> was the most effective at reducing hemoglobin concentration in brain, liver and kidney. (<b>D</b>) In the spleen, the hemoglobin concentration was extremely high in all groups (33.3–77.8%). The superscripted *** in (<b>C</b>) represent significant differences (<span class="html-italic">p</span> &lt; 0.001) compared with the PBS group.</p>
Full article ">Figure 6
<p>Fluorescent dextran concentrations in different tissues (<span class="html-italic">n</span> = 3). Compared to PBS group, 100 mM NaNO<sub>2</sub> increased FITC-Dextran concentration in the (<b>A</b>) brain, (<b>D</b>) liver, and (<b>J</b>) spleen, but not in the (<b>G</b>) kidney. Regarding Texas Red-Dextran, 100 mM NaNO<sub>2</sub> increased its concentration in all tissues (<b>B</b>,<b>E</b>,<b>H</b>,<b>K</b>). As a result, there was minor difference in FITC/Texas Red ratio between groups in the (<b>C</b>) brain and (<b>F</b>) liver. However, in the (<b>I</b>) kidney and (<b>L</b>) spleen, a significant decrease in 100 mM NaNO<sub>2</sub> group was observed compared with PBS group. (One-way ANOVA with post hoc Tukey’s test.) * Represents significant differences (<span class="html-italic">p</span> &lt; 0.05); and ** is <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 7
<p>K<sub>p,b</sub> and K<sub>p,t</sub> of human IgG1 and transferrin (<b>A</b>–<b>D</b>). Regarding K<sub>p,b</sub> and K<sub>p,t</sub> of hIgG1 and hTf in the brain and liver, perfusion reduced the K<sub>p,b</sub> and K<sub>p,t</sub> value in comparison of no perfusion group. However, no difference was found between perfusion groups (<b>E</b>,<b>F</b>). For the kidney, compared to PBS perfusion, 100 mM NaNO<sub>2</sub> reduced K<sub>p,t</sub> of hIgG1 and hTf without significance. (<b>G</b>,<b>H</b>) 100 mM NaNO<sub>2</sub> significantly lowered the K<sub>p,t</sub> of hIgG1 in the spleen but not for K<sub>p,t</sub> of hTf compared with PBS group (one-way ANOVA with post hoc Dunnett’s test). *** Represents significant differences (<span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">
13 pages, 2259 KiB  
Case Report
Post-Carotid Artery Stenting Hyperperfusion Syndrome in a Hypotensive Patient: Case Report and Systematic Review of Literature
by Matija Zupan, Matej Perovnik, Janja Pretnar Oblak and Senta Frol
Life 2024, 14(11), 1472; https://doi.org/10.3390/life14111472 - 12 Nov 2024
Viewed by 541
Abstract
Cerebral hyperperfusion syndrome (CHS) is a serious post-procedural complication of carotid artery stenting (CAS). The pathophysiological mechanisms of CHS in the absence of arterial hypertension (AH) remain only partially understood. We performed a systematic literature search of the PubMed database using the terms [...] Read more.
Cerebral hyperperfusion syndrome (CHS) is a serious post-procedural complication of carotid artery stenting (CAS). The pathophysiological mechanisms of CHS in the absence of arterial hypertension (AH) remain only partially understood. We performed a systematic literature search of the PubMed database using the terms »cerebral hyperperfusion syndrome«, »hypotension«, »hyperperfusion«, »stroke«, »intracranial hemorrhages«, »risk factors«, »carotid revascularization«, »carotid stenting«, »carotid endarterectomy«, »blood-brain barrier«, »endothelium«, »contrast encephalopathy«, and combinations. We present a case of a normotensive female patient who developed CHS post-CAS for symptomatic carotid stenosis while being hypotensive with complete recovery. We identified 393 papers, among which 65 were deemed relevant to the topic. The weighted average prevalence of CHS after CAS is 1.2% [0.0–37.7%] with that of intracranial hemorrhage (ICH) being 0.51% [0–9.3%]. Recently symptomatic carotid stenosis or contralateral carotid revascularization, urgent intervention, acute carotid occlusion, contralateral ≥70% stenosis, and the presence of leptomeningeal collaterals were associated with CHS. A prolonged hemodynamic instability after CAS conveys a higher risk for CHS. However, none of the articles mentioned isolated hypotension as a risk factor for CHS. Whereas mortality after ICH post-CAS ranges from 40 to 75%, in the absence of ICH, CHS generally carries a good prognosis. AH is not obligatory in CHS development. Even though impaired cerebral autoregulation and post-revascularization changes in cerebral hemodynamics seem to play a pivotal role in CHS pathophysiology, our case highlights the complexity of CHS, involving factors like endothelial dysfunction and sudden reperfusion. Further research is needed to refine diagnostic and management approaches for this condition. Full article
(This article belongs to the Section Medical Research)
Show Figures

Figure 1

Figure 1
<p>Imaging before neurological worsening. (<b>A</b>) A CT pre-procedure showing postischemic encephalomalacia in the left frontal region (arrow). (<b>B</b>,<b>C</b>) A CT angiography (CTA) pre-procedure showing a hemodynamically significant left internal carotid artery (ICA) stenosis of 80–90% (arrows). (<b>D</b>) A CTA pre-procedure showing a patent left posterior communicating artery. (<b>E</b>) A DSA showing a 50% left ICA stenosis (arrow). (<b>F</b>) The DSA showed a patent stent without residual stenosis at the end of the procedure (arrow).</p>
Full article ">Figure 2
<p>Neurological worsening. (<b>A</b>,<b>B</b>) A post-procedure CT showing sulcal effacement in the left cerebral hemisphere denoting edema (arrows). (<b>C</b>) A CTA post-procedure showing patent intracranial arteries. (<b>D</b>) A CTA post-procedure showing a patent stent in the left ICA (arrow). (<b>E</b>) A CT perfusion (CTP) post-procedure showing attenuated cerebral blood flow (CBF) in the left temporal region (circle). (<b>F</b>) The CTP shows attenuated cerebral blood volume (CBV) in the left temporal region (circle). (<b>G</b>) A CTP showing delayed TMAX in the left temporal region (circle). (<b>H</b>) A follow-up CT two days post-procedure denoting the improvement of the edema in the left hemisphere. (<b>I</b>) A follow-up DUS one month post-procedure showing a patent stent in the left ICA with normal hemodynamics.</p>
Full article ">Figure 3
<p>Flowchart of the selection procedure according to the PRISMA2020 guidelines.</p>
Full article ">
45 pages, 2120 KiB  
Review
Functional Food Nutrients, Redox Resilience Signaling and Neurosteroids for Brain Health
by Maria Scuto, Miroslava Majzúnová, Gessica Torcitto, Silvia Antonuzzo, Francesco Rampulla, Eleonora Di Fatta and Angela Trovato Salinaro
Int. J. Mol. Sci. 2024, 25(22), 12155; https://doi.org/10.3390/ijms252212155 - 12 Nov 2024
Viewed by 375
Abstract
The interplay between functional food nutrients and neurosteroids has garnered significant attention for its potential to enhance stress resilience in health and/or disease. Several bioactive nutrients, including medicinal herbs, flavonoids, and bioavailable polyphenol-combined nanoparticles, as well as probiotics, vitamin D and omega-3 fatty [...] Read more.
The interplay between functional food nutrients and neurosteroids has garnered significant attention for its potential to enhance stress resilience in health and/or disease. Several bioactive nutrients, including medicinal herbs, flavonoids, and bioavailable polyphenol-combined nanoparticles, as well as probiotics, vitamin D and omega-3 fatty acids, have been shown to improve blood–brain barrier (BBB) dysfunction, endogenous neurosteroid homeostasis and brain function. These nutrients can inhibit oxidative stress and neuroinflammation, which are linked to the pathogenesis of various neurological disorders. Interestingly, flavonoids exhibit dose-dependent effects, activating the nuclear factor erythroid 2–related factor 2 (Nrf2) pathway at the physiological/low dose (neurohormesis). This leads to the upregulation of antioxidant phase II genes and proteins such as heme oxygenase-1 (HO-1) and sirtuin-1 (Sirt1), which are activated by curcumin and resveratrol, respectively. These adaptive neuronal response mechanisms help protect against reactive oxygen species (ROS) and neurotoxicity. Impaired Nrf2 and neurosteroid hormone signaling in the brain can exacerbate selective vulnerability to neuroinflammatory conditions, contributing to the onset and progression of neurodegenerative and psychiatric disorders, including Alzheimer’s disease, anxiety and depression and other neurological disorders, due to the vulnerability of neurons to stress. This review focuses on functional food nutrients targeting Nrf2 antioxidant pathway and redox resilience genes to regulate the neurosteroid homeostasis and BBB damage associated with altered GABAergic neurotransmission. By exploring the underlying molecular mechanisms using innovative technologies, we aim to develop promising neuroprotective strategies and personalized nutritional and neuroregenerative therapies to prevent or attenuate oxidative stress and neuroinflammation, ultimately promoting brain health. Full article
(This article belongs to the Special Issue The Role of Hormones and Nutrients in Health and Disease)
Show Figures

Figure 1

Figure 1
<p>Neurohormetic nutrients and neurosteroids promote brain health through the activation of Nrf2 pathway and GABARs. Neurohormetic nutrients, including polyphenols (resveratrol, Hidrox<sup>®</sup>, sulforaphane, curcumin and 3-epigallocatechin gallate) but also probiotics and ω3 fatty acids (EPA and DHA), protect neurons against oxidative injury and neuroinflammation in a dose-dependent manner. Interestingly, moderate/low doses of food nutrients can modulate the antioxidant pathway and stress resilience genes and proteins, particularly, Hsp70 and HO-1, γ-GCs, Sirt1 and FOXO3, which efficiently remove ROS and provide neuroprotection during CNS disorders. In addition, functional nutrients induce brain health by upregulating GABA via GABARs activation. Likewise, neurosteroids such as vitamin D, 17β-estradiol, DHEA, DHEAS, progesterone and particularly allopregnanolone are potent neuromodulators of GABA through GABARs. The activation of GABARs inhibits neuroinflammatory cytokine cascade and microglia activation, thereby preventing BBB dysfunction and the onset and progression of neurodegenerative and psychiatric disorders. We hypothesize that the synergistic action of neurohormetic nutrients and neurosteroids could potentiate their neuroprotective and neuroregenerative effects through a potential crosstalk between the Nrf2 pathway and GABAergic signaling via GABARs for the prevention and therapy of CNS disorders, ultimately promoting brain health and longevity in humans.</p>
Full article ">Figure 2
<p>Potential molecular pathways ↑ upregulated or ↓ downregulated by neurosteroids in nervous system disorders in a dose-dependent manner (Neurohormesis).</p>
Full article ">Figure 3
<p>Overview of the potential crosstalk between Nrf2 pathway and GABAergic signaling activated by functional nutrients and neurosteroids through GABARs for neuroprotection and neuro-regenerative therapy via neurohormetic dose–response effects. GAD: glutamic acid decarboxylase. ↑ upregulated and ↓ downregulated.</p>
Full article ">
17 pages, 1662 KiB  
Article
Kratom Alkaloids: A Blood–Brain Barrier Specific Membrane Permeability Assay-Guided Isolation and Cyclodextrin Complexation Study
by András Dohárszky, Erika Mária Vági, Árpád Könczöl, Alexandra Simon, Erzsébet Várnagy, Miras Muratov, Kristóf István Steiger, Bianka Várnai, Szabolcs Béni, Eszter Riethmüller and Ida Fejős
Molecules 2024, 29(22), 5302; https://doi.org/10.3390/molecules29225302 - 9 Nov 2024
Viewed by 702
Abstract
Mitragynine is an “atypic opioid” analgesic with an alternative mechanism of action and a favorable side-effect profile. Our aim was to optimize the alkaloid extraction procedure from kratom leaves and to determine and isolate the most relevant compounds capable of penetrating the central [...] Read more.
Mitragynine is an “atypic opioid” analgesic with an alternative mechanism of action and a favorable side-effect profile. Our aim was to optimize the alkaloid extraction procedure from kratom leaves and to determine and isolate the most relevant compounds capable of penetrating the central nervous system. The PAMPA-BBB study revealed that mitragynine and its coalkaloids, speciociliatine, speciogynine, and paynantheine, possess excellent in vitro BBB permeability. An optimized sequence of CPC, flash chromatography, and preparative HPLC methods was used to isolate the four identified BBB+ alkaloids. To improve the bioavailability of the isolated alkaloids, their cyclodextrin (CD) complexation behavior was investigated via affinity capillary electrophoresis using almost 40 CD derivatives. The apparent alkaloid–CD complex stability constants were determined and compared, and the most relevant CDs phase-solubility studies were also performed. Both the neutral and negatively charged derivatives were able to form complexes with all four kratom alkaloids. It was found that cavity size, substituent type, and degree of substitution also influenced complex formation. The negatively charged Sugammadex, Subetadex, and the sufoalkylated-beta-CD analogs were able to form the most stable complexes, exceeding 1000 M−1. These results serve as a good basis for further solubility and stability enhancement studies of kratom alkaloids. Full article
(This article belongs to the Section Natural Products Chemistry)
Show Figures

Figure 1

Figure 1
<p>Comparison of the extraction procedures (Soxhlet extraction, maceration, ultrasound-assisted extraction (UAE), and supercritical CO<sub>2</sub> extraction (SFE)) applying methanol (MeOH) and ethanol (EtOH) regarding the yield and the mitragynine content of the extracts. Detailed results and further conditions can be found in <a href="#app1-molecules-29-05302" class="html-app">Table S2 (in the Supplementary Materials)</a> and in <a href="#sec3dot2-molecules-29-05302" class="html-sec">Section 3.2</a>.</p>
Full article ">Figure 2
<p>Demonstrative UHPLC-UV chromatogram of UAE MeOH (50 °C, 1:10 feed to solvent ratio) extract (red trace, top) and of PAMPA-BBB acceptor phase (green trace, bottom), along with the chemical structure of the kratom alkaloids detected in the acceptor phase recorded at 226 nm.</p>
Full article ">Figure 3
<p>(<b>a</b>) Schematic structure of the cyclodextrins (R stands for substituents), (<b>b</b>) representative electropherograms of mitragynine (M)–sulfobutyl-ether-β-cyclodextrin DS~6.5 complexes in the presence of increasing cyclodextrin concentrations using dimethyl sulfoxide as the electroosmotic flow (EOF) marker. Different colours indicate different cyclodextrin concentrations. Additional conditions can be found in <a href="#sec3dot7-molecules-29-05302" class="html-sec">Section 3.7</a>.</p>
Full article ">Figure 4
<p>A<sub>L</sub>- and A<sub>N</sub>-type isotherms of mitragynine–cyclodextrin complexes. Additional conditions and cyclodextrin abbreviations can be found in <a href="#sec3dot1-molecules-29-05302" class="html-sec">Section 3.1</a> and <a href="#sec3dot8-molecules-29-05302" class="html-sec">Section 3.8</a>. The R<sup>2</sup> values for each fitting are listed in <a href="#molecules-29-05302-t003" class="html-table">Table 3</a>.</p>
Full article ">
14 pages, 3172 KiB  
Review
A Review of Sturge–Weber Syndrome Brain Involvement, Cannabidiol Treatment and Molecular Pathways
by Katharine Elizabeth Joslyn, Nicholas Flinn Truver and Anne Marie Comi
Molecules 2024, 29(22), 5279; https://doi.org/10.3390/molecules29225279 - 8 Nov 2024
Viewed by 524
Abstract
Sturge–Weber syndrome (SWS) is a rare congenital neurocutaneous disorder typically caused by a somatic mosaic mutation in R183Q GNAQ. At-risk children present at birth with a capillary malformation port-wine birthmark. The primary diagnostic characteristic of the disorder includes leptomeningeal enhancement of the [...] Read more.
Sturge–Weber syndrome (SWS) is a rare congenital neurocutaneous disorder typically caused by a somatic mosaic mutation in R183Q GNAQ. At-risk children present at birth with a capillary malformation port-wine birthmark. The primary diagnostic characteristic of the disorder includes leptomeningeal enhancement of the brain, which demonstrates abnormal blood vessels and results in impaired venous drainage and impaired local cerebral perfusion. Impaired cerebral blood flow is complicated by seizures resulting in strokes, hemiparesis and visual field deficits, hormonal deficiencies, behavioral impairments, and intellectual disability. Therefore, anti-seizure medication in combination with low-dose aspirin is a common therapeutic treatment strategy. Recently published data indicate that the underlying mutation in endothelial cells results in the hyperactivation of downstream pathways and impairment of the blood–brain barrier. Cannabidiol (CBD) has been used to treat medically refractory seizures in SWS due to its anti-seizure, anti-inflammatory, and neuroprotective properties. Pilot research suggests that CBD improves cognitive impairment, emotional regulation, and quality of life in patients with SWS. Recent preclinical studies also suggest overlapping molecular pathways in SWS and in CBD, suggesting that CBD may be uniquely effective for SWS brain involvement. This review aims to summarize early data on CBD’s efficacy for preventing and treating epilepsy and neuro-cognitive impairments in patients with SWS, likely molecular pathways impacted, and provide insights for future translational research to improve clinical treatment for patients with SWS. Full article
(This article belongs to the Special Issue Recent Advances in Cannabis and Hemp Research)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Patient 1: T1-weighted post-contrast axial MRI showing unilateral right leptomeningeal enhancement (box) and enlarged choroid plexus (arrow). (<b>b</b>) Patient 1: Susceptibility-weighted axial MRI showing right occipital and temporal lobe cortical calcification and dilated deep draining vessels (box). (<b>c</b>) Patient 2: Infant with SWS presenting a unilateral left-sided facial PWB and glaucoma.</p>
Full article ">Figure 2
<p>Diagram of cannabidiol in the context of common anti-seizure medications prescribed in SWS (not to scale). Overall, CBD has a pro-inhibitory effect via various receptor pathways. As proposed by Rosenberg et al. [<a href="#B43-molecules-29-05279" class="html-bibr">43</a>], GPR55 promotes the downregulation of GABA<sub>A</sub> receptor densities via the breakdown of gephyrin scaffolding; however, this effect is blocked by CBD. Activation of TRPV1 by CBD may regulate vasodilation and astrocytic movement post-seizure. Additionally, the regulation of the adenosine transporter (ENT-1) may play a bigger role in SWS by promoting both GABA release via A2A and AMPA endocytosis via A1. Adapted from Löscher and Klein 2021 [<a href="#B44-molecules-29-05279" class="html-bibr">44</a>]. Abbreviations: CBD (Cannabidiol); GABA (gamma-aminobutyric acid); GABA<sub>A</sub> (gamma-aminobutyric acid receptor A); AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor); NMDA (N-methyl-D-aspartate receptor); TRPV1 (transient receptor potential vanilloid 1); GAD (glutamic acid decarboxylase); GAT-1 (gamma-aminobutyric acid transporter 1); A1 (adenosine A1 receptor); A2A (adenosine receptor subtype 2A); ENT-1 (equilibrative nucleoside transporter 1); GPR55 (G-protein coupled receptor 55); SV2A (Synaptic vesicle glycoprotein 2A).</p>
Full article ">Figure 3
<p>The canonical Gαq pathway is dysregulated by the R183Q somatic mutation that likely causes capillary malformations in the brain, skin, and eye and dysregulation of cellular mechanisms, thereby resulting in symptoms of SWS. Red arrows indicate an upregulated path in the R183Q GNAQ mutation; blue arrows indicate a blocked or downregulated path in the mutation. Since the αq remains in the active form, PLCβ excessively cleaves PIP2 and recursively activates downstream targets. Unidentified are the compensatory changes that happen naturally that mitigate this process, given that SWS is not cancerous. Abbreviations: GPCR (G-protein coupled receptor); GDP (Guanosine diphosphate); GTP (Guanosine triphosphate); PLCß (Phospholipase C beta); PIP2 (Phosphatidylinositol 4,5-bisphosphate); DAG (Diacylglycerol); PKC (Protein kinase C); MEK (Mitogen-activated protein kinase kinase); ERK (Extracellular signal-regulated kinase); MAPK (Mitogen-activated protein kinase); mTOR (mammalian target of rapamycin); IP3 (Inositol trisphosphate).</p>
Full article ">
26 pages, 1192 KiB  
Review
From Microcirculation to Aging-Related Diseases: A Focus on Endothelial SIRT1
by Martin Law, Pei-Chun Wang, Zhong-Yan Zhou and Yu Wang
Pharmaceuticals 2024, 17(11), 1495; https://doi.org/10.3390/ph17111495 - 7 Nov 2024
Viewed by 483
Abstract
Silent information regulator sirtuin 1 (SIRT1) is an NAD+-dependent deacetylase with potent anti-arterial aging activities. Its protective function in aging-related diseases has been extensively studied. In the microcirculation, SIRT1 plays a crucial role in preventing microcirculatory endothelial senescence by suppressing inflammation and oxidative [...] Read more.
Silent information regulator sirtuin 1 (SIRT1) is an NAD+-dependent deacetylase with potent anti-arterial aging activities. Its protective function in aging-related diseases has been extensively studied. In the microcirculation, SIRT1 plays a crucial role in preventing microcirculatory endothelial senescence by suppressing inflammation and oxidative stress while promoting mitochondrial function and optimizing autophagy. It suppresses hypoxia-inducible factor-1α (HIF-1α)-mediated pathological angiogenesis while promoting healthy, physiological capillarization. As a result, SIRT1 protects against microvascular dysfunction, such as diabetic microangiopathy, while enhancing exercise-induced skeletal muscle capillarization and energy metabolism. In the brain, SIRT1 upregulates tight junction proteins and strengthens their interactions, thus maintaining the integrity of the blood−brain barrier. The present review summarizes recent findings on the regulation of microvascular function by SIRT1, the underlying mechanisms, and various approaches to modulate SIRT1 activity in microcirculation. The importance of SIRT1 as a molecular target in aging-related diseases, such as diabetic retinopathy and stroke, is underscored, along with the need for more clinical evidence to support SIRT1 modulation in the microcirculation. Full article
Show Figures

Figure 1

Figure 1
<p>Endothelial silent information regulator 1 (SIRT1) exerts microcirculatory vasoprotective effects by upregulating antioxidant proteins, mitochondrial function, mitophagy, microcirculatory nitric oxide (NO) bioavailability, cholesterol export, and tight junction protein expression while suppressing inflammation and endothelial glycocalyx shedding, alongside optimizing autophagic flux.</p>
Full article ">Figure 2
<p>SIRT1 suppresses HIF-1α activity during hypoxia to prevent pathological angiogenesis. (<b>a</b>) Under normoxic conditions, hypoxia-inducible factor-1a (HIF-1α) is degraded via the ubiquitin-proteasome pathway, while Notch signaling is inhibited by SIRT1 via promoting Notch sensitivity to Jagged1 and deacetylating the Notch intracellular domain, which promotes healthy angiogenesis. (<b>b</b>) Under hypoxic conditions, acetylated HIF-1α leads to complete recruitment of p300 to the HIF-1α-HIF-1β complex, resulting in a high activity state on the hypoxia response element (HRE) and drastic upregulation of vascular endothelial growth factor (VEGF) and VEGF receptors (VEGFR) to induce pathogenic angiogenesis. (<b>c</b>) SIRT1 upregulation during hypoxia deacetylates HIF-1a, blocking the recruitment of p300 and reducing its activity on the HRE. In addition, SIRT1 promotes healthy angiogenesis by inhibiting Notch signaling.</p>
Full article ">
18 pages, 1393 KiB  
Article
Manipulation of Lipid Nanocapsules as an Efficient Intranasal Platform for Brain Deposition of Clozapine as an Antipsychotic Drug
by Ahmed A. Katamesh, Hend Mohamed Abdel-Bar, Mohammed Khaled Bin Break, Shimaa M. Hassoun, Gehad Subaiea, Amr Radwan and Hadel A. Abo El-Enin
Pharmaceutics 2024, 16(11), 1417; https://doi.org/10.3390/pharmaceutics16111417 - 5 Nov 2024
Viewed by 510
Abstract
Background/objectives: The blood–brain barrier (BBB) significantly limits the treatment of central nervous system disorders, such as schizophrenia, by restricting drug delivery to the brain. This study explores the potential of intranasal clozapine-loaded lipid nanocapsules (IN LNCsClo) as a targeted and effective [...] Read more.
Background/objectives: The blood–brain barrier (BBB) significantly limits the treatment of central nervous system disorders, such as schizophrenia, by restricting drug delivery to the brain. This study explores the potential of intranasal clozapine-loaded lipid nanocapsules (IN LNCsClo) as a targeted and effective delivery system to the brain. Methods: LNCsClo were prepared using the phase inversion technique and characterized in terms of size, zeta potential, entrapment efficiency (EE%), and in vitro drug release. The pharmacokinetic, safety, and pharmacodynamic effects of LNCsClo were then evaluated in a rat model through intranasal (IN) administration and compared with those of oral and intravenous (IV) Clo solutions. Results: LNCsClo were prepared using a phase inversion technique, resulting in a nanocarrier with a particle size of 28.6 ± 3.6 nm, homogenous dispersion, and high EE% (84.66 ± 5.66%). Pharmacokinetic analysis demonstrated that IN LNCsClo provided enhanced Clo brain bioavailability, rapid CNS targeting, and prolonged drug retention compared to oral and intravenous routes. Notably, the area under the curve (AUC) for brain concentration showed more than two-fold and eight-fold increases with LNCsClo, compared to IV and oral solutions, respectively, indicating improved brain-targeting efficiency. Safety assessments indicated that LNCsClo administration mitigated Clo-associated metabolic side effects, such as hyperglycemia, insulin imbalance, and liver enzyme alterations. Additionally, pharmacodynamic studies showed that LNCsClo significantly improved antipsychotic efficacy and reduced schizophrenia-induced hyperactivity, while preserving motor function. Conclusions: These results highlight the potential of IN LNCsClo as a novel drug delivery system, offering improved therapeutic efficacy, reduced systemic side effects, and better patient compliance in the treatment of schizophrenia and potentially other CNS disorders. Full article
(This article belongs to the Section Nanomedicine and Nanotechnology)
Show Figures

Figure 1

Figure 1
<p>In vitro characterization of the LNCs<sub>Clo</sub>. Morphological characterization of the prepared LNCs<sub>Clo</sub> using transmission electron micrography (<b>A</b>). The LNCs<sub>Clo</sub> appeared as a spherical nonaggregate nanostructure. In vitro release profile of Clo from the LNCs<sub>Clo</sub> in simulated nasal fluids at 35 °C (<b>B</b>). Drug release from the LNCs is measured by dialyzing the LNCs<sub>Clo</sub> against simulated nasal fluids (pH 7.4). Drug concentration in the dialysate is assessed by HPLC. Datapoints represent mean and SD (<span class="html-italic">n</span> = 3). (<b>C</b>) Effects of storage at 4 °C on the LNCs<sub>Clo</sub> particle size, zeta potential, and EE%. One-way analysis of variance (ANOVA) was used to compare the different parameters between groups, followed by the Tukey HSD test; ns is nonsignificant.</p>
Full article ">Figure 2
<p>Clozapine concentrations in rat (<b>A</b>) plasma and (<b>B</b>) brain tissues, after administration of various formulations. Animals received a dose of 2.5 mg/kg of Clo either via IN LNCs<sub>Clo,</sub> IV solution, or oral solution. At each time point, 6 animals were sacrificed from each group, and the concentration of Clo in plasma and brain tissues was quantified using HPLC. A significantly higher brain Clo concentration was observed at all time points following IN administration of LNCs<sub>Clo</sub> compared to IV and oral solutions (<span class="html-italic">p</span> &lt; 0.05). Datapoints represent mean ± SE (<span class="html-italic">n</span> = 6). The ANOVA test, followed by the Tukey HSD test, was used to compare the different parameters between groups, where <span class="html-italic">p</span> &lt; 0.05 was considered significant.</p>
Full article ">Figure 3
<p>Effect of clozapine on plasma glucose level (<b>A</b>) and insulin levels (<b>B</b>). Animals received a dose of 2.5 mg/kg/day of Clo either via IN LNCs<sub>Clo</sub> or an oral solution for 8 weeks. Every week, the fasting plasma glucose and insulin levels were measured. A significantly higher plasma glucose concentration and lower plasma insulin were observed at all time points following oral administration of Clo compared to the IN LNCs<sub>Clo</sub> (<span class="html-italic">p</span> &lt; 0.05). Datapoints represent mean ± SE (<span class="html-italic">n</span> = 10). The ANOVA test, followed by the Tukey HSD test, was used to compare the different parameters between groups, where <span class="html-italic">p</span> &lt; 0.05 was considered significant.</p>
Full article ">Figure 4
<p>Effect of clozapine on serum (<b>A</b>) ALT, (<b>B</b>) AST and (<b>C</b>) triglyceride levels. Animals received a dose of 2.5 mg/kg/day of Clo either via IN LNCs<sub>Clo</sub> or an oral solution for 8 weeks. Every week, fasting serum ALT, AST, and triglyceride levels were measured. Significantly higher serum ALT, AST, and triglyceride levels were observed at all time points following oral administration of Clo compared to IN LNCs<sub>Clo</sub> (<span class="html-italic">p</span> &lt; 0.05). Datapoints represent mean ± SE (<span class="html-italic">n</span> = 10). The ANOVA test, followed by the Tukey HSD test, was used to compare the different parameters between groups, where <span class="html-italic">p</span>&lt; 0.05 was considered significant.</p>
Full article ">Figure 5
<p>Assessment of the pharmacodynamics effects of intranasal LNCs<sub>Clo</sub>, IV Clo solution, and oral Clo solution using the (<b>A</b>) paw test and (<b>B</b>) ketamine-induced schizophrenia in rats via an open field test. A significantly higher anti-schizophrenic effect was observed following IN administration of LNCs<sub>Clo</sub> compared to IV and oral Clo solutions. Datapoints represent mean ± SE (<span class="html-italic">n</span> = 10). One-way analysis of variance (ANOVA) was used to compare the different parameters between groups, followed by the Tukey HSD test, * <span class="html-italic">p</span> &lt; 0.05, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
22 pages, 7402 KiB  
Article
Development of Nanocomposite Microspheres for Nasal Administration of Deferiprone in Neurodegenerative Disorders
by Radka Boyuklieva, Plamen Katsarov, Plamen Zagorchev, Silviya Abarova, Asya Hristozova and Bissera Pilicheva
J. Funct. Biomater. 2024, 15(11), 329; https://doi.org/10.3390/jfb15110329 - 5 Nov 2024
Viewed by 644
Abstract
Elevated brain iron levels are characteristic of many neurodegenerative diseases. As an iron chelator with short biological half-life, deferiprone leads to agranulocytosis and neutropenia with a prolonged therapeutic course. Its inclusion in sustained-release dosage forms may reduce the frequency of administration. On the [...] Read more.
Elevated brain iron levels are characteristic of many neurodegenerative diseases. As an iron chelator with short biological half-life, deferiprone leads to agranulocytosis and neutropenia with a prolonged therapeutic course. Its inclusion in sustained-release dosage forms may reduce the frequency of administration. On the other hand, when administered by an alternative route of administration, such as the nasal route, systemic exposure to deferiprone will be reduced, thereby reducing the occurrence of adverse effects. Direct nose-to-brain delivery has been raised as a non-invasive strategy to deliver drugs to the brain, bypassing the blood–brain barrier. The aim of the study was to develop and characterize nanocomposite microspheres suitable for intranasal administration by combining nano- and microparticle-based approaches. Nanoparticles with an average particle size of 213 ± 56 nm based on the biodegradable polymer poly-ε-caprolactone were developed using the solvent evaporation method. To ensure the deposition of the particles in the nasal cavity and avoid exhalation or deposition into the small airways, the nanoparticles were incorporated into composite structures of sodium alginate obtained by spray drying. Deferiprone demonstrated sustained release from the nanocomposite microspheres and high iron-chelating activity. Full article
(This article belongs to the Special Issue Medical Application of Functional Biomaterials (2nd Edition))
Show Figures

Figure 1

Figure 1
<p>Chemical structure of DFP (<b>A</b>) and iron ion binding sites (<b>B</b>).</p>
Full article ">Figure 2
<p>Schematic illustration of nanoparticle preparation by double emulsion technique (figure created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>).</p>
Full article ">Figure 3
<p>Schematic illustration of the experimental set-up for the study of mucoadhesive ability.</p>
Full article ">Figure 4
<p>DLS histograms of nanoparticles from batches NP1-DFP (<b>A</b>) and NP2-DFP (<b>B</b>).</p>
Full article ">Figure 5
<p>SEM micrograph of DFP-loaded nanoparticles from model NP1-DFP (20,000×).</p>
Full article ">Figure 6
<p>TEM micrographs of nanoparticles loaded with DFP from model NP1-DFP (28,000×).</p>
Full article ">Figure 7
<p>DFP release profiles from batches NP1-DFP and NP2-DFP.</p>
Full article ">Figure 8
<p>SEM micrograph of nanocomposite microspheres from batch M-NP1-DFP (3500×).</p>
Full article ">Figure 9
<p>Particle size distribution of batch M-NP1-DFP.</p>
Full article ">Figure 10
<p>Release profiles of DFP from nanoparticles (NP1-DFP) and composite microspheres (M-NP1-DFP).</p>
Full article ">Figure 11
<p>FTIR spectra of DFP, PCL, sodium alginate, and nanocomposite microparticles of model M-NP1-DFP.</p>
Full article ">Figure 12
<p>DSC thermograms of sodium alginate (<b>A</b>), PCL (<b>B</b>), DFP (<b>C</b>), and particles from batches M-NP1-DFP (<b>D</b>) and M-NP-Placebo (<b>E</b>).</p>
Full article ">Figure 13
<p>Chronological change of the maximal adhesion force in batch M-NP1-DFP. The symbols *, #, indicate the presence of a statistically significant difference compared to the initial moment (*) and after the established steady state of the process 15 min (#).</p>
Full article ">Figure 14
<p>Comparison of Fe-chelating activity of DFP solution, nanocomposite microspheres without the drug (M-NP-Placebo), and DFP-loaded nanocomposite microspheres (M-NP1-DFP). Data is presented as mean values ± SD, <span class="html-italic">n</span> = 3. * Indicates a statistically significant result (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 15
<p>Fluorescence spectra of HSA (2.7 mg/mL) upon addition of different volumes of samples containing dispersed particles from batch M-NP1-DFP and placebo particles at 37 °C.</p>
Full article ">
Back to TopTop