Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Exp Zool B Mol Dev Evol ; 334(7-8): 405-422, 2020 11.
Article in English | MEDLINE | ID: mdl-32488995

ABSTRACT

Carotenoids are lipid-soluble yellow to orange pigments produced by plants, bacteria, and fungi. They are consumed by animals and metabolized to produce molecules essential for gene regulation, vision, and pigmentation. Cave animals represent an interesting opportunity to understand how carotenoid utilization evolves. Caves are devoid of light, eliminating primary production of energy through photosynthesis and, therefore, limiting carotenoid availability. Moreover, the selective pressures that favor carotenoid-based traits, like pigmentation and vision, are relaxed. Astyanax mexicanus is a species of fish with multiple river-adapted (surface) and cave-adapted populations (i.e., Tinaja, Pachón, Molino). Cavefish exhibit regressive features, such as loss of eyes and melanin pigment, and constructive traits, like increased sensory neuromasts and starvation resistance. Here, we show that, unlike surface fish, Tinaja and Pachón cavefish accumulate carotenoids in the visceral adipose tissue. Carotenoid accumulation is not observed in Molino cavefish, indicating that it is not an obligatory consequence of eye loss. We used quantitative trait loci mapping and RNA sequencing to investigate genetic changes associated with carotenoid accumulation. Our findings suggest that multiple stages of carotenoid processing may be altered in cavefish, including absorption and transport of lipids, cleavage of carotenoids into unpigmented molecules, and differential development of intestinal cell types involved in carotenoid assimilation. Our study establishes A. mexicanus as a model to study the genetic basis of natural variation in carotenoid accumulation and how it impacts physiology.


Subject(s)
Carotenoids/metabolism , Characidae/genetics , Animals , Biological Evolution , Carotenoids/analysis , Caves , Characidae/anatomy & histology , Characidae/metabolism , Chromatography, High Pressure Liquid , Chromosome Mapping , Eye/anatomy & histology , Female , Intra-Abdominal Fat/chemistry , Male , Sequence Analysis, DNA , Sequence Analysis, RNA , Transcriptome
2.
Bioorg Med Chem Lett ; 30(8): 127014, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32081448

ABSTRACT

Robust transport of therapeutic peptides and other medicinal molecules across tight epithelial barriers would overcome the major obstacle to oral delivery. We have already demonstrated that peptides conjugated to gangliosides (GM1 and GM3) having non-native short N-acyl groups hijack the endogenous process of intracellular lipid sorting resulting in transcytosis and delivery across epithelial barriers in vitro and in vivo. Here, we report synthetic methodologies to covalently conjugate peptides directly to short-acyl-chain C6-ceramides. We found that the short-acyl-chain ceramide domain is solely responsible for transcytosis in vitro. This clarifies and expands the platform of short-acyl-chain sphingolipids for conjugated peptide delivery across tight mucosal cell barriers from gangliosides to just the ceramide itself.


Subject(s)
Ceramides/metabolism , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Peptides/metabolism , Biological Transport, Active , Cells, Cultured , Ceramides/chemistry , Dose-Response Relationship, Drug , Epithelial Cells/chemistry , Humans , Intestinal Mucosa/chemistry , Intestinal Mucosa/cytology , Molecular Structure , Peptides/chemistry , Structure-Activity Relationship
3.
Bio Protoc ; 8(20)2018 10 20.
Article in English | MEDLINE | ID: mdl-30505885

ABSTRACT

Absorption and secretion of peptide and protein cargoes across single-cell thick mucosal and endothelial barriers occurs by active endocytic and vesicular trafficking that connects one side of the epithelial or endothelial cell (the lumen) with the other (the serosa or blood). Assays that assess this pathway must robustly control for non-specific and passive solute flux through weak or damaged intercellular junctions that seal the epithelial or endothelial cells together. Here we describe an in vitro cell culture Transwell assay for transcytosis of therapeutic peptides linked covalently to various species of the glycosphingolipid GM1. We recently used this assay to develop technology that harnesses endogenous mechanism of lipid sorting across epithelial cell barriers to enable oral delivery of peptide and protein therapeutics.

4.
Elife ; 72018 05 31.
Article in English | MEDLINE | ID: mdl-29851380

ABSTRACT

Transport of biologically active molecules across tight epithelial barriers is a major challenge preventing therapeutic peptides from oral drug delivery. Here, we identify a set of synthetic glycosphingolipids that harness the endogenous process of intracellular lipid-sorting to enable mucosal absorption of the incretin hormone GLP-1. Peptide cargoes covalently fused to glycosphingolipids with ceramide domains containing C6:0 or smaller fatty acids were transported with 20-100-fold greater efficiency across epithelial barriers in vitro and in vivo. This was explained by structure-function of the ceramide domain in intracellular sorting and by the affinity of the glycosphingolipid species for insertion into and retention in cell membranes. In mice, GLP-1 fused to short-chain glycosphingolipids was rapidly and systemically absorbed after gastric gavage to affect glucose tolerance with serum bioavailability comparable to intraperitoneal injection of GLP-1 alone. This is unprecedented for mucosal absorption of therapeutic peptides, and defines a technology with many other clinical applications.


Subject(s)
Absorption, Physiological , Glycosphingolipids/metabolism , Mucous Membrane/metabolism , Peptides/therapeutic use , Animals , Biological Transport, Active , Blood Glucose/metabolism , Cell Nucleus/metabolism , Ceramides/chemistry , Dogs , Epithelial Cells/metabolism , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/metabolism , Glucagon-Like Peptide 1/metabolism , Madin Darby Canine Kidney Cells , Male , Mice, Inbred C57BL , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Reproducibility of Results , Solutions , Structure-Activity Relationship , Transcytosis
5.
J Microbiol ; 56(3): 183-188, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29492875

ABSTRACT

Mucosal surfaces that line our gastrointestinal tract are continuously exposed to trillions of bacteria that form a symbiotic relationship and impact host health and disease. It is only beginning to be understood that the cross-talk between the host and microbiome involve dynamic changes in commensal bacterial population, secretion, and absorption of metabolites between the host and microbiome. As emerging evidence implicates dysbiosis of gut microbiota in the pathology and progression of various diseases such as inflammatory bowel disease, obesity, and allergy, conventional treatments that either overlook the microbiome in the mechanism of action, or eliminate vast populations of microbes via wide-spectrum antibiotics need to be reconsidered. It is also becoming clear the microbiome can influence the body's response to therapeutic treatments for cancers. As such, targeting the microbiome as treatment has garnered much recent attention and excitement from numerous research labs and biotechnology companies. Treatments range from fecal microbial transplantation to precision-guided molecular approaches. Here, we survey recent progress in the development of innovative therapeutics that target the microbiome to treat disease, and highlight key findings in the interplay between host microbes and therapy.


Subject(s)
Dysbiosis/therapy , Gastrointestinal Diseases/therapy , Gastrointestinal Microbiome , Gastrointestinal Tract/drug effects , Probiotics/therapeutic use , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , Dysbiosis/microbiology , Fecal Microbiota Transplantation , Gastrointestinal Diseases/etiology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiopathology , Humans , Hypersensitivity/etiology , Hypersensitivity/therapy , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/therapy , Neoplasms/therapy , Obesity/etiology , Obesity/therapy , Symbiosis
6.
Article in English | MEDLINE | ID: mdl-28213463

ABSTRACT

Polarized epithelial cells line diverse surfaces throughout the body forming selective barriers between the external environment and the internal milieu. To cross these epithelial barriers, large solutes and other cargoes must undergo transcytosis, an endocytic pathway unique to polarized cell types, and significant for the development of cell polarity, uptake of viral and bacterial pathogens, transepithelial signaling, and immunoglobulin transport. Here, we review recent advances in our knowledge of the transcytotic pathway for proteins and lipids. We also discuss briefly the promise of harnessing the molecules that undergo transcytosis as vehicles for clinical applications in drug delivery.


Subject(s)
Endocytosis , Epithelial Cells/metabolism , Biological Transport/physiology , Cell Polarity , Drug Delivery Systems , Endosomes/metabolism , Endosomes/physiology , Humans , Lipid Metabolism , Models, Biological
7.
Biophys J ; 111(12): 2547-2550, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27914621

ABSTRACT

Current models of lipid rafts propose that lipid domains exist as nanoscale compositional fluctuations and these fluctuations can potentially be stabilized into larger domains, consequently better compartmentalizing cellular functions. However, the mechanisms governing stabilized raft assembly and function remain unclear. Here, we test the role of glycolipid crosslinking as a raft targeting and ordering mechanism using the well-studied raft marker cholera toxin B pentamer (CTxB) that binds up to five GM1 glycosphingolipids to enter host cells. We show that when applied to cell-derived giant plasma membrane vesicles, a variant of CTxB containing only a single functional GM1 binding site exhibits significantly reduced partitioning to the ordered phase compared to wild-type CTxB with five binding sites. Moreover, monovalent CTxB does not stabilize membrane domains, unlike wild-type CTxB. These results support the long-held hypothesis that CTxB stabilizes raft domains via a lipid crosslinking mechanism and establish a role for crosslinking in the partitioning of CTxB to ordered domains.


Subject(s)
Cholera Toxin/metabolism , Glycosphingolipids/chemistry , Glycosphingolipids/metabolism , Membrane Microdomains/metabolism , Animals , COS Cells , Chlorocebus aethiops , Protein Transport
8.
Traffic ; 16(6): 572-90, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25690058

ABSTRACT

How the plasma membrane is bent to accommodate clathrin-independent endocytosis remains uncertain. Recent studies suggest Shiga and cholera toxin induce membrane curvature required for their uptake into clathrin-independent carriers by binding and cross-linking multiple copies of their glycosphingolipid receptors on the plasma membrane. But it remains unclear if toxin-induced sphingolipid crosslinking provides sufficient mechanical force for deforming the plasma membrane, or if host cell factors also contribute to this process. To test this, we imaged the uptake of cholera toxin B-subunit into surface-derived tubular invaginations. We found that cholera toxin mutants that bind to only one glycosphingolipid receptor accumulated in tubules, and that toxin binding was entirely dispensable for membrane tubulations to form. Unexpectedly, the driving force for tubule extension was supplied by the combination of microtubules, dynein and dynactin, thus defining a novel mechanism for generating membrane curvature during clathrin-independent endocytosis.


Subject(s)
Cell Membrane/metabolism , Endocytosis , Microtubules/metabolism , Animals , COS Cells , Chlorocebus aethiops , Cholera Toxin/metabolism , Clathrin/metabolism , Dyneins/metabolism , HeLa Cells , Humans , Protein Binding , Receptors, Transferrin/metabolism
9.
J Control Release ; 175: 72-8, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24370893

ABSTRACT

The incretin hormone Glucagon-like peptide 1 (GLP-1) requires delivery by injection for the treatment of Type 2 diabetes mellitus. Here, we test if the properties of glycosphingolipid trafficking in epithelial cells can be applied to convert GLP-1 into a molecule suitable for mucosal absorption. GLP-1 was coupled to the extracellular oligosaccharide domain of GM1 species containing ceramides with different fatty acids and with minimal loss of incretin bioactivity. When applied to apical surfaces of polarized epithelial cells in monolayer culture, only GLP-1 coupled to GM1-ceramides with short- or cis-unsaturated fatty acids trafficked efficiently across the cell to the basolateral membrane by transcytosis. In vivo studies showed mucosal absorption after nasal administration. The results substantiate our recently reported dependence on ceramide structure for trafficking the GM1 across polarized epithelial cells and support the idea that specific glycosphingolipids can be harnessed as molecular vehicles for mucosal delivery of therapeutic peptides.


Subject(s)
Ceramides/chemistry , Drug Carriers/chemistry , G(M1) Ganglioside/chemistry , Glucagon-Like Peptide 1/administration & dosage , Incretins/administration & dosage , Amino Acid Sequence , Animals , Cell Line , Ceramides/metabolism , Diabetes Mellitus, Type 2/drug therapy , Drug Carriers/metabolism , G(M1) Ganglioside/metabolism , Glucagon-Like Peptide 1/chemistry , HEK293 Cells , Humans , Incretins/chemistry , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Transcytosis
10.
J Biol Chem ; 288(36): 25804-25809, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23884419

ABSTRACT

Cholera toxin causes diarrheal disease by binding ganglioside GM1 on the apical membrane of polarized intestinal epithelial cells and trafficking retrograde through sorting endosomes, the trans-Golgi network (TGN), and into the endoplasmic reticulum. A fraction of toxin also moves from endosomes across the cell to the basolateral plasma membrane by transcytosis, thus breeching the intestinal barrier. Here we find that sorting of cholera toxin into this transcytotic pathway bypasses retrograde transport to the TGN. We also find that GM1 sphingolipids can traffic from apical to basolateral membranes by transcytosis in the absence of toxin binding but only if the GM1 species contain cis-unsaturated or short acyl chains in the ceramide domain. We found previously that the same GM1 species are needed to efficiently traffic retrograde into the TGN and endoplasmic reticulum and into the recycling endosome, implicating a shared mechanism of action for sorting by lipid shape among these pathways.


Subject(s)
Ceramides/metabolism , Cholera Toxin/metabolism , Cholera Toxin/pharmacology , G(M1) Ganglioside/metabolism , Transcytosis/drug effects , Animals , Ceramides/genetics , Dogs , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endosomes/genetics , Endosomes/metabolism , G(M1) Ganglioside/genetics , Madin Darby Canine Kidney Cells
11.
Dev Cell ; 23(3): 573-86, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22975326

ABSTRACT

The glycosphingolipid GM1 binds cholera toxin (CT) on host cells and carries it retrograde from the plasma membrane (PM) through endosomes, the trans-Golgi (TGN), and the endoplasmic reticulum (ER) to induce toxicity. To elucidate how a membrane lipid can specify trafficking in these pathways, we synthesized GM1 isoforms with alternate ceramide domains and imaged their trafficking in live cells. Only GM1 with unsaturated acyl chains sorted efficiently from PM to TGN and ER. Toxin binding, which effectively crosslinks GM1 lipids, was dispensable, but membrane cholesterol and the lipid raft-associated proteins actin and flotillin were required. The results implicate a protein-dependent mechanism of lipid sorting by ceramide structure and provide a molecular explanation for the diversity and specificity of retrograde trafficking by CT in host cells.


Subject(s)
Cell Membrane/chemistry , Ceramides/chemistry , Cholera Toxin/chemistry , Endoplasmic Reticulum/chemistry , G(M1) Ganglioside/chemistry , Biological Transport , Cell Membrane/metabolism , Cells, Cultured , Ceramides/metabolism , Cholera Toxin/metabolism , Endoplasmic Reticulum/metabolism , G(M1) Ganglioside/chemical synthesis , G(M1) Ganglioside/metabolism , Humans , Protein Isoforms/chemical synthesis , Protein Isoforms/chemistry , Protein Isoforms/metabolism
12.
Article in English | MEDLINE | ID: mdl-22919642

ABSTRACT

Some bacterial toxins and viruses have evolved the capacity to bind mammalian glycosphingolipids to gain access to the cell interior, where they can co-opt the endogenous mechanisms of cellular trafficking and protein translocation machinery to cause toxicity. Cholera toxin (CT) is one of the best-studied examples, and is the virulence factor responsible for massive secretory diarrhea seen in cholera. CT enters host cells by binding to monosialotetrahexosylganglioside (GM1 gangliosides) at the plasma membrane where it is transported retrograde through the trans-Golgi network (TGN) into the endoplasmic reticulum (ER). In the ER, a portion of CT, the CT-A1 polypeptide, is unfolded and then "retro-translocated" to the cytosol by hijacking components of the ER associated degradation pathway (ERAD) for misfolded proteins. CT-A1 rapidly refolds in the cytosol, thus avoiding degradation by the proteasome and inducing toxicity. Here, we highlight recent advances in our understanding of how the bacterial AB(5) toxins induce disease. We highlight the molecular mechanisms by which these toxins use glycosphingolipid to traffic within cells, with special attention to how the cell senses and sorts the lipid receptors. We also discuss several new studies that address the mechanisms of toxin unfolding in the ER and the mechanisms of CT A1-chain retro-translocation to the cytosol.


Subject(s)
Bacterial Toxins/metabolism , Glycosphingolipids/metabolism , Animals , Cell Membrane/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Eukaryotic Cells/metabolism , Golgi Apparatus/metabolism , Humans , Mammals , Protein Binding , Protein Transport
13.
PLoS One ; 6(9): e24693, 2011.
Article in English | MEDLINE | ID: mdl-21935440

ABSTRACT

BACKGROUND: The glucose-dependent insulinotropic polypeptide (GIP) and the glucagon-like peptide-1 (GLP-1) receptors are considered complementary therapeutic targets for type 2 diabetes. Using recombinant membrane-tethered ligand (MTL) technology, the present study focused on defining optimized modulators of these receptors, as well as exploring how local anchoring influences soluble peptide function. METHODOLOGY/PRINCIPAL FINDINGS: Serial substitution of residue 7 in membrane-tethered GIP (tGIP) led to a wide range of activities at the GIP receptor, with [G(7)]tGIP showing enhanced efficacy compared to the wild type construct. In contrast, introduction of G(7) into the related ligands, tGLP-1 and tethered exendin-4 (tEXE4), did not affect signaling at the cognate GLP-1 receptor. Both soluble and tethered GIP and GLP-1 were selective activators of their respective receptors. Although soluble EXE4 is highly selective for the GLP-1 receptor, unexpectedly, tethered EXE4 was found to be a potent activator of both the GLP-1 and GIP receptors. Diverging from the pharmacological properties of soluble and tethered GIP, the newly identified GIP-R agonists, (i.e. [G(7)]tGIP and tEXE4) failed to trigger cognate receptor endocytosis. In an attempt to recapitulate the dual agonism observed with tEXE4, we conjugated soluble EXE4 to a lipid moiety. Not only did this soluble peptide activate both the GLP-1 and GIP receptors but, when added to receptor expressing cells, the activity persists despite serial washes. CONCLUSIONS: These findings suggest that conversion of a recombinant MTL to a soluble membrane anchored equivalent offers a means to prolong ligand function, as well as to design agonists that can simultaneously act on more than one therapeutic target.


Subject(s)
Cell Membrane/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Cell Line , Enzyme-Linked Immunosorbent Assay , Exenatide , Gastric Inhibitory Polypeptide/pharmacology , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor , Humans , Incretins/pharmacology , Microscopy, Confocal , Peptides/pharmacology , Receptors, Glucagon/metabolism , Venoms/pharmacology
14.
Proc Natl Acad Sci U S A ; 108(38): 15846-51, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21911378

ABSTRACT

Phosphatidylserine (PS) is a relatively minor constituent of biological membranes. Despite its low abundance, PS in the plasma membrane (PM) plays key roles in various phenomena such as the coagulation cascade, clearance of apoptotic cells, and recruitment of signaling molecules. PS also localizes in endocytic organelles, but how this relates to its cellular functions remains unknown. Here we report that PS is essential for retrograde membrane traffic at recycling endosomes (REs). PS was most concentrated in REs among intracellular organelles, and evectin-2 (evt-2), a protein of previously unknown function, was targeted to REs by the binding of its pleckstrin homology (PH) domain to PS. X-ray analysis supported the specificity of the binding of PS to the PH domain. Depletion of evt-2 or masking of intracellular PS suppressed membrane traffic from REs to the Golgi. These findings uncover the molecular basis that controls the RE-to-Golgi transport and identify a unique PH domain that specifically recognizes PS but not polyphosphoinositides.


Subject(s)
Endosomes/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Phosphatidylserines/metabolism , Animals , COS Cells , Chlorocebus aethiops , Crystallography, X-Ray , Endosomes/ultrastructure , Golgi Apparatus/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Intracellular Membranes/ultrastructure , Membrane Proteins/chemistry , Membrane Proteins/genetics , Microscopy, Fluorescence , Microscopy, Immunoelectron , Models, Biological , Phosphatidylserines/chemistry , Protein Binding , Protein Structure, Tertiary , Protein Transport , RNA Interference , Vero Cells
15.
J Am Chem Soc ; 133(40): 15878-81, 2011 Oct 12.
Article in English | MEDLINE | ID: mdl-21905700

ABSTRACT

Cell surface heptahelical G protein-coupled receptors (GPCRs) mediate critical cellular signaling pathways and are important pharmaceutical drug targets. (1) In addition to traditional small-molecule approaches, lipopeptide-based GPCR-derived pepducins have emerged as a new class of pharmaceutical agents. (2, 3) To better understand how pepducins interact with targeted receptors, we developed a cell-based photo-cross-linking approach to study the interaction between the pepducin agonist ATI-2341 and its target receptor, chemokine C-X-C-type receptor 4 (CXCR4). A pepducin analogue, ATI-2766, formed a specific UV-light-dependent cross-link to CXCR4 and to mutants with truncations of the N-terminus, the known chemokine docking site. These results demonstrate that CXCR4 is the direct binding target of ATI-2341 and suggest a new mechanism for allosteric modulation of GPCR activity. Adaptation and application of our findings should prove useful in further understanding pepducin modulation of GPCRs as well as enable new experimental approaches to better understand GPCR signal transduction.


Subject(s)
Peptides/chemistry , Peptides/pharmacology , Receptors, CXCR4/agonists , Receptors, CXCR4/metabolism , Allosteric Regulation/drug effects , Amino Acid Sequence , Cell Line , Humans , Models, Molecular , Molecular Sequence Data , Photochemical Processes , Ultraviolet Rays
16.
J Clin Invest ; 120(12): 4399-4409, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21041954

ABSTRACT

Cholera toxin (CT) causes the massive secretory diarrhea associated with epidemic cholera. To induce disease, CT enters the cytosol of host cells by co-opting a lipid-based sorting pathway from the plasma membrane, through the trans-Golgi network (TGN), and into the endoplasmic reticulum (ER). In the ER, a portion of the toxin is unfolded and retro- translocated to the cytosol. Here, we established zebrafish as a genetic model of intoxication and examined the Derlin and flotillin proteins, which are thought to be usurped by CT for retro-translocation and lipid sorting, respectively. Using antisense morpholino oligomers and siRNA, we found that depletion of Derlin-1, a component of the Hrd-1 retro-translocation complex, was dispensable for CT-induced toxicity. In contrast, the lipid raft-associated proteins flotillin-1 and -2 were required. We found that in mammalian cells, CT intoxication was dependent on the flotillins for trafficking between plasma membrane/endosomes and two pathways into the ER, only one of which appears to intersect the TGN. These results revise current models for CT intoxication and implicate protein scaffolding of lipid rafts in the endo-somal sorting of the toxin-GM1 complex.


Subject(s)
Cholera Toxin/toxicity , Membrane Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Base Sequence , Biological Transport, Active , COS Cells , Cell Line , Chlorocebus aethiops , Cholera Toxin/pharmacokinetics , Endosomes/metabolism , G(M1) Ganglioside/metabolism , Humans , Membrane Microdomains/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , RNA, Small Interfering/genetics , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
17.
Toxins (Basel) ; 2(3): 310-25, 2010 03.
Article in English | MEDLINE | ID: mdl-22069586

ABSTRACT

Cholera toxin (CT), an AB(5)-subunit toxin, enters host cells by binding the ganglioside GM1 at the plasma membrane (PM) and travels retrograde through the trans-Golgi Network into the endoplasmic reticulum (ER). In the ER, a portion of CT, the enzymatic A1-chain, is unfolded by protein disulfide isomerase and retro-translocated to the cytosol by hijacking components of the ER associated degradation pathway for misfolded proteins. After crossing the ER membrane, the A1-chain refolds in the cytosol and escapes rapid degradation by the proteasome to induce disease by ADP-ribosylating the large G-protein Gs and activating adenylyl cyclase. Here, we review the mechanisms of toxin trafficking by GM1 and retro-translocation of the A1-chain to the cytosol.


Subject(s)
Cholera Toxin/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Cell Membrane/metabolism , Cholera Toxin/chemistry , G(M1) Ganglioside/metabolism , Humans , Protein Conformation , Protein Transport
18.
J Mol Biol ; 388(1): 21-9, 2009 Apr 24.
Article in English | MEDLINE | ID: mdl-19281822

ABSTRACT

An in vitro selection search for DNAs capable of catalyzing photochemistry yielded two distinctive deoxyribozymes (DNAzymes) with photolyase activity: UV1C, which repaired thymine dimers within DNA using a UV light of >300 nm wavelength and no extraneous cofactor, and Sero1C, which required the tryptophan metabolite serotonin as cofactor in addition to the UV light. Catalysis by Sero1C conformed to Michaelis-Menten kinetics, and analysis of the action spectrum of Sero1C confirmed that serotonin did indeed serve as a catalytic cofactor rather than as a structural cofactor. Sero1C and UV1C showed strikingly distinct wavelength optima for their respective photoreactivation catalyses. Although the rate enhancements characteristic of the two DNAzymes were similar, the cofactor-requiring Sero1C repaired a substantially broader range of substrates compared to UV1C, including thymine, uracil, and a range of chimeric deoxypyrimidine and ribopyrimidine dimers. Similarities and differences in the properties of these two photolyase DNAzymes suggest, first, that the harnessing of less damaging UV light for the repair of photolesions may have been a primordial catalytic activity of nucleic acids, and, second, the broader substrate range of Sero1C may highlight an evolutionary advantage to coopting amino-acid-like cofactors by functionality-poor nucleic acid enzymes.


Subject(s)
DNA, Catalytic/chemistry , DNA/chemistry , Pyrimidine Dimers/chemistry , Serotonin/chemistry , Base Sequence , DNA Repair , DNA, Catalytic/metabolism , Kinetics , Models, Biological , Molecular Sequence Data , Nucleic Acid Conformation , Photochemistry , Pyrimidine Dimers/metabolism , Substrate Specificity
19.
J Mol Biol ; 365(5): 1326-36, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17141270

ABSTRACT

Among the unexpected chemistries that can be catalyzed by nucleic acid enzymes is photochemistry. We have reported the in vitro selection of a small, cofactor-independent deoxyribozyme, UV1C, capable of repairing thymine dimers in a DNA substrate, most optimally with light at a wavelength of >300 nm. We hypothesized that a guanine quadruplex functioned both as a light antenna and an electron source for the repair of the substrate within the enzyme-substrate complex. Here, we report structural and mechanistic investigations of that hypothesis. Contact-crosslinking and guanosine to inosine mutational studies reveal that the thymine dimer and the guanine quadruplex are positioned close to each other in the deoxyribozyme-substrate complex, and permit us to refine the structure and topology of the folded deoxyribozyme. In exploring the substrate utilization capabilities of UV1C, we find it to be able to repair uracil dimers as well as thymine dimers, as long as they are present in an overall deoxyribonucleotide milieu. Some surprising similarities with bacterial CPD photolyase enzymes are noted.


Subject(s)
DNA, Catalytic/metabolism , Deoxyribodipyrimidine Photo-Lyase/metabolism , Base Sequence , Catalysis/drug effects , Cross-Linking Reagents/pharmacology , Deoxyribose/chemistry , Deoxyribose/metabolism , Guanine/metabolism , Inosine/genetics , Models, Biological , Molecular Sequence Data , Nucleic Acid Conformation/drug effects , Point Mutation/genetics , Pyrimidine Dimers/chemistry , Pyrimidine Dimers/metabolism , RNA/chemistry , Ribose/chemistry , Ribose/metabolism , Substrate Specificity/drug effects
20.
FEMS Microbiol Lett ; 266(2): 129-37, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17156122

ABSTRACT

Cholera toxin (CT), and members of the AB(5) family of toxins enter host cells and hijack the cell's endogenous pathways to induce toxicity. CT binds to a lipid receptor on the plasma membrane (PM), ganglioside GM1, which has the ability to associate with lipid rafts. The toxin can then enter the cell by various modes of receptor-mediated endocytosis and traffic in a retrograde manner from the PM to the Golgi and the endoplasmic reticulum (ER). Once in the ER, a portion of the toxin is unfolded and retro-translocated to the cytosol so as to induce disease. GM1 is the vehicle that carries CT from PM to ER. Thus, the toxin pathway from PM to ER is a lipid-based sorting pathway, which is potentially meditated by the determinants of the GM1 ganglioside structure itself.


Subject(s)
Cell Membrane/metabolism , Cholera Toxin/metabolism , Endocytosis/physiology , Endoplasmic Reticulum/metabolism , Animals , Humans , Models, Biological , Protein Transport/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...