Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Exp Neurol ; 377: 114782, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38641126

RESUMEN

Elevated transport of Caveolin-1 (CAV-1) vesicles within vascular endothelial cells constitutes a significant secondary pathogenic event contributing to the compromise of the blood-brain barrier (BBB) post-traumatic brain injury (TBI). While Wnt/ß-catenin signaling is recognized for its critical involvement in angiogenesis and the maintenance of BBB integrity, its influence on vascular endothelial transcytosis in the aftermath of TBI is not well-defined. This study aims to elucidate the impact of Wnt/ß-catenin signaling on cerebrovascular vesicular transcytosis following TBI. In this experiment, adult male wild-type (WT) C57BL/6 mice underwent various interventions. TBI was induced utilizing the controlled cortical impact technique. Post-TBI, mice were administered either an inhibitor or an agonist of Wnt signaling via intraperitoneal injection. Recombinant adeno-associated virus (rAAV) was administered intracerebroventricularly to modulate the expression of the CAV-1 inhibitory protein, Major facilitator superfamily domain-containing 2a (Mfsd2a). This research utilized Evans blue assay, Western blot analysis, immunofluorescence, transmission electron microscopy, and neurobehavioral assessments. Post-TBI observations revealed substantial increases in macromolecule (Evans blue and albumin) leakage, CAV-1 transport vesicle count, astrocyte end-feet edema, and augmented aquaporin-4 (AQP4) expression, culminating in BBB disruption. The findings indicate that Wnt signaling pathway inhibition escalates CAV-1 transport vesicle activity and aggravates BBB compromise. Conversely, activating this pathway could alleviate BBB damage by curtailing CAV-1 vesicle presence. Post-TBI, there is a diminution in Mfsd2a expression, which is directly influenced by the modulation of WNT signals. Employing a viral approach to regulate Mfsd2a, we established that its down-regulation undermines the protective benefits derived from reducing CAV-1 transport vesicles through WNT signal enhancement. Moreover, we verified that the WNT signaling agonist LiCl notably ameliorates neurological deficits following TBI in mice. Collectively, our data imply that Wnt/ß-catenin signaling presents a potential therapeutic target for safeguarding against BBB damage and enhancing neurological function after TBI.


Asunto(s)
Barrera Hematoencefálica , Lesiones Traumáticas del Encéfalo , Caveolina 1 , Ratones Endogámicos C57BL , Transcitosis , Vía de Señalización Wnt , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Ratones , Masculino , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/fisiología , Transcitosis/efectos de los fármacos , Transcitosis/fisiología , Caveolina 1/metabolismo , Simportadores
2.
Adv Sci (Weinh) ; 9(21): e2201414, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35652273

RESUMEN

Bile acid-modified nanoparticles provide a convenient strategy to improve oral bioavailability of poorly permeable drugs by exploiting specific interactions with bile acid transporters. However, the underlying mechanisms are unknown, especially considering the different absorption sites of free bile acids (ileum) and digested fat molecules from bile acid-emulsified fat droplets (duodenum). Here, glycocholic acid (GCA)-conjugated polystyrene nanoparticles (GCPNs) are synthesized and their transport in Caco-2 cell models is studied. GCA conjugation enhances the uptake by interactions with apical sodium-dependent bile acid transporter (ASBT). A new pathway correlated with both ASBT and chylomicron pathways is identified. Meanwhile, the higher uptake of GCPNs does not lead to higher transcytosis to the same degree compared with unmodified nanoparticles (CPNs). The pharmacological and genomics study confirm that GCA conjugation changes the endocytosis mechanisms and downregulates the cellular response to the transport at gene levels, which works as a negative feedback loop and explains the higher cellular retention of GCPNs. These findings offer a solid foundation in the bile acid-based nanomedicine design, with utilizing advantages of the ASBT-mediated uptake, as well as inspiration to take comprehensive consideration of the cellular response with more developed technologies.


Asunto(s)
Ácidos y Sales Biliares , Quilomicrones , Nanopartículas , Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/farmacología , Células CACO-2 , Quilomicrones/efectos de los fármacos , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Humanos , Nanopartículas/química , Transportadores de Anión Orgánico Sodio-Dependiente/farmacología , Transducción de Señal/efectos de los fármacos , Simportadores/farmacología , Transcitosis/efectos de los fármacos , Transcitosis/fisiología
3.
Front Immunol ; 12: 636966, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34557180

RESUMEN

Since 2003, the world has been confronted with three new betacoronaviruses that cause human respiratory infections: SARS-CoV, which causes severe acute respiratory syndrome (SARS), MERS-CoV, which causes Middle East respiratory syndrome (MERS), and SARS-CoV-2, which causes Coronavirus Disease 2019 (COVID-19). The mechanisms of coronavirus transmission and dissemination in the human body determine the diagnostic and therapeutic strategies. An important problem is the possibility that viral particles overcome tissue barriers such as the intestine, respiratory tract, blood-brain barrier, and placenta. In this work, we will 1) consider the issue of endocytosis and the possibility of transcytosis and paracellular trafficking of coronaviruses across tissue barriers with an emphasis on the intestinal epithelium; 2) discuss the possibility of antibody-mediated transcytosis of opsonized viruses due to complexes of immunoglobulins with their receptors; 3) assess the possibility of the virus transfer into extracellular vesicles during intracellular transport; and 4) describe the clinical significance of these processes. Models of the intestinal epithelium and other barrier tissues for in vitro transcytosis studies will also be briefly characterized.


Asunto(s)
Endocitosis , Mucosa Intestinal/virología , SARS-CoV-2/metabolismo , Anticuerpos Antivirales/metabolismo , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19/transmisión , COVID-19/virología , Ensayos Clínicos como Asunto , Endocitosis/efectos de los fármacos , Humanos , Mucosa Intestinal/metabolismo , Modelos Biológicos , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , Uniones Estrechas/metabolismo , Uniones Estrechas/virología , Transcitosis/efectos de los fármacos , Acoplamiento Viral , Tratamiento Farmacológico de COVID-19
4.
J Med Chem ; 64(18): 13152-13173, 2021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34505508

RESUMEN

In the development of central nervous system (CNS) drugs, the blood-brain barrier (BBB) restricts many drugs from entering the brain to exert therapeutic effects. Although many novel delivery methods of large molecule drugs have been designed to assist transport, small molecule drugs account for the vast majority of the CNS drugs used clinically. From this perspective, we review studies from the past five years that have sought to modify small molecules to increase brain exposure. Medicinal chemists make it easier for small molecules to cross the BBB by improving diffusion, reducing efflux, and activating carrier transporters. On the basis of their excellent work, we summarize strategies for structural modification of small molecules to improve BBB penetration. These strategies are expected to provide a reference for the future development of small molecule CNS drugs.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Fármacos del Sistema Nervioso Central/metabolismo , Animales , Fármacos del Sistema Nervioso Central/química , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Estructura Molecular , Permeabilidad , Transcitosis/efectos de los fármacos
5.
ACS Chem Biol ; 16(1): 106-115, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33315366

RESUMEN

The cell membrane of brain endothelial cells is enriched in omega-3 phospholipid species. Numerous omega-3 phospholipid species were recently proposed to be important for maintaining the low rate of transcytosis and, thus, could be important for regulating one of the mechanisms of the blood brain barrier (BBB). However, the spatial distribution of these phospholipid species within the brain was previously unknown. Here, we combined advanced mass spectrometry imaging techniques to generate a map of these phospholipids in the brain at near single cell resolution. Furthermore, we explored the effects of omega-3 dietary deprivation on both docosahexaenoic acid (DHA)-containing phospholipids and the global brain phospholipid profile. We demonstrate the unique spatial distribution of individual DHA-containing phospholipids, which may be important for the regiospecific properties of the BBB. Finally, 24 diet discriminative phospholipids were identified and showed an increase in saturated phospholipid species and ceramide containing phospholipid species under omega-3 dietary deficiency.


Asunto(s)
Ácidos Grasos Omega-3/metabolismo , Fosfolípidos/farmacología , Transcitosis/efectos de los fármacos , Animales , Barrera Hematoencefálica , Femenino , Masculino , Ratones , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos
6.
Burns ; 47(4): 894-905, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33143990

RESUMEN

BACKGROUND: Hypertrophic scar (HS) formation, a type of dermal fibroproliferative condition, is a frequent complication in wound healing resulting from burns, severe trauma, and surgical procedures. The effects of Panax Notoginseng Saponins (PNS) on the HS formation remain relatively under-explored. Hence, this study was intended to interrogate anti-apoptosis and anti-fibrosis effects of PNS on the hypertrophic scar fibroblasts (HSFs) during HS formation and assess the involvement of TRPM7 and PI3K/AKT signaling pathway. METHODS: Using MTT and CCK-8 assays, we evaluated cell cytotoxicity and cell viability. Collagen I/III (col 1/3) and α-SMA expression levels were assessed through immunofluorescence and western blot, and cell migration, cell apoptosis and cell cycle were examined with applications of wound healing, TUNEL staining and flow cytometry. TRPM7, PI3K/AKT, TGF-ß1 and related-proteins were quantified using RT-qPCR and western blot. RESULTS: PNS administration could suppress TRPM7 expression and the viability of HSFs in a dose-dependent manner. Moreover, PNS could restrain the HS formation and ECM deposition by decreasing col 1/3 and α-SMA synthesis, suppressing cell migration, and boosting apoptosis and G1 arrest. Notably, this study revealed that PNS inhibited PI3K/AKT activation in HSFs. Besides, knockdown of TRPM7 enhanced therapeutic effects of PNS on HSFs, but overexpression markedly reversed above mentioned effects of PNS on HSFs. CONCLUSION: This study suggested that PNS hampered scar formation might via inhibiting ECM and stimulating cell apoptosis by modulating the PI3K/AKT signaling. Overall, these findings in the present study could support the use of PNS for preventing HS formation, and TRPM7 may be a novel molecular target for treating HS.


Asunto(s)
Cicatriz Hipertrófica/prevención & control , Panax notoginseng , Extractos Vegetales/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Canales Catiónicos TRPM/antagonistas & inhibidores , Transcitosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cicatriz Hipertrófica/tratamiento farmacológico , Humanos , Extractos Vegetales/uso terapéutico , Canales Catiónicos TRPM/efectos de los fármacos , Canales Catiónicos TRPM/metabolismo
7.
J Nutr Biochem ; 87: 108517, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33011286

RESUMEN

Flavonols are bioactive substances in plant foods. In this study, two flavonols galangin and kaempferol were heated at 100°C for 30 min prior to assessing their effects on barrier function of rat intestinal epithelial (IEC-6) cells. Both heated and unheated flavonols (2.5-20 µmol/L dosages) were nontoxic to the cells up to 48 h post-treatment, and could promote cell viability values to 102.2-141.2% of control. By treatment with 5 µmol/L flavonols for 24 and 48 h, the treated cells time-dependently showed better improved physical and biological barrier functions than the control cells without any flavonol treatment, including higher transepithelial electrical resistance and antibacterial effect but reduced paracellular permeability and bacterial translocation. The results from real-time PCR and western-blot assays indicated that the cells treated with heated and unheated flavonols of 5 µmol/L dosage had up-regulated mRNA (1.13-1.81 folds) and protein (1.15-5.11 folds) expression for zonula occluden-1, occludin, and claudin-1 that are vital to the tight junctions of the cells. Moreover, protein expression of RhoA and ROCK were down-regulated into 0.41-0.98 and 0.40-0.92 folds, respectively, demonstrating a Rho inactivation that led to enhanced cell barrier integrity via the RhoA/ROCK pathway. Overall, galangin was more active than kaempferol to perform three biofunctions like improving cell barrier function, up-regulating tight junctions protein expression, and down-regulating RhoA/ROCK expression. Moreover, the heated flavonols were less effective than the unheated counterparts to perform these biofunctions. It is concluded that this heat treatment of galangin and kaempferol could inhibit their benefits to improve barrier function of IEC-6 cells.


Asunto(s)
Flavonoides/farmacología , Mucosa Intestinal/efectos de los fármacos , Quempferoles/farmacología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Calor , Mucosa Intestinal/metabolismo , Permeabilidad/efectos de los fármacos , Ratas , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Transcitosis/efectos de los fármacos
8.
Drug Deliv ; 27(1): 1474-1490, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33100061

RESUMEN

The emergence of nanomaterials for drug delivery provides the opportunity to avoid the side effects of systemic drug administration and injury caused by the removal of tumors, delivering great promise for future cancer treatments. However, the efficacy of current nano drugs is not significantly better than that of the original drug treatments. The important reason is that nano drugs enter the tumor vasculature, remaining close to the blood vessels and unable to enter the tumor tissue or tumor cells to complete the drug delivery process. The low efficiency of drug penetration into tumors has become a bottleneck restricting the development of nano-drugs. Herein, we present a systematic overview of recent advances on the design of nano-drug carriers in drug delivery systems for enhancing drug penetration into tumors. The review is organized into four sections: The drug penetration process in tumor tissue includes paracellular and transcellular transport, which is summarized first. Strategies that promote tumor penetration are then introduced, including methods of remodeling the tumor microenvironment, charge inversion, dimensional change, and surface modification of ligands which promote tissue penetration. Conclusion and the prospects for the future development of drug penetration are finally briefly illustrated. The review is intended to provide thoughts for effective treatment of cancer by summarizing strategies for promoting the endocytosis of nano drugs into tumor cells.


Asunto(s)
Antineoplásicos/administración & dosificación , Portadores de Fármacos/administración & dosificación , Sistemas de Liberación de Medicamentos/tendencias , Neoplasias/tratamiento farmacológico , Transcitosis/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Animales , Antineoplásicos/metabolismo , Portadores de Fármacos/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Humanos , Nanopartículas/administración & dosificación , Nanopartículas/metabolismo , Neoplasias/metabolismo , Transcitosis/fisiología , Microambiente Tumoral/fisiología
9.
Pharm Biol ; 58(1): 518-527, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32501184

RESUMEN

Context: Oridonin (ORI) has obvious anticancer effects, but its solubility is poor. Nanocrystal (NC) is a novel nano-drug delivery system for increasing bioavailability for ORI. However, the endocytosis and transcytosis behaviours of oridonin nanocrystals (ORI-NCs) through epithelial membrane are still unclear.Objectives: ORI-NCs were prepared and characterized. The in vitro cytotoxicity and endocytosis and transcytosis process on Madin-Darby canine kidney (MDCK) monolayer were investigated.Materials and methods: Anti-solvent precipitation method was adopted in preparation of ORI-NCs. Differential scanning calorimetry (DSC) and X-ray diffraction (XRD) were adopted to explore crystallography of ORI-NCs. Sulforhodamine B (SRB) method was used to test the inhibition effect on proliferation of MDCK cells. Quantitative analysis by HPLC was performed to study the endocytosis and transcytosis of ORI-NCs and ORI bulk drug, and the process was observed by confocal laser spectrum microscopy (CLSM) and flow cytometry.Results: The particle size of ORI-NCs was about 274 nm. The crystallography form of ORI was not changed after prepared into NCs. The dissolution rate of ORI-NCs was higher than pure ORI in 120 min. At higher concentrations (34, 84 and 135 µg/mL), ORI-NCs significantly reduced the cell viability compared with free ORI (p < 0.05, p < 0.01). ORI-NCs demonstrated higher endocytosis in MDCK cells than free ORI (p < 0.01). In the transport process, ORI-NC was taken up into cells in an intact form, and excreted out from basolateral membrane of polarized epithelial cells in an intact form. The internalization and transmembrane amount increased as a function of time.Conclusions: ORI-NCs transported through the MDCK monolayers in an intact form.


Asunto(s)
Diterpenos de Tipo Kaurano/metabolismo , Endocitosis/fisiología , Células Epiteliales/metabolismo , Nanopartículas/metabolismo , Transcitosis/fisiología , Animales , Diterpenos de Tipo Kaurano/farmacología , Perros , Endocitosis/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células de Riñón Canino Madin Darby , Nanopartículas/administración & dosificación , Tamaño de la Partícula , Transcitosis/efectos de los fármacos
10.
Chem Pharm Bull (Tokyo) ; 68(4): 316-325, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32238649

RESUMEN

Discriminatory drug delivery into target cells is essential to effectively elicit the drug activity and to avoid off-target side effects; however, transporting drugs across the cell membrane is difficult due to factors such as molecular size, hydrophilicity, intercellular adhesiveness, and efflux transporters, particularly, in the brain capillary endothelial cells. Drug delivery into the brain is blocked by the blood-brain barrier (BBB). Thus, developing drugs for the central nervous system (CNS) diseases remains a challenge. The approach based on receptor-mediated transcytosis (RMT) can overcome this impassable problem at the BBB. Well-designed molecules for RMT form conjugates with the ligand and drugs via linkers or nanoparticles. Cell penetrating peptides (CPPs), receptor-targeting peptides, and monoclonal antibodies (mAbs) are often used as ligands. The binding of ligand to the receptor on the endothelial cell surface induces endocytosis. Existing exosomes comprising the conjugates move in the cytoplasm and fuse with the opposite plasma membrane to release them. Subsequently, the transcytosed conjugate-loaded drugs or released drugs from the conjugates elicit activity in the brain. As receptors, transferrin receptor (TfR), low-density lipoprotein receptor (LDLR), and insulin receptor (InsR) have been used to intendedly induce transcytosis. Presently, several clinical trials on CNS drugs for Alzheimer's and Parkinson disease are hindered due to poor drug distribution into the brain. Therefore, this strategy based on RMT is a promising method for CNS drugs to be transported into the brain. In this review, I introduce the practicality and possibility of drug delivery into brain across the BBB using RMT.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Fármacos del Sistema Nervioso Central/farmacología , Sistemas de Liberación de Medicamentos , Receptores de Superficie Celular/metabolismo , Transcitosis/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Fármacos del Sistema Nervioso Central/química , Humanos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Receptores de Superficie Celular/efectos de los fármacos
11.
Transl Stroke Res ; 11(5): 1012-1027, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31907728

RESUMEN

Blood-brain barrier (BBB) disruption is one of the critical mechanisms of brain injury induced by subarachnoid hemorrhage (SAH). Past studies have often focused on the tight junctions of endothelial cells. However, low transcellular transport levels also play an important role in the normal functioning of the BBB. Major facilitator superfamily domain-containing 2a (Mfsd2a) has been demonstrated to be essential for the maintenance of the normal BBB. Our present study aimed to explore the roles and mechanisms of Mfsd2a in BBB disruption after SAH. In this study, a prechiasmatic cistern single-injection model was used to produce experimental SAH in Sprague-Dawley rats. Specific small-interfering RNA and plasmids were used to downregulate and upregulate the expression of Mfsd2a prior to assessments in our SAH model. Omega-3 fatty acid deficiency diet was used to reduce DHA in rat brain. The expression level of Mfsd2a decreased significantly after SAH and reached its lowest level at 72 h post-SAH, which then gradually recovered. At 72 h after SAH, BBB function was disrupted; upregulation of Mfsd2a reversed this damage, whereas downregulation of Mfsd2a exacerbated this damage. These effects were primarily mediated through transcellular transport, especially for changes in caveolae compared to those of tight junctions. After stopping the supply of omega-3 fatty acids, the effect of Mfsd2a on inhibition of caveolae and protection of the blood-brain barrier was eliminated. Taken together, Mfsd2a inhibits caveolae-based transcellular transport by transporting omega-3 fatty acids to protect the BBB after SAH.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Caveolas/metabolismo , Hemorragia Subaracnoidea/tratamiento farmacológico , Transcitosis/efectos de los fármacos , Animales , Lesiones Encefálicas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/metabolismo
12.
Autophagy ; 16(6): 1111-1129, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31448673

RESUMEN

Diabetes is a recognized high-risk factor for the development of atherosclerosis, in which macroautophagy/autophagy is emerging to play essential roles. The retention of low-density lipoprotein (LDL) particles in subendothelial space following transcytosis across the endothelium is the initial step of atherosclerosis. Here, we identified that high glucose could promote atherosclerosis by stimulating transcytosis of LDL. By inhibiting AMPK-MTOR-PIK3C3 pathway, high glucose suppresses the CAV-CAVIN-LC3B-mediated autophagic degradation of CAV1; therefore, more CAV1 is accumulated in the cytosol and utilized to form more caveolae in the cell membrane and facilitates the LDL transcytosis across endothelial cells. For a proof of concept, higher levels of lipids were accumulated in the subendothelial space of umbilical venous walls from pregnant women with gestational diabetes mellitus (GDM), compared to those of pregnant women without GDM. Our results reveal that high glucose stimulates LDL transcytosis by a novel CAV1-CAVIN1-LC3B signaling-mediated autophagic degradation pathway. ABBREVIATIONS: 3-MA: 3-methyladenine; ACTB: actin beta; AMPK: AMP-activated protein kinase; Bafi: bafilomycin A1; CAV1: caveolin-1; CAVIN1: caveolae associated protein 1; CSD: the CAV1 scaffolding domain; GDM: gestational diabetes mellitus; IMD: intramembrane domain; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule- associated protein 1 light chain 3; MFI: mean fluorescence intensity; MTOR: mechanistic target of rapamycin kinase; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; SQSTM1/p62: sequestosome 1.


Asunto(s)
Autofagia/genética , Caveolina 1/metabolismo , Glucosa/metabolismo , Lipoproteínas LDL/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcitosis/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Caveolina 1/genética , Fosfatidilinositol 3-Quinasas Clase III/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Glucosa/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Microscopía Electrónica de Transmisión , Proteínas Asociadas a Microtúbulos/genética , Mutación , Embarazo , Proteínas Quinasas/metabolismo , Estabilidad Proteica , Proteínas de Unión al ARN/genética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Transcitosis/efectos de los fármacos
13.
Nanoscale ; 11(45): 22054-22069, 2019 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-31720664

RESUMEN

Nanoparticles capable of penetrating the blood-brain barrier (BBB) will greatly advance the delivery of therapies against brain disorders. Carbon nanotubes hold great potential as delivery vehicles due to their high aspect-ratio and cell-penetrating ability. Studies have shown multiwalled carbon nanotubes (MWCNT) cross the BBB, however they have largely relied on labelling methods to track and quantify transport, or on individual electron microscopy images to qualitatively assess transcytosis. Therefore, new direct and quantitative methods, using well-defined and unlabelled MWCNT, are needed to compare BBB translocation of different MWCNT types. Using highly controlled anionic (-), cationic (+) and non-ionic (0) functionalized MWCNT (fMWCNT), we correlate UV-visible spectroscopy with quantitative transmission electron microscopy, quantified from c. 270 endothelial cells, to examine cellular uptake, BBB transport and neurotoxicity of unlabelled fMWCNT. Our results demonstrate that: (i) a large fraction of cationic and non-ionic, but not anionic fMWCNT become trapped at the luminal brain endothelial cell membrane; (ii) despite high cell association, fMWCNT uptake by brain endothelial cells is low (<1.5% ID) and does not correlate with BBB translocation, (iii) anionic fMWCNT have highest transport levels across an in vitro model of the human BBB compared to non-ionic or cationic nanotubes; and (iv) fMWCNT are not toxic to hippocampal neurons at relevant abluminal concentrations; however, fMWCNT charge has an effect on carbon nanotube neurotoxicity at higher fMWCNT concentrations. This quantitative combination of microscopy and spectroscopy, with cellular assays, provides a crucial strategy to predict brain penetration efficiency and neurotoxicity of unlabelled MWCNT and other nanoparticle technologies relevant to human health.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Membrana Celular/metabolismo , Células Endoteliales/metabolismo , Ensayo de Materiales , Nanotubos de Carbono/química , Transcitosis/efectos de los fármacos , Animales , Transporte Biológico , Barrera Hematoencefálica/ultraestructura , Línea Celular Transformada , Membrana Celular/ultraestructura , Células Endoteliales/ultraestructura , Humanos , Nanotubos de Carbono/ultraestructura , Ratas
14.
ACS Nano ; 13(11): 12470-12486, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31693858

RESUMEN

The development of effective therapies for stroke continues to face repeated translational failures. Brain endothelial cells form paracellular and transcellular barriers to many blood-borne therapies, and the development of efficient delivery strategies is highly warranted. Here, in a mouse model of stroke, we show selective recruitment of clinically used liposomes into the ischemic brain that correlates with biphasic blood brain barrier (BBB) breakdown. Intravenous administration of liposomes into mice exposed to transient middle cerebral artery occlusion took place at early (0.5 and 4 h) and delayed (24 and 48 h) time points, covering different phases of BBB disruption after stroke. Using a combination of in vivo real-time imaging and histological analysis we show that selective liposomal brain accumulation coincides with biphasic enhancement in transcellular transport followed by a delayed impairment to the paracellular barrier. This process precedes neurological damage in the acute phase and maintains long-term liposomal colocalization within the neurovascular unit, which could have great potential for neuroprotection. Levels of liposomal uptake by glial cells are similarly selectively enhanced in the ischemic region late after experimental stroke (2-3 days), highlighting their potential for blocking delayed inflammatory responses or shifting the polarization of microglia/macrophages toward brain repair. These findings demonstrate the capability of liposomes to maximize selective translocation into the brain after stroke and identify two windows for therapeutic manipulation. This emphasizes the benefits of selective drug delivery for efficient tailoring of stroke treatments.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Liposomas , Accidente Cerebrovascular/metabolismo , Animales , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Infarto de la Arteria Cerebral Media/metabolismo , Liposomas/química , Liposomas/farmacocinética , Liposomas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Transcitosis/efectos de los fármacos
15.
PLoS Pathog ; 15(11): e1008014, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31703116

RESUMEN

Clostridium perfringens epsilon toxin (ETX) is responsible for causing the economically devastating disease, enterotoxaemia, in livestock. It is well accepted that ETX causes blood brain barrier (BBB) permeability, however the mechanisms involved in this process are not well understood. Using in vivo and in vitro methods, we determined that ETX causes BBB permeability in mice by increasing caveolae-dependent transcytosis in brain endothelial cells. When mice are intravenously injected with ETX, robust ETX binding is observed in the microvasculature of the central nervous system (CNS) with limited to no binding observed in the vasculature of peripheral organs, indicating that ETX specifically targets CNS endothelial cells. ETX binding to CNS microvasculature is dependent on MAL expression, as ETX binding to CNS microvasculature of MAL-deficient mice was not detected. ETX treatment also induces extravasation of molecular tracers including 376Da fluorescein salt, 60kDA serum albumin, 70kDa dextran, and 155kDA IgG. Importantly, ETX-induced BBB permeability requires expression of both MAL and caveolin-1, as mice deficient in MAL or caveolin-1 did not exhibit ETX-induced BBB permeability. Examination of primary murine brain endothelial cells revealed an increase in caveolae in ETX-treated cells, resulting in dynamin and lipid raft-dependent vacuolation without cell death. ETX-treatment also results in a rapid loss of EEA1 positive early endosomes and accumulation of large, RAB7-positive late endosomes and multivesicular bodies. Based on these results, we hypothesize that ETX binds to MAL on the apical surface of brain endothelial cells, causing recruitment of caveolin-1, triggering caveolae formation and internalization. Internalized caveolae fuse with early endosomes which traffic to late endosomes and multivesicular bodies. We believe that these multivesicular bodies fuse basally, releasing their contents into the brain parenchyma.


Asunto(s)
Toxinas Bacterianas/farmacología , Barrera Hematoencefálica/fisiopatología , Encéfalo/fisiopatología , Caveolina 1/fisiología , Permeabilidad de la Membrana Celular/fisiología , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/fisiología , Transcitosis/efectos de los fármacos , Animales , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Caveolas/efectos de los fármacos , Caveolas/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
Carbohydr Polym ; 207: 428-439, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30600025

RESUMEN

Herein, we describe a novel amphipathic chitosan derivative (N-octyl-N'-phthalyl-O-phosphoryl chitosan, abbreviated as OPPC) as an effective oral delivery platform for P-gp substrates, especially paclitaxel (PTX). OPPC could readily self-assemble into micelles, solubilize and encapsulate PTX into the hydrophobic inner core of OPPC with superior loading capacity to chitosan. PTX/OPPC micelles possessed improved intestinal epithelial permeability and oral bioavailability of PTX evaluated by in situ perfusion and pharmacokinetic studies. In vivo fluorescence imaging revealed enhanced stability and integrity of OPPC micelles in mice gastrointestine. Furthermore, cellular uptake studies revealed effective transport and accumulation of OPPC micelles loading PTX or rhodamine-123 into Caco-2 cells via clathrin/cavelin-mediated endocytosis and OPPC-mediated P-gp inhibition. Mechanistically, the inhibition of P-gp efflux pumps by OPPC resulted from the reduction of membrane fluidity and decreased P-gp ATPase activity. In summary, OPPC micelles may serve as an efficient and promising delivery system for enhancing oral bioavailability of P-gp substrates.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Quitosano/análogos & derivados , Quitosano/química , Portadores de Fármacos/química , Paclitaxel/farmacocinética , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Administración Oral , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/química , Células CACO-2 , Quitosano/síntesis química , Quitosano/toxicidad , Regulación hacia Abajo , Portadores de Fármacos/síntesis química , Portadores de Fármacos/toxicidad , Liberación de Fármacos , Endocitosis/efectos de los fármacos , Humanos , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Masculino , Fluidez de la Membrana/efectos de los fármacos , Ratones Endogámicos BALB C , Micelas , Paclitaxel/administración & dosificación , Paclitaxel/química , Ratas Sprague-Dawley , Solubilidad , Transcitosis/efectos de los fármacos , Verapamilo/farmacocinética
17.
J Pharmacol Exp Ther ; 368(2): 317-325, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30420359

RESUMEN

Prostaglandin transporter Oatp2a1/Slco2a1 is expressed at the apical (AP) membranes of type-1 alveolar epithelial (AT1) cells. To investigate the role of OATP2A1 in prostaglandin E2 (PGE2) handling by alveolar epithelium, we studied PGE2 transport across and secretion from monolayers of rat AT1-like (AT1-L) cells obtained by trans-differentiation of type-2 alveolar epithelial cells isolated from male Wistar rats. Rat AT1-L cells expressed Oatp2a1/Slco2a1, together with smaller amounts of Mrp4/Abcc4 and Oct1/Slc22a1 PGE2 uptake was saturable with Km 43.9 ± 21.9 nM. Transcellular transport of PGE2 across AT1-L cells grown on permeable filters in the AP-to-basolateral (BL) direction was 5-fold greater than that in the reverse direction and was saturable with Km 118 ± 26.8 nM; it was significantly inhibited by OATP inhibitors bromosulfophthalein (BSP) and suramin, and an MRP4 inhibitor, Ceefourin 1. We simultaneously monitored the effects of BSP on the distribution of PGE2 produced by bradykinin-treated AT1-L cells and PGE2-d4 externally added on the AP side of the cells. In the presence of BSP, PGE2 increased more rapidly on the AP side, whereas PGE2-d4 decreased more slowly on the AP side. The decrease in PGE2-d4 from the AP side corresponded well to the increase on the BL side, indicating that intracellular metabolism did not occur. These results suggest that Oatp2a1 and Mrp4 mediate transepithelial transport of PGE2 in the AP-to-BL direction. Therefore, OATP2A1 may be an important regulator of PGE2 in alveolar epithelium by reducing secretion of PGE2 and facilitating "resecretion" of PGE2 present in the alveolar lumen to the interstitial space or blood.


Asunto(s)
Dinoprostona/metabolismo , Transportadores de Anión Orgánico/metabolismo , Alveolos Pulmonares/metabolismo , Mucosa Respiratoria/metabolismo , Transcitosis/fisiología , Animales , Benzotiazoles/farmacología , Relación Dosis-Respuesta a Droga , Masculino , Transportadores de Anión Orgánico/antagonistas & inhibidores , Alveolos Pulmonares/efectos de los fármacos , Ratas , Ratas Wistar , Mucosa Respiratoria/efectos de los fármacos , Transcitosis/efectos de los fármacos , Triazoles/farmacología
18.
Eur Neuropsychopharmacol ; 29(2): 195-210, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30554860

RESUMEN

Methylphenidate (MPH) is the classic treatment for attention deficit hyperactivity disorder (ADHD) among children and adults. Despite its beneficial effects, non-medical use of MPH is nowadays a problem with high impact on society. Thus, our goal was to uncover the neurovascular and cognitive effects of MPH chronic use during a critical period of development in control conditions. For that, male Wistar Kyoto rats were treated with MPH (1.5 or 5 mg/kg/day at weekdays, per os) from P28 to P55. We concluded that the higher dose of MPH caused hippocampal blood-brain barrier (BBB) hyperpermeability by vesicular transport (transcytosis) concomitantly with the presence of peripheral immune cells in the brain parenchyma. These observations were confirmed by in vitro studies, in which the knockdown of caveolin-1 in human brain endothelial cells prevented the increased permeability and leukocytes transmigration triggered by MPH (100 µM, 24 h). Furthermore, MPH led to astrocytic atrophy and to a decrease in the levels of several synaptic proteins and impairment of AKT/CREB signaling, together with working memory deficit assessed in the Y-maze test. On the contrary, we verified that the lower dose of MPH (1.5 mg/kg/day) increased astrocytic processes and upregulated several neuronal proteins as well as signaling pathways involved in synaptic plasticity culminating in working memory improvement. In conclusion, the present study reveals that a lower dose of MPH in normal rats improves memory performance being associated with the modulation of astrocytic morphology and synaptic machinery. However, a higher dose of MPH leads to BBB dysfunction and memory impairment.


Asunto(s)
Estimulantes del Sistema Nervioso Central/farmacología , Hipocampo/efectos de los fármacos , Memoria/efectos de los fármacos , Metilfenidato/farmacología , Transcitosis/efectos de los fármacos , Animales , Animales Recién Nacidos , Antioxidantes/metabolismo , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Hipocampo/anatomía & histología , Hipocampo/ultraestructura , Peroxidación de Lípido/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/ultraestructura , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Transcitosis/fisiología , Regulación hacia Arriba/efectos de los fármacos
19.
Arterioscler Thromb Vasc Biol ; 38(10): 2283-2294, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354216

RESUMEN

Objective- The atheroprotective effects of estrogen are independent of circulating lipid levels. Whether estrogen regulates transcytosis of LDL (low-density lipoprotein) across the coronary endothelium is unknown. Approach and Results- Using total internal reflection fluorescence microscopy, we quantified transcytosis of LDL across human coronary artery endothelial cells from multiple donors. LDL transcytosis was significantly higher in cells from men compared with premenopausal women. Estrogen significantly attenuated LDL transcytosis by endothelial cells from male but not female donors; transcytosis of albumin was not affected. Estrogen caused downregulation of endothelial SR-BI (scavenger receptor class B type 1), and overexpression of SR-BI was sufficient to restore LDL transcytosis. Similarly, depletion of SR-BI by siRNA attenuated endothelial LDL transcytosis and prevented any further effect of estrogen. In contrast, treatment with estrogen had no effect on SR-BI expression by liver cells. Inhibition of estrogen receptors α and ß had no effect on estrogen-mediated attenuation of LDL transcytosis. However, estrogen's effect on LDL transcytosis was blocked by depletion of the GPER (G-protein-coupled estrogen receptor). GPER was found to be enriched in endothelial cells compared with hepatocytes and is reported to signal via transactivation of the EGFR (epidermal growth factor receptor); inhibition of EGFR prevented the effect of estrogen on LDL transcytosis and SR-BI mRNA. Last, SR-BI expression was significantly higher in human coronary artery endothelial cells from male compared with premenopausal female donors. Conclusions- Estrogen significantly inhibits LDL transcytosis by downregulating endothelial SR-BI; this effect requires GPER.


Asunto(s)
Vasos Coronarios/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Estradiol/farmacología , Lipoproteínas LDL/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Depuradores de Clase B/metabolismo , Transcitosis/efectos de los fármacos , Células Cultivadas , Vasos Coronarios/metabolismo , Regulación hacia Abajo , Células Endoteliales/metabolismo , Femenino , Humanos , Masculino , Microscopía Fluorescente/métodos , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Depuradores de Clase B/genética , Factores Sexuales , Transducción de Señal/efectos de los fármacos
20.
BioDrugs ; 32(6): 547-559, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30306341

RESUMEN

Antibody, immuno- and gene therapies developed for neurological indications face a delivery challenge posed by various anatomical and physiological barriers within the central nervous system (CNS); most notably, the blood-brain barrier (BBB). Emerging delivery technologies for biotherapeutics have focused on trans-cellular pathways across the BBB utilizing receptor-mediated transcytosis (RMT). 'Traditionally' targeted RMT receptors, transferrin receptor (TfR) and insulin receptor (IR), are ubiquitously expressed and pose numerous translational challenges during development, including species differences and safety risks. Recent advances in antibody engineering technologies and discoveries of RMT targets and BBB-crossing antibodies that are more BBB-selective have combined to create a new preclinical pipeline of BBB-crossing biotherapeutics with improved efficacy and safety. Novel BBB-selective RMT targets and carrier antibodies have exposed additional opportunities for re-targeting gene delivery vectors or nanocarriers for more efficient brain delivery. Emergence and refinement of core technologies of genetic engineering and editing as well as biomanufacturing of viral vectors and cell-derived products have de-risked the path to the development of systemic gene therapy approaches for the CNS. In particular, brain-tropic viral vectors and extracellular vesicles have recently expanded the repertoire of brain delivery strategies for biotherapeutics. Whereas protein biotherapeutics and bispecific antibodies enabled for BBB transcytosis are rapidly heading towards clinical trials, systemic gene therapy approaches for CNS will likely remain in research phase for the foreseeable future. The promise and limitations of these emerging cross-BBB delivery technologies are further discussed in this article.


Asunto(s)
Productos Biológicos/administración & dosificación , Barrera Hematoencefálica/efectos de los fármacos , Enfermedades del Sistema Nervioso Central/terapia , Sistemas de Liberación de Medicamentos/métodos , Vectores Genéticos/administración & dosificación , Animales , Antígenos CD/metabolismo , Productos Biológicos/uso terapéutico , Barrera Hematoencefálica/metabolismo , Enfermedades del Sistema Nervioso Central/genética , Terapia Genética/métodos , Humanos , Receptor de Insulina/metabolismo , Receptores de Transferrina/metabolismo , Transcitosis/efectos de los fármacos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA