Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Tissue Barriers ; 11(1): 2039003, 2023 Jan 02.
Article in English | MEDLINE | ID: mdl-35262466

ABSTRACT

Cholix (Chx) is secreted by non-pandemic strains of Vibrio cholerae in the intestinal lumen. For this exotoxin to induce cell death in non-polarized cells in the intestinal lamina propria, it must traverse the epithelium in the fully intact form. We identified host cell elements in polarized enterocytes associated with Chx endocytosis and apical to basal (A→B) vesicular transcytosis. This pathway overcomes endogenous mechanisms of apical vesicle recycling and lysosomal targeting by interacting with several host cell proteins that include the 75 kDa glucose-regulated protein (GRP75). Apical endocytosis of Chx appears to involve the single membrane spanning protein TMEM132A, and interaction with furin before it engages GRP75 in apical vesicular structures. Sorting within these apical vesicles results in Chx being trafficked to the basal region of cells in association with the Lectin, Mannose Binding 1 protein LMAN1. In this location, Chx interacts with the basement membrane-specific heparan sulfate proteoglycan perlecan in recycling endosomes prior to its release from this basal vesicular compartment to enter the underlying lamina propria. While the furin and LMAN1 elements of this Chx transcytosis pathway undergo cellular redistribution that are reflective of the polarity shifts noted for coatamer complexes COPI and COPII, GRP75 and perlecan fail to show these dramatic rearrangements. Together, these data define essential steps in the A→B transcytosis pathway accessed by Chx to reach the intestinal lamina propria where it can engage and intoxicate certain non-polarized cells.


The Vibrio cholerae exotoxin protein cholix interacts with a number of host cell proteins, including GRP75, to facilitate its vesicular transcytosis across polarized intestinal epithelial cells following apical endocytosis.


Subject(s)
Furin , Transcytosis , Endocytosis , Membrane Proteins
2.
Int J Pharm X ; 4: 100125, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36065415

ABSTRACT

Parenteral administrations are a mainstay of clinical drug delivery. Intramuscular (IM) injections deposit drug directly into skeletal muscle bellies, providing rapid systemic uptake due to the highly vascularized nature of this site. The potential to inject particulate or non-aqueous materials have also made IM injections useful for long-acting formulations. These attributes have supported a plethora of medicines being approved for IM administration. Despite these many approvals across multiple pharmaceutical categories, mechanisms that control drug release from the injection site, and thus its pharmacokinetic properties, remain poorly understood. Several pre-clinical in vivo animals have been used to model IM drug fate in patients, but these approaches have not consistently predicted clinical outcomes. This lack of a predictive in vivo model and no standardized in vitro tools have limited the options of pharmaceutical scientists to rationally design formulations for IM delivery. Here, we describe a novel, tractable in vitro model informed by dominant extracellular matrix (ECM) components present at the IM injection site. Three charge variants of green florescent protein (GFP) and the impact of three common formulation components were examined in an initial test of this in vitro model. A strongly positively charged GFP was restricted in its release from hydrogels composed of ECM components type I collagen and hyaluronic acid compared to standard and strongly negatively charged GFP. Introduction of commonly used buffers (histidine or acetate) or the non-ionic surfactant polysorbate 20 altered the release properties of these GFP variants in a manner that was dependent upon ECM element composition. In sum, this Simulator of IntraMuscular Injections, termed SIMI, demonstrated distinct release profiles of a protein biopharmaceutical surrogate that could be exploited to interrogate the impact of formulation components to expedite novel drug development and reduce current dependence on potentially non-predictive pre-clinical in vivo models.

3.
Pharmaceutics ; 13(8)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34452131

ABSTRACT

The low permeability of nanoparticles (NPs) across the intestinal epithelium remains a major challenge for their application of delivering macromolecular therapeutic agents via the oral route. Previous studies have demonstrated the epithelial transcytosis capacity of a non-toxic version of Pseudomonas aeruginosa exotoxin A (ntPE). Here, we show that ntPE can be used to deliver the protein cargo green fluorescent protein (GFP) or human growth hormone (hGH), as genetic fusions, across intact rat jejunum in a model where the material is administered by direct intra-luminal injection (ILI) in vivo in a transcytosis process that required less than 15 min. Next, ntPE chemically coupled onto biodegradable alginate/chitosan condensate nanoparticles (AC NPs-ntPE) were shown to transport similarly to ntPE-GFP and ntPE-hGH across rat jejunum. Finally, AC NPs-ntPE loaded with GFP as a model cargo were demonstrated to undergo a similar transcytosis process that resulted in GFP being colocalized with CD11c+ cells in the lamina propria after 30 min. Control NP preparations, not decorated with ntPE, were not observed within polarized epithelial cells or within the cells of the lamina propria. These studies demonstrate the capacity of ntPE to facilitate the transcytosis of a covalently associated protein cargo as well as a biodegradable NP that can undergo transcytosis across the intestinal epithelium to deliver a noncovalently associated protein cargo. In sum, these studies support the potential applications of ntPE to facilitate the oral delivery of macromolecular therapeutics under conditions of covalent or non-covalent association.

4.
J Immunol ; 205(11): 3191-3204, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33148717

ABSTRACT

IL-10 is a potent anti-inflammatory cytokine capable of suppressing a number of proinflammatory signals associated with intestinal inflammatory diseases, such as ulcerative colitis and Crohn's disease. Clinical use of human IL-10 (hIL-10) has been limited by anemia and thrombocytopenia following systemic injection, side effects that might be eliminated by a gut-restricted distribution. We have identified a transcytosis pathway used by cholix, an exotoxin secreted by nonpandemic forms of the intestinal pathogen Vibrio cholerae A nontoxic fragment of the first 386 aa of cholix was genetically fused to hIL-10 to produce recombinant AMT-101. In vitro and in vivo characterization of AMT-101 showed it to efficiently cross healthy human intestinal epithelium (SMI-100) by a vesicular transcytosis process, activate hIL-10 receptors in an engineered U2OS osteosarcoma cell line, and increase cellular phospho-STAT3 levels in J774.2 mouse macrophage cells. AMT-101 was taken up by inflamed intestinal mucosa and activated pSTAT3 in the lamina propria with limited systemic distribution. AMT-101 administered to healthy mice by oral gavage or to cynomolgus monkeys (nonhuman primates) by colonic spray increased circulating levels of IL-1R antagonist (IL-1Ra). Oral gavage of AMT-101 in two mouse models of induced colitis prevented associated pathological events and plasma cytokine changes. Overall, these studies suggest that AMT-101 can efficiently overcome the epithelial barrier to focus biologically active IL-10 to the intestinal lamina propria.


Subject(s)
Colitis/metabolism , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Animals , Cells, Cultured , Colon/metabolism , Crohn Disease/metabolism , Cytokines/metabolism , Female , Humans , Inflammation/metabolism , Macaca fascicularis , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Mucous Membrane/metabolism , Rats , Rats, Wistar , Transcytosis/physiology
5.
Tissue Barriers ; 8(1): 1710429, 2020.
Article in English | MEDLINE | ID: mdl-31928299

ABSTRACT

Cholix (Chx) is expressed by the intestinal pathogen Vibrio cholerae as a single chain of 634 amino acids (~70.7 kDa protein) that folds into three distinct domains, with elements of the second and third domains being involved in accessing the cytoplasm of nonpolarized cells and inciting cell death via ADP-ribosylation of elongation factor 2, respectively. In order to reach nonpolarized cells within the intestinal lamina propria, however, Chx must cross the polarized epithelial barrier in an intact form. Here, we provide invitro and invivo demonstrations that a nontoxic Chx transports across intestinal epithelium via a vesicular trafficking pathway that rapidly achieves vesicular apical to basal (A→B) transcytosis and avoids routing to lysosomes. Specifically, Chx traffics in apical endocytic Rab7+ vesicles and in basal exocytic Rab11+ vesicles with a transition between these domains occurring in the ER-Golgi intermediate compartment (ERGIC) through interactions with the lectin mannose-binding protein 1 (LMAN1) protein that undergoes an intracellular re-distribution that coincides with the re-organization of COPI+ and COPII+ vesicular structures. Truncation studies demonstrated that domain I of Chx alone was sufficient to efficiently complete A→B transcytosis and capable of ferrying genetically conjoined human growth hormone (hGH). These studies provide evidence for a pathophysiological strategy where native Chx exotoxin secreted in the intestinal lumen by nonpandemic V. cholerae can reach nonpolarized cells within the lamina propria in an intact form by using a nondestructive pathway to cross in the intestinal epithelial that appears useful for oral delivery of biopharmaceuticals.One-Sentence Summary: Elements within the first domain of the Cholix exotoxin protein are essential and sufficient for the apical to basal transcytosis of this Vibrio cholerae-derived virulence factor across polarized intestinal epithelial cells.


Subject(s)
ADP-Ribosylation Factors/chemistry , Bacterial Toxins/chemistry , Protein Domains/physiology , Transcytosis/physiology , Humans
6.
J Insect Sci ; 19(1)2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30753607

ABSTRACT

The sweetpotato vine borer, Omphisa anastomosalis (Guenée), is a primarily Asian pest of sweetpotato, Ipomoea batatas L. Damage by O. anastomosalis infestation can cause root yield losses of 30%-50%. A binary sex pheromone for O. anastomosalis, consisting of Type I [(10E,14E)-10,14-hexadecadienal (E10,E14-16:Ald)] and Type II [(3Z,6Z,9Z)-3,6,9-tricosatriene (Z3,Z6,Z9-23:H)] components, was identified in Vietnam from extracts of female pheromone glands. A structurally similar Type II compound [(3Z,6Z,9Z)-3,6,9-docosatriene (Z3,Z6,Z9-22:H)], not recovered from female pheromone glands, was also found to synergize the attractiveness of the Type I component. Additional field work has been needed to determine whether these synergistic enhancements of attractiveness also occur in other parts of the geographical distribution of this moth species. Herein, results of studies are reported which document that both Z3,Z6,Z9-23:H and Z3,Z6,Z9-22:H also synergistically enhance male response to E10,E14-16:Ald in Hawaii sweetpotato field populations. Trap catch tends to be enhanced with increase of dose and lower Type I:Type II ratios. Among the compound doses and ratios tested, trap catch increased with the addition of the Type II component by over 13 times relative to traps baited with the Type I component alone, which significantly enhanced sweetpotato vine borer detection. Using a 2.0 mg: 4.0 mg Type I: Type II loading, there was continued catch over 12 wk, during which time the Type I component weathered at a faster rate than the Type II component. This binary sex pheromone seems to have promise for both monitoring and suppression of field populations of O. anastomosalis throughout its geographical range.


Subject(s)
Aldehydes/pharmacology , Chemotaxis , Moths/physiology , Pheromones/pharmacology , Polyenes/pharmacology , Sex Attractants/pharmacology , Animals , Female , Hawaii , Ipomoea batatas/growth & development
7.
Mol Immunol ; 48(4): 452-62, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21055811

ABSTRACT

The structure of the complement-binding domain of Staphylococcus aureus protein Sbi (Sbi-IV) in complex with ligand C3d is presented. The 1.7Å resolution structure reveals the molecular details of the recognition of thioester-containing fragment C3d of the central complement component C3, involving interactions between residues of Sbi-IV helix α2 and the acidic concave surface of C3d. The complex provides a structural basis for the binding preference of Sbi for native C3 over C3b and explains how Sbi-IV inhibits the interaction between C3d and complement receptor 2. A second C3d binding site on Sbi-IV is identified in the crystal structure that is not observed in related S. aureus C3 inhibitors Efb-C and Ehp. This binding mode perhaps hints as to how Sbi-IV, as part of Sbi, forms a C3b-Sbi adduct and causes futile consumption of C3, an extraordinary aspect of Sbi function that is not shared by any other known Staphylococcal complement inhibitor.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Carrier Proteins/chemistry , Carrier Proteins/immunology , Complement C3d/chemistry , Complement C3d/immunology , Staphylococcus aureus/immunology , Amino Acid Sequence , Amino Acids/immunology , Crystallography, X-Ray , Humans , Ligands , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Scattering, Small Angle , Structure-Activity Relationship , Surface Properties , Titrimetry
8.
J Immunol ; 184(4): 1946-55, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20083651

ABSTRACT

We recently characterized an interaction between the Staphylococcus aureus immune evasion molecule Staphylococcus aureus binder of Ig (Sbi) and complement C3, an interaction mediated primarily through the binding of C3d(g) to Sbi domain IV. Events related to these studies prompted us to investigate via mutagenesis the binding interface of C3d for Sbi domain IV (Sbi-IV), as well as to revisit the controversial issue of the complement receptor 2 (CR2) binding site of C3d. Specifically, we had shown that Sbi domains III and IV fragment binding to C3dg inhibited the latter's binding to CR2. Moreover, a published cocrystal structure of C3d bound to complement inhibitory C-terminal domain of extracellular fibrinogen-binding protein (Efb-C), a structural and functional homolog of Sbi-IV, showed Efb-C binding to a region on the concave face of C3d previously implicated in CR2 binding by our mutagenesis data but not confirmed in the CR2(short consensus repeat [SCR]1-2):C3d cocrystal structure. We have now analyzed by surface plasmon resonance the binding of a series of variant C3dg molecules to biosensor-bound Sbi-IV or CR2(SCR1-2). We found that mutations to the concave face acidic pocket of C3d significantly affected binding to both Sbi-IV and CR2, although there was divergence in which residues were most important in each case. By contrast, no binding defects were seen for mutations made to the sideface of C3d implicated from the cocrystal structure to be involved in binding CR2(SCR1-2). The results with Sbi-IV suggest a mode of binding highly similar to that visualized in the Efb-C:C3d complex. The results with CR2 confirm our earlier mapping studies and cast even further doubt on the physiologic relevance of the complex visualized in the C3d:CR2 cocrystal.


Subject(s)
Bacterial Proteins/chemistry , Carrier Proteins/chemistry , Complement C3d/chemistry , Immune Evasion , Receptors, Complement 3d/chemistry , Staphylococcus aureus/immunology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Complement C3b/antagonists & inhibitors , Complement C3b/genetics , Complement C3b/metabolism , Complement C3d/genetics , Complement C3d/metabolism , Crystallization , Crystallography, X-Ray , DNA Mutational Analysis , Humans , Immune Evasion/genetics , Mice , Mutagenesis, Site-Directed , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/genetics , Peptide Fragments/metabolism , Receptors, Complement 3d/antagonists & inhibitors , Receptors, Complement 3d/genetics , Staphylococcus aureus/genetics
9.
Proteomics ; 10(1): 48-58, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19899078

ABSTRACT

The incorporation of the specialized carbohydrate affinity ligand methacrylamido phenylboronic acid in polyacrylamide gels for SDS-PAGE analysis has been successful for the separation of carbohydrates and has here been adapted for the analysis of post-translationally modified proteins. While conventional SDS-PAGE analysis cannot distinguish between glycated and unglycated proteins, methacrylamido phenylboronate acrylamide gel electrophoresis (mP-AGE) in low loading shows dramatic retention of delta-gluconolactone modified proteins, while the mobility of the unmodified proteins remains unchanged. With gels containing 1% methacrylamido phenylboronate, mP-AGE analysis of gluconoylated recombinant protein Sbi results in the retention of the modified protein at a position expected for a protein that has quadrupled its expected molecular size. Subsequently, mP-AGE was tested on HSA, a protein that is known to undergo glycation under physiological conditions. mP-AGE could distinguish between various carbohydrate-protein adducts, using in vitro glycated HSA, and discriminate early from late glycation states of the protein. Enzymatically glycosylated proteins show no altered retention in the phenylboronate-incorporated gels, rendering this method highly selective for glycated proteins. We reveal that a trident interaction between phenylboronate and the Amadori cis 1,2 diol and amine group provides the molecular basis of this specificity. These results epitomize mP-AGE as an important new proteomics tool for the detection, separation, visualization and identification of protein glycation. This method will aid the design of inhibitors of unwanted carbohydrate modifications in recombinant protein production, ageing, diabetes, cardiovascular diseases and Alzheimer's disease.


Subject(s)
Boronic Acids/chemistry , Electrophoresis, Polyacrylamide Gel/methods , Glycosylation , Proteomics/methods , Serum Albumin/analysis , Electrophoresis, Polyacrylamide Gel/economics , Humans , Molecular Structure , Phosphorylation , Protein Processing, Post-Translational , Proteomics/economics , Serum Albumin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...