Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 9.584
1.
Fluids Barriers CNS ; 21(1): 39, 2024 May 06.
Article En | MEDLINE | ID: mdl-38711118

BACKGROUND: Triptans are anti-migraine drugs with a potential central site of action. However, it is not known to what extent triptans cross the blood-brain barrier (BBB). The aim of this study was therefore to determine if triptans pass the brain capillary endothelium and investigate the possible underlying mechanisms with focus on the involvement of the putative proton-coupled organic cation (H+/OC) antiporter. Additionally, we evaluated whether triptans interacted with the efflux transporter, P-glycoprotein (P-gp). METHODS: We investigated the cellular uptake characteristics of the prototypical H+/OC antiporter substrates, pyrilamine and oxycodone, and seven different triptans in the human brain microvascular endothelial cell line, hCMEC/D3. Triptan interactions with P-gp were studied using the IPEC-J2 MDR1 cell line. Lastly, in vivo neuropharmacokinetic assessment of the unbound brain-to-plasma disposition of eletriptan was conducted in wild type and mdr1a/1b knockout mice. RESULTS: We demonstrated that most triptans were able to inhibit uptake of the H+/OC antiporter substrate, pyrilamine, with eletriptan emerging as the strongest inhibitor. Eletriptan, almotriptan, and sumatriptan exhibited a pH-dependent uptake into hCMEC/D3 cells. Eletriptan demonstrated saturable uptake kinetics with an apparent Km of 89 ± 38 µM and a Jmax of 2.2 ± 0.7 nmol·min-1·mg protein-1 (n = 3). Bidirectional transport experiments across IPEC-J2 MDR1 monolayers showed that eletriptan is transported by P-gp, thus indicating that eletriptan is both a substrate of the H+/OC antiporter and P-gp. This was further confirmed in vivo, where the unbound brain-to-unbound plasma concentration ratio (Kp,uu) was 0.04 in wild type mice while the ratio rose to 1.32 in mdr1a/1b knockout mice. CONCLUSIONS: We have demonstrated that the triptan family of compounds possesses affinity for the H+/OC antiporter proposing that the putative H+/OC antiporter plays a role in the BBB transport of triptans, particularly eletriptan. Our in vivo studies indicate that eletriptan is subjected to simultaneous brain uptake and efflux, possibly facilitated by the putative H+/OC antiporter and P-gp, respectively. Our findings offer novel insights into the potential central site of action involved in migraine treatment with triptans and highlight the significance of potential transporter related drug-drug interactions.


Blood-Brain Barrier , Brain , Endothelial Cells , Mice, Knockout , Pyrrolidines , Tryptamines , Tryptamines/pharmacology , Tryptamines/metabolism , Tryptamines/pharmacokinetics , Animals , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Humans , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Brain/metabolism , Cell Line , Mice , Mice, Inbred C57BL , Biological Transport/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Male , Antiporters/metabolism , Pyrilamine/metabolism , Pyrilamine/pharmacology , ATP Binding Cassette Transporter, Subfamily B/metabolism
3.
Sci Rep ; 14(1): 8908, 2024 04 17.
Article En | MEDLINE | ID: mdl-38632344

Diseases related to the central nervous system (CNS) are major health concerns and have serious social and economic impacts. Developing new drugs for CNS-related disorders presents a major challenge as it actively involves delivering drugs into the CNS. Therefore, it is imperative to develop in silico methodologies to reliably identify potential lead compounds that can penetrate the blood-brain barrier (BBB) and help to thoroughly understand the role of different physicochemical properties fundamental to the BBB permeation of molecules. In this study, we have analysed the chemical space of the CNS drugs and compared it to the non-CNS-approved drugs. Additionally, we have collected a feature selection dataset from Muehlbacher et al. (J Comput Aided Mol Des 25(12):1095-1106, 2011. 10.1007/s10822-011-9478-1) and an in-house dataset. This information was utilised to design a molecular fingerprint that was used to train machine learning (ML) models. The best-performing models reported in this study achieved accuracies of 0.997 and 0.98, sensitivities of 1.0 and 0.992, specificities of 0.971 and 0.962, MCCs of 0.984 and 0.958, and ROC-AUCs of 0.997 and 0.999 on an imbalanced and a balanced dataset, respectively. They demonstrated overall good accuracies and sensitivities in the blind validation dataset. The reported models can be applied for fast and early screening drug-like molecules with BBB potential. Furthermore, the bbbPythoN package can be used by the research community to both produce the BBB-specific molecular fingerprints and employ the models mentioned earlier for BBB-permeability prediction.


Blood-Brain Barrier , Central Nervous System , Biological Transport/physiology , Machine Learning , Permeability
4.
J Math Biol ; 88(6): 69, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38664246

Flow in a porous medium can be driven by the deformations of the boundaries of the porous domain. Such boundary deformations locally change the volume fraction accessible by the fluid, creating non-uniform porosity and permeability throughout the medium. In this work, we construct a deformation-driven porous medium transport model with spatially and temporally varying porosity and permeability that are dependent on the boundary deformations imposed on the medium. We use this model to study the transport of interstitial fluid along the basement membranes in the arterial walls of the brain. The basement membrane is modeled as a deforming annular porous channel with the compressible pore space filled with an incompressible, Newtonian fluid. The role of a forward propagating peristaltic heart pulse wave and a reverse smooth muscle contraction wave on the flow within the basement membranes is investigated. Our results identify combinations of wave amplitudes that can induce either forward or reverse transport along these transport pathways in the brain. The magnitude and direction of fluid transport predicted by our model can help in understanding the clearance of fluids and solutes along the Intramural Periarterial Drainage route and the pathology of cerebral amyloid angiopathy.


Brain , Extracellular Fluid , Extracellular Fluid/metabolism , Extracellular Fluid/physiology , Porosity , Humans , Brain/metabolism , Brain/blood supply , Brain/physiology , Basement Membrane/metabolism , Basement Membrane/physiology , Mathematical Concepts , Biological Transport/physiology , Models, Biological , Computer Simulation , Models, Neurological , Animals , Permeability
5.
Mol Pharm ; 21(5): 2473-2483, 2024 May 06.
Article En | MEDLINE | ID: mdl-38579335

In recent years, the drainage of fluids, immune cells, antigens, fluorescent tracers, and other solutes from the brain has been demonstrated to occur along lymphatic outflow pathways to the deep cervical lymph nodes in the neck. To the best of our knowledge, no studies have evaluated the lymphatic transport of therapeutics from the brain. The objective of this study was to determine the lymphatic transport of model therapeutics of different molecular weights and lipophilicity from the brain using cervical lymph cannulation and ligation models in rats. To do this, anesthetized Sprague-Dawley rats were cannulated at the carotid artery and cannulated, ligated, or left intact at the cervical lymph duct. Rats were administered 14C-ibuprofen (206.29 g/mol, logP 3.84), 3H-halofantrine HCl (536.89 g/mol, logP 8.06), or 3H-albumin (∼65,000 g/mol) via direct injection into the brain striatum at a rate of 0.5 µL/min over 16 min. Plasma or cervical lymph samples were collected for up to 6-8 h following dosing, and brain and lymph nodes were collected at 6 or 8 h. Samples were subsequently analyzed for radioactivity levels via scintillation counting. For 14C-ibuprofen, plasma concentrations over time (plasma AUC0-6h) were >2 fold higher in lymph-ligated rats than in lymph-intact rats, suggesting that ibuprofen is cleared from the brain primarily via nonlymphatic routes (e.g., across the blood-brain barrier) but that this clearance is influenced by changes in lymphatic flow. For 3H-halofantrine, >73% of the dose was retained at the brain dosing site in lymph-intact and lymph-ligated groups, and plasma AUC0-8h values were low in both groups (<0.3% dose.h/mL), consistent with the high retention in the brain. It was therefore not possible to determine whether halofantrine undergoes lymphatic transport from the brain within the duration of the study. For 3H-albumin, plasma AUC0-8h values were not significantly different between lymph-intact, lymph-ligated, and lymph-cannulated rats. However, >4% of the dose was recovered in cervical lymph over 8 h. Lymph/plasma concentration ratios of 3H-albumin were also very high (up to 53:1). Together, these results indicate that 3H-albumin is transported from the brain not only via lymphatic routes but also via the blood. Similar to other tissues, the lymphatics may thus play a significant role in the transport of macromolecules, including therapeutic proteins, from the brain but are unlikely to be a major transport pathway from the brain for small molecule drugs that are not lipophilic. Our rat cervical lymph cannulation model can be used to quantify the lymphatic drainage of different molecules and factors from the brain.


Brain , Ibuprofen , Lymph Nodes , Rats, Sprague-Dawley , Animals , Rats , Brain/metabolism , Male , Lymph Nodes/metabolism , Ibuprofen/pharmacokinetics , Ibuprofen/administration & dosage , Ibuprofen/chemistry , Phenanthrenes/pharmacokinetics , Phenanthrenes/chemistry , Phenanthrenes/administration & dosage , Biological Transport/physiology , Albumins/pharmacokinetics , Albumins/metabolism
6.
Biosystems ; 239: 105210, 2024 May.
Article En | MEDLINE | ID: mdl-38599512

Most nutrient uptake problems are modeled by the convection-diffusion equation (CDE) abiding by Fick's law. Because nutrients needed by plants exist in the soil solution as a form of ions and the soil is a typical fractal structure of heterogeneity, it makes the solute transport appear anomalous diffusion in soil. Taking anomalous diffusion as a transport process, we propose time and space fractional nutrient uptake models based on the classic Nye-Tinker-Barber model. There does not appear apparent sub-diffusion of nitrate in the time fractional model until four months and the time fractional models are appropriate for describing long-term dynamics and slow sorption reaction; the space fractional model can capture super-diffusion in short term and it is suitable for describing nonlocal phenomena and daily variations driven by transpiration and metabolism; the anomalous diffusion more apparently appears near the root surface in the modeling simulation.


Models, Biological , Nutrients , Plant Roots , Plant Roots/metabolism , Diffusion , Nutrients/metabolism , Biological Transport/physiology , Soil/chemistry , Nitrates/metabolism , Computer Simulation
7.
Food Funct ; 15(7): 3496-3506, 2024 Apr 02.
Article En | MEDLINE | ID: mdl-38463011

A potential osteogenic tetradecapeptide with the amino acid sequence GETNPADSKPGSIR (P-GM-2) was identified from Gadus morhua. The present study aimed to elucidate its absorption and transport properties using Caco-2/HT29-MTX co-culture monolayers and to evaluate its osteogenic activity using an ovariectomized mouse model. The results showed that P-GM-2 could cross Caco-2/HT29-MTX co-culture barriers intactly with an apparent permeability coefficient of 4.02 × 10-6 cm s-1via the TJ-mediated passive paracellular pathway. Pharmacokinetic results revealed that P-GM-2 was detectable in the blood of mice within 5 min of oral administration and reached its maximum concentration at 30 min. Furthermore, the oral administration of P-GM-2 for a duration of three months has been found to effectively regulate the secretion of key markers of bone turnover, thereby protecting against bone microstructure degeneration and bone loss in ovariectomized mice. Importantly, no toxicity related to the treatment was observed. Taken together, these findings offer valuable insights into the absorption and transport mechanisms of P-GM-2, highlighting its potential as a safe and effective active ingredient for preventing osteoporosis.


Intestinal Absorption , Peptides , Humans , Mice , Animals , Caco-2 Cells , Intestinal Absorption/physiology , HT29 Cells , Permeability , Peptides/pharmacology , Peptides/metabolism , Biological Transport/physiology
8.
J Mater Chem B ; 12(8): 2123-2138, 2024 Feb 21.
Article En | MEDLINE | ID: mdl-38314923

Oxidative stress and blood-brain barrier (BBB) injury are two major stress disorders before and after ischemic stroke (IS) therapy. The intense inflammatory response also causes damage to nerve cells, affecting the repair of brain tissue. In this study, polyphenolic nanoparticles (PPNs) with strong free radical scavenging ability were designed to treat IS multimodally. To investigate the mechanism of polyphenolic polymerization, solid nanoparticles were synthesized using four kinds of polyphenol compounds as the basic unit under the control of temperature. The form of polymerization between monomers with different structures led to changes in the chemical properties of the corresponding nanoparticles as well as the antioxidant capacity at the cellular level. Particularly, PPNs can significantly improve cerebral infarction and penetrate and repair the BBB, and even downregulate levels of inflammatory cytokines. Molecular signaling pathway studies have shown that PPNs can provide comprehensive treatment of IS by promoting the expression of tight junction protein and enhancing the activity of antioxidant enzymes. Therefore, PPNs combined with the antioxidant, anti-inflammatory and BBB repair ability not only provide a perfect therapeutic pathway but also give ideas for the development of natural material carriers that have a wide application prospect.


Blood-Brain Barrier , Ischemic Stroke , Humans , Blood-Brain Barrier/metabolism , Reactive Oxygen Species/metabolism , Ischemic Stroke/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Antioxidants/metabolism , Biological Transport/physiology
9.
Mol Biol Cell ; 35(3): ar39, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38170584

DIFFRAC is a powerful method for systematically comparing proteome content and organization between samples in a high-throughput manner. By subjecting control and experimental protein extracts to native chromatography and quantifying the contents of each fraction using mass spectrometry, it enables the quantitative detection of alterations to protein complexes and abundances. Here, we applied DIFFRAC to investigate the consequences of genetic loss of Ift122, a subunit of the intraflagellar transport-A (IFT-A) protein complex that plays a vital role in the formation and function of cilia and flagella, on the proteome of Tetrahymena thermophila. A single DIFFRAC experiment was sufficient to detect changes in protein behavior that mirrored known effects of IFT-A loss and revealed new biology. We uncovered several novel IFT-A-regulated proteins, which we validated through live imaging in Xenopus multiciliated cells, shedding new light on both the ciliary and non-ciliary functions of IFT-A. Our findings underscore the robustness of DIFFRAC for revealing proteomic changes in response to genetic or biochemical perturbation.


Proteome , Proteomics , Protein Transport/physiology , Proteome/metabolism , Biological Transport/physiology , Cilia/metabolism , Flagella/metabolism , Phenotype
10.
Life Sci Alliance ; 7(3)2024 Mar.
Article En | MEDLINE | ID: mdl-38171596

The mouse cortical collecting duct cell line presents a tight epithelium with regulated ion and water transport. The epithelial sodium channel (ENaC) is localized in the apical membrane and constitutes the rate-limiting step for sodium entry, thereby enabling transepithelial transport of sodium ions. The membrane-bound serine protease Tmprss2 is co-expressed with the alpha subunit of ENaC. αENaC gene expression followed the Tmprss2 expression, and the absence of Tmprss2 resulted not only in down-regulation of αENaC gene and protein expression but also in abolished transepithelial sodium transport. In addition, RNA-sequencing analyses unveiled drastic down-regulation of the membrane-bound protease CAP3/St14, the epithelial adhesion molecule EpCAM, and the tight junction proteins claudin-7 and claudin-3 as also confirmed by immunohistochemistry. In summary, our data clearly demonstrate a dual role of Tmprss2 in maintaining not only ENaC-mediated transepithelial but also EpCAM/claudin-7-mediated paracellular barrier; the tight epithelium of the mouse renal mCCD cells becomes leaky. Our working model proposes that Tmprss2 acts via CAP3/St14 on EpCAM/claudin-7 tight junction complexes and through regulating transcription of αENaC on ENaC-mediated sodium transport.


Claudins , Sodium , Animals , Mice , Biological Transport/physiology , Claudins/genetics , Claudins/metabolism , Epithelial Cell Adhesion Molecule/metabolism , Ion Transport , Sodium/metabolism
11.
Biol Pharm Bull ; 47(1): 79-87, 2024.
Article En | MEDLINE | ID: mdl-38171781

Herein, we investigated whether a fluorescent probe for an organic anion transporter (OAT), fluorescein (FLS), could be accumulated by human kidney 2 (HK-2) cells derived from human kidney proximal tubular epithelia. HK-2 cells took up FLS in a pH-dependent and concentration-dependent manner. FLS accumulation by HK-2 cells was inhibited by monocarboxylic acids, ibuprofen, rosuvastatin, and indoleacetic acid but not by typical substrates for OATs. A typical protonophore, carbonyl cyanide p-trichloromethoxyphenylhydrazone completely abolished FLS accumulation by HK-2 cells. The FLS efflux process from the preloaded HK-2 cells exhibited substantial trans-stimulation by the excess amount of extracellular FLS transport inhibitable monocarboxylate compounds such as 2,4-dichloro phenoxyacetic acid, fluvastatin, ibuprofen, indoleacetic acid, salicylic acid and rosuvastatin, indicating that the FLS transporter can recognize and accumulate them into the cells in a pH-dependent manner. The involvement of the FLS transporter in the reabsorption of monocarboxylic compounds was indicated by demonstrating that the pH-dependent FLS uptake is inhibited by various monocarboxylates in rabbit renal brush border membrane vesicles. pH-dependent FLS uptake was trans-stimulated by the inhibitable monocarboxylates. Collectively, the present data indicate that the pH-dependent transporters expressed in HK-2 cells are involved in the reabsorption of monocarboxylates from the urinary fluid into the tubular epithelia.


Ibuprofen , Monocarboxylic Acid Transporters , Animals , Humans , Rabbits , Fluorescein/metabolism , Rosuvastatin Calcium/metabolism , Monocarboxylic Acid Transporters/metabolism , Kidney/metabolism , Biological Transport/physiology , Indoleacetic Acids , Hydrogen-Ion Concentration
12.
Expert Opin Drug Deliv ; 21(1): 71-89, 2024.
Article En | MEDLINE | ID: mdl-38217410

INTRODUCTION: Successful neuropharmacology requires optimization of CNS drug delivery and, by extension, free drug concentrations at brain molecular targets. Detailed assessment of blood-brain barrier (BBB) physiological characteristics is necessary to achieve this goal. The 'next frontier' in CNS drug delivery is targeting BBB uptake transporters, an approach that requires evaluation of brain endothelial cell transport processes so that effective drug accumulation and improved therapeutic efficacy can occur. AREAS COVERED: BBB permeability of drugs is governed by tight junction protein complexes (i.e., physical barrier) and transporters/enzymes (i.e., biochemical barrier). For most therapeutics, a component of blood-to-brain transport involves passive transcellular diffusion. Small molecule drugs that do not possess acceptable physicochemical characteristics for passive permeability may utilize putative membrane transporters for CNS uptake. While both uptake and efflux transport mechanisms are expressed at the brain microvascular endothelium, uptake transporters can be targeted for optimization of brain drug delivery and improved treatment of neurological disease states. EXPERT OPINION: Uptake transporters represent a unique opportunity to optimize brain drug delivery by leveraging the endogenous biology of the BBB. A rigorous understanding of these transporters is required to improve translation from the bench to clinical trials and stimulate the development of new treatment paradigms for neurological diseases.


Blood-Brain Barrier , Brain , Blood-Brain Barrier/metabolism , Brain/metabolism , Membrane Transport Proteins/metabolism , Biological Transport/physiology , Pharmaceutical Preparations/metabolism , Central Nervous System Agents
13.
J Nanobiotechnology ; 22(1): 43, 2024 Jan 30.
Article En | MEDLINE | ID: mdl-38287357

The central nervous system (CNS) maintains homeostasis with its surrounding environment by restricting the ingress of large hydrophilic molecules, immune cells, pathogens, and other external harmful substances to the brain. This function relies heavily on the blood-cerebrospinal fluid (B-CSF) and blood-brain barrier (BBB). Although considerable research has examined the structure and function of the BBB, the B-CSF barrier has received little attention. Therapies for disorders associated with the central nervous system have the potential to benefit from targeting the B-CSF barrier to enhance medication penetration into the brain. In this study, we synthesized a nanoprobe ANG-PEG-UCNP capable of crossing the B-CSF barrier with high targeting specificity using a hydrocephalus model for noninvasive magnetic resonance ventriculography to understand the mechanism by which the CSF barrier may be crossed and identify therapeutic targets of CNS diseases. This magnetic resonance nanoprobe ANG-PEG-UCNP holds promising potential as a safe and effective means for accurately defining the ventricular anatomy and correctly locating sites of CSF obstruction.


Blood-Brain Barrier , Brain , Brain/diagnostic imaging , Central Nervous System , Biological Transport/physiology , Magnetic Resonance Imaging
14.
CNS Neurol Disord Drug Targets ; 23(3): 315-330, 2024.
Article En | MEDLINE | ID: mdl-36999187

The blood-brain barrier (BBB) plays a crucial role in the central nervous system by tightly regulating the influx and efflux of biological substances between the brain parenchyma and peripheral circulation. Its restrictive nature acts as an obstacle to protect the brain from potentially noxious substances such as blood-borne toxins, immune cells, and pathogens. Thus, the maintenance of its structural and functional integrity is vital in the preservation of neuronal function and cellular homeostasis in the brain microenvironment. However, the barrier's foundation can become compromised during neurological or pathological conditions, which can result in dysregulated ionic homeostasis, impaired transport of nutrients, and accumulation of neurotoxins that eventually lead to irreversible neuronal loss. Initially, the BBB is thought to remain intact during neurodegenerative diseases, but accumulating evidence as of late has suggested the possible association of BBB dysfunction with Parkinson's disease (PD) pathology. The neurodegeneration occurring in PD is believed to stem from a myriad of pathogenic mechanisms, including tight junction alterations, abnormal angiogenesis, and dysfunctional BBB transporter mechanism, which ultimately causes altered BBB permeability. In this review, the major elements of the neurovascular unit (NVU) comprising the BBB are discussed, along with their role in the maintenance of barrier integrity and PD pathogenesis. We also elaborated on how the neuroendocrine system can influence the regulation of BBB function and PD pathogenesis. Several novel therapeutic approaches targeting the NVU components are explored to provide a fresh outlook on treatment options for PD.


Blood-Brain Barrier , Parkinson Disease , Humans , Blood-Brain Barrier/physiology , Parkinson Disease/pathology , Brain/pathology , Central Nervous System , Biological Transport/physiology
15.
Handb Exp Pharmacol ; 284: 213-230, 2024.
Article En | MEDLINE | ID: mdl-37528323

Blood-brain barrier (BBB) is a special biological property of the brain neurovascular unit (including brain microvessels and capillaries), which facilitates the transport of nutrients into the central nervous system (CNS) and exchanges metabolites but restricts passage of blood-borne neurotoxic substances and drugs/xenobiotics into CNS. BBB plays a crucial role in maintaining the homeostasis and normal physiological functions of CNS but severely impedes the delivery of drugs and biotherapeutics into CNS for treatment of neurological disorders. A variety of technologies have been developed in the past decade for brain drug delivery. Most of these technologies are still in preclinical stage and some are undergoing clinical studies. Only a few have been approved by regulatory agencies for clinical applications. This chapter will overview the strategies and technologies/approaches for brain drug delivery and discuss some of the recent advances in the field.


Blood-Brain Barrier , Brain , Humans , Blood-Brain Barrier/metabolism , Brain/metabolism , Biological Transport/physiology , Central Nervous System , Drug Delivery Systems , Pharmaceutical Preparations/metabolism
16.
J Cereb Blood Flow Metab ; 44(3): 446-448, 2024 03.
Article En | MEDLINE | ID: mdl-38000041

Blood-brain barrier dysfunction (BBB) occurs rapidly after stroke and contributes to edema, inflammation, and secondary brain injury including haemorrhage. Two recent studies shed light on the temporal extent of post-stroke BBB dysfunction as well as its consequences for drug delivery. Zhang et al. found increases in BBB permeability that persist up to one-year post-ischemia. Despite increased paracellular leakage, Stanton et al. showed that transcellular transporter systems are required to deliver therapeutics into brain parenchyma. Both studies remind us of the complexity of BBB responses after stroke and provide novel entry points for future research into the underlying mechanisms.


Brain Ischemia , Stroke , Humans , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Stroke/metabolism , Brain/metabolism , Biological Transport/physiology , Brain Ischemia/metabolism
17.
J Pharm Sci ; 113(5): 1209-1219, 2024 May.
Article En | MEDLINE | ID: mdl-37984697

To clarify the regulation of drug absorption by the enteric nervous system, we investigated how adrenergic agonists (adrenaline (ADR), clonidine (CLO), dobutamine (DOB)) and dibutyryl cAMP (DBcAMP) affected P-glycoprotein (P-gp) function by utilizing isolated rat jejunal sheets and Caco-2 cell monolayers. ADR and CLO significantly decreased the secretory transport (Papptotal) of rhodamine-123 and tended to decrease the transport via P-gp (PappP-gp) and passive transport (Papppassive). In contrast, DBcAMP significantly increased and DOB tended to increase Papptotal and both tended to increase PappP-gpand Papppassive. Changes in P-gp expression on brush border membrane by adrenergic agonists and DBcAMP were significantly correlated with PappP-gp, while P-gp expression was not changed in whole cell homogenates, suggesting that the trafficking of P-gp would be responsible for its functional changes. Papppassive was inversely correlated with transmucosal or transepithelial electrical resistance, indicating that adrenergic agonists affected the paracellular permeability. Adrenergic agonists also changed cAMP levels, which were significantly correlated with PappP-gp. Furthermore, protein kinase A (PKA) or PKC inhibitor significantly decreased PappP-gp in Caco-2 cell monolayers, suggesting that they would partly contribute to the changes in P-gp activity. In conclusion, adrenergic agonists regulated P-gp function and paracellular permeability, which would be caused via adrenoceptor stimulation.


ATP Binding Cassette Transporter, Subfamily B, Member 1 , Adrenergic Agonists , Humans , Rats , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Caco-2 Cells , Bucladesine/metabolism , Biological Transport/physiology , ATP Binding Cassette Transporter, Subfamily B/metabolism , Epinephrine , Intestinal Absorption
18.
Protein Sci ; 33(2): e4879, 2024 Feb.
Article En | MEDLINE | ID: mdl-38131105

Lipopolysaccharide (LPS) synthesis in Gram-negative bacteria is completed at the outer leaflet of the inner membrane (IM). Following synthesis, seven LPS transport (Lpt) proteins facilitate the movement of LPS to the outer membrane (OM), an essential process that if disrupted at any stage has lethal effects on bacterial viability. LptB2 FG, the IM component of the Lpt bridge system, is a type VI ABC transporter that provides the driving force for LPS extraction from the IM and subsequent transport across a stable protein bridge to the outer leaflet of the OM. LptC is a periplasmic protein anchored to the IM by a single transmembrane (TM) helix intercalating within the lateral gate formed by LptF TM5 and LptG TM1. LptC facilitates the hand-off of LPS from LptB2 FG to the periplasmic protein LptA and has been shown to regulate the ATPase activity of LptB2 FG. Here, using an engineered chromosomal knockout system in Escherichia coli to assess the effects of LptC mutations in vivo, we identified six partial loss of function LptC mutations in the first unbiased alanine screen of this essential protein. To investigate the functional effects of these mutations, nanoDSF (differential scanning fluorimetry) and site-directed spin labeling (SDSL) electron paramagnetic resonance (EPR) spectroscopy in combination with an in vitro ATPase assay show that specific residues in the TM helix of LptC destabilize the LptB2 FGC complex and regulate the ATPase activity of LptB.


Escherichia coli Proteins , Periplasmic Proteins , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Lipopolysaccharides/metabolism , Membrane Proteins/chemistry , Periplasmic Proteins/metabolism , Biological Transport/physiology , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/chemistry , ATP-Binding Cassette Transporters/metabolism
19.
Dis Model Mech ; 16(11)2023 Nov 01.
Article En | MEDLINE | ID: mdl-38037877

By controlling the passage of small molecules across lipid bilayers, membrane transporters influence not only the uptake and efflux of nutrients, but also the metabolic state of the cell. With more than 450 members, the Solute Carriers (SLCs) are the largest transporter super-family, clustering into families with different substrate specificities and regulatory properties. Cells of different types are, therefore, able to tailor their transporter expression signatures depending on their metabolic requirements, and the physiological importance of these proteins is illustrated by their mis-regulation in a number of disease states. In cancer, transporter expression is heterogeneous, and the SLC family has been shown to facilitate the accumulation of biomass, influence redox homeostasis, and also mediate metabolic crosstalk with other cell types within the tumour microenvironment. This Review explores the roles of membrane transporters in physiological and malignant settings, and how these roles can affect drug response, through either indirect modulation of sensitivity or the direct transport of small-molecule therapeutic compounds into cells.


Membrane Transport Proteins , Neoplasms , Humans , Membrane Transport Proteins/metabolism , Solute Carrier Proteins/chemistry , Solute Carrier Proteins/metabolism , Biological Transport/physiology , Neoplasms/drug therapy , Cell Physiological Phenomena , Tumor Microenvironment
20.
Int J Mol Sci ; 24(22)2023 Nov 14.
Article En | MEDLINE | ID: mdl-38003477

The blood-brain barrier (BBB) is a unique and selective feature of the central nervous system's vasculature. BBB dysfunction has been observed as an early sign of Alzheimer's Disease (AD) before the onset of dementia or neurodegeneration. The intricate relationship between the BBB and the pathogenesis of AD, especially in the context of neurovascular coupling and the overlap of pathophysiology in neurodegenerative and cerebrovascular diseases, underscores the urgency to understand the BBB's role more deeply. Preserving or restoring the BBB function emerges as a potentially promising strategy for mitigating the progression and severity of AD. Molecular and genetic changes, such as the isoform ε4 of apolipoprotein E (ApoEε4), a significant genetic risk factor and a promoter of the BBB dysfunction, have been shown to mediate the BBB disruption. Additionally, receptors and transporters like the low-density lipoprotein receptor-related protein 1 (LRP1), P-glycoprotein (P-gp), and the receptor for advanced glycation end products (RAGEs) have been implicated in AD's pathogenesis. In this comprehensive review, we endeavor to shed light on the intricate pathogenic and therapeutic connections between AD and the BBB. We also delve into the latest developments and pioneering strategies targeting the BBB for therapeutic interventions, addressing its potential as a barrier and a carrier. By providing an integrative perspective, we anticipate paving the way for future research and treatments focused on exploiting the BBB's role in AD pathogenesis and therapy.


Alzheimer Disease , Cerebrovascular Disorders , Humans , Alzheimer Disease/metabolism , Blood-Brain Barrier/metabolism , Amyloid beta-Peptides/metabolism , Biological Transport/physiology , Cerebrovascular Disorders/metabolism
...