Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Genes Dev ; 37(21-24): 998-1016, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38092521

ABSTRACT

Reductions in brain kynurenic acid levels, a neuroinhibitory metabolite, improve cognitive function in diverse organisms. Thus, modulation of kynurenic acid levels is thought to have therapeutic potential in a range of brain disorders. Here we report that the steroid 5-androstene 3ß, 17ß-diol (ADIOL) reduces kynurenic acid levels and promotes associative learning in Caenorhabditis elegans We identify the molecular mechanisms through which ADIOL links peripheral metabolic pathways to neural mechanisms of learning capacity. Moreover, we show that in aged animals, which normally experience rapid cognitive decline, ADIOL improves learning capacity. The molecular mechanisms that underlie the biosynthesis of ADIOL as well as those through which it promotes kynurenic acid reduction are conserved in mammals. Thus, rather than a minor intermediate in the production of sex steroids, ADIOL is an endogenous hormone that potently regulates learning capacity by causing reductions in neural kynurenic acid levels.


Subject(s)
Kynurenic Acid , Steroids , Animals , Kynurenic Acid/pharmacology , Hormones , Mammals
2.
Front Chem ; 11: 1161775, 2023.
Article in English | MEDLINE | ID: mdl-37123874

ABSTRACT

Fat metabolism is an important modifier of aging and longevity in Caenorhabditis elegans. Given the anatomy and hermaphroditic nature of C. elegans, a major challenge is to distinguish fats that serve the energetic needs of the parent from those that are allocated to the progeny. Broadband coherent anti-Stokes Raman scattering (BCARS) microscopy has revealed that the composition and dynamics of lipid particles are heterogeneous both within and between different tissues of this organism. Using BCARS, we have previously succeeded in distinguishing lipid-rich particles that serve as energetic reservoirs of the parent from those that are destined for the progeny. While BCARS microscopy produces high-resolution images with very high information content, it is not yet a widely available platform. Here we report a new approach combining the lipophilic vital dye Nile Red and two-photon fluorescence lifetime imaging microscopy (2p-FLIM) for the in vivo discrimination of lipid particle sub-types. While it is widely accepted that Nile Red staining yields unreliable results for detecting lipid structures in live C. elegans due to strong interference of autofluorescence and non-specific staining signals, our results show that simple FLIM phasor analysis can effectively separate those signals and is capable of differentiating the non-polar lipid-dominant (lipid-storage), polar lipid-dominant (yolk lipoprotein) particles, and the intermediates that have been observed using BCARS microscopy. An advantage of this approach is that images can be acquired using common, commercially available 2p-FLIM systems within about 10% of the time required to generate a BCARS image. Our work provides a novel, broadly accessible approach for analyzing lipid-containing structures in a complex, live whole organism context.

3.
Genes Dev ; 34(15-16): 1033-1038, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32675325

ABSTRACT

Kynurenic acid (KynA) levels link peripheral metabolic status to neural functions including learning and memory. Since neural KynA levels dampen learning capacity, KynA reduction has been proposed as a therapeutic strategy for conditions of cognitive deficit such as neurodegeneration. While KynA is generated locally within the nervous system, its precursor, kynurenine (Kyn), is largely derived from peripheral resources. The mechanisms that import Kyn into the nervous system are poorly understood. Here, we provide genetic, anatomical, biochemical, and behavioral evidence showing that in C. elegans an ortholog of the human LAT1 transporter, AAT-1, imports Kyn into sites of KynA production.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/metabolism , Kynurenic Acid/metabolism , Large Neutral Amino Acid-Transporter 1/physiology , Neurons/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Eating , Kynurenine/metabolism , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Learning/physiology , Mutation
4.
Nat Chem Biol ; 16(10): 1087-1095, 2020 10.
Article in English | MEDLINE | ID: mdl-32572275

ABSTRACT

Caenorhabditis elegans serves as a model for understanding adiposity and its connections to aging. Current methodologies do not distinguish between fats serving the energy needs of the parent, akin to mammalian adiposity, from those that are distributed to the progeny, making it difficult to accurately interpret the physiological implications of fat content changes induced by external perturbations. Using spectroscopic coherent Raman imaging, we determine the protein content, chemical profiles and dynamics of lipid particles in live animals. We find fat particles in the adult intestine to be diverse, with most destined for the developing progeny. In contrast, the skin-like epidermis contains fats that are the least heterogeneous, the least dynamic and have high triglyceride content. These attributes are most consistent with stored somatic energy reservoirs. These results challenge the prevailing practice of assessing C. elegans adiposity by measurements that are dominated by the intestinal fat content.


Subject(s)
Caenorhabditis elegans/physiology , Lipids/chemistry , Spectrum Analysis, Raman/methods , Animals , Lipid Metabolism/physiology
5.
Genes Dev ; 32(1): 14-19, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29386332

ABSTRACT

A general feature of animal aging is decline in learning and memory. Here we show that in Caenorhabditis elegans, a significant portion of this decline is due to accumulation of kynurenic acid (KYNA), an endogenous antagonist of neural N-methyl-D-aspartate receptors (NMDARs). We show that activation of a specific pair of interneurons either through genetic means or by depletion of KYNA significantly improves learning capacity in aged animals even when the intervention is applied in aging animals. KYNA depletion also improves memory. We show that insulin signaling is one factor in KYNA accumulation.


Subject(s)
Aging/metabolism , Kynurenic Acid/metabolism , Learning , Memory , Aging/psychology , Animals , Caenorhabditis elegans/metabolism , Insulin/metabolism , Signal Transduction
6.
PLoS Biol ; 15(8): e2002032, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28763436

ABSTRACT

In species ranging from humans to Caenorhabditis elegans, dietary restriction (DR) grants numerous benefits, including enhanced learning. The precise mechanisms by which DR engenders benefits on processes related to learning remain poorly understood. As a result, it is unclear whether the learning benefits of DR are due to myriad improvements in mechanisms that collectively confer improved cellular health and extension of organismal lifespan or due to specific neural mechanisms. Using an associative learning paradigm in C. elegans, we investigated the effects of DR as well as manipulations of insulin, mechanistic target of rapamycin (mTOR), AMP-activated protein kinase (AMPK), and autophagy pathways-processes implicated in longevity-on learning. Despite their effects on a vast number of molecular effectors, we found that the beneficial effects on learning elicited by each of these manipulations are fully dependent on depletion of kynurenic acid (KYNA), a neuroinhibitory metabolite. KYNA depletion then leads, in an N-methyl D-aspartate receptor (NMDAR)-dependent manner, to activation of a specific pair of interneurons with a critical role in learning. Thus, fluctuations in KYNA levels emerge as a previously unidentified molecular mechanism linking longevity and metabolic pathways to neural mechanisms of learning. Importantly, KYNA levels did not alter lifespan in any of the conditions tested. As such, the beneficial effects of DR on learning can be attributed to changes in a nutritionally sensitive metabolite with neuromodulatory activity rather than indirect or secondary consequences of improved health and extended longevity.


Subject(s)
Association Learning/physiology , Caloric Restriction , Interneurons/metabolism , Kynurenic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Caenorhabditis elegans , Longevity
7.
PLoS Negl Trop Dis ; 11(7): e0005680, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28704396

ABSTRACT

BACKGROUND: Reliance on just one drug to treat the prevalent tropical disease, schistosomiasis, spurs the search for new drugs and drug targets. Inhibitors of human cyclic nucleotide phosphodiesterases (huPDEs), including PDE4, are under development as novel drugs to treat a range of chronic indications including asthma, chronic obstructive pulmonary disease and Alzheimer's disease. One class of huPDE4 inhibitors that has yielded marketed drugs is the benzoxaboroles (Anacor Pharmaceuticals). METHODOLOGY/PRINCIPAL FINDINGS: A phenotypic screen involving Schistosoma mansoni and 1,085 benzoxaboroles identified a subset of huPDE4 inhibitors that induced parasite hypermotility and degeneration. To uncover the putative schistosome PDE4 target, we characterized four PDE4 sequences (SmPDE4A-D) in the parasite's genome and transcriptome, and cloned and recombinantly expressed the catalytic domain of SmPDE4A. Among a set of benzoxaboroles and catechol inhibitors that differentially inhibit huPDE4, a relationship between the inhibition of SmPDE4A, and parasite hypermotility and degeneration, was measured. To validate SmPDE4A as the benzoxaborole molecular target, we first generated Caenorhabditis elegans lines that express a cDNA for smpde4a on a pde4(ce268) mutant (hypermotile) background: the smpde4a transgene restored mutant worm motility to that of the wild type. We then showed that benzoxaborole inhibitors of SmPDE4A that induce hypermotility in the schistosome also elicit a hypermotile response in the C. elegans lines that express the smpde4a transgene, thereby confirming SmPDE4A as the relevant target. CONCLUSIONS/SIGNIFICANCE: The orthogonal chemical, biological and genetic strategies employed identify SmPDE4A's contribution to parasite motility and degeneration, and its potential as a drug target. Transgenic C. elegans is highlighted as a potential screening tool to optimize small molecule chemistries to flatworm molecular drug targets.


Subject(s)
Anthelmintics/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Schistosoma mansoni/drug effects , Animals , Animals, Genetically Modified/genetics , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Catalytic Domain , Cloning, Molecular , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Locomotion/drug effects , Schistosoma mansoni/anatomy & histology , Schistosoma mansoni/physiology
8.
Trends Endocrinol Metab ; 27(8): 586-596, 2016 08.
Article in English | MEDLINE | ID: mdl-27289335

ABSTRACT

An overview of Caenorhabditis elegans as an experimental organism for studying energy balance is presented. Some of the unresolved questions that complicate the interpretation of lipid measurements from C. elegans are highlighted. We review studies that show that both lipid synthesis and lipid breakdown pathways are activated and needed for the longevity of hermaphrodites that lack their germlines. These findings illustrate the heterogeneity of triglyceride-rich lipid particles in C. elegans and reveal specific lipid signals that promote longevity. Finally, we provide a brief overview of feeding behavioral responses of C. elegans to varying nutritional conditions and highlight an unanticipated metabolic pathway that allows the incorporation of experience in feeding behavior.


Subject(s)
Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Lipid Metabolism/physiology , Longevity/physiology , Animals , Behavior, Animal , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Humans , Lipid Metabolism/genetics , Longevity/genetics , Signal Transduction/genetics , Signal Transduction/physiology
9.
Proc Natl Acad Sci U S A ; 113(10): E1343-51, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26903627

ABSTRACT

Common environmental contaminants such as bisphenols and phthalates and persistent contaminants such as polychlorinated biphenyls are thought to influence tissue homeostasis and carcinogenesis by acting as disrupters of endocrine function. In this study we investigated the direct effects of exposure to bisphenol A (BPA), mono-n-butyl phthalate (Pht), and polychlorinated biphenyl 153 (PCB153) on the proteome of primary organotypic cultures of the mouse mammary gland. At low-nanomolar doses each of these agents induced distinct effects on the proteomes of these cultures. Although BPA treatment produced effects that were similar to those induced by estradiol, there were some notable differences, including a reduction in the abundance of retinoblastoma-associated protein and increases in the Rho GTPases Ras-related C3 botulinum toxin substrate 1 (Rac1) and cell division cycle protein CDC42. Both Pht and PCB153 induced changes that were distinct from those induced by estrogen, including decreased levels of the transcriptional corepressor C-terminal binding protein 1. Interestingly, the three chemicals appeared to alter the abundance of distinct splice forms of many proteins as well as the abundance of several proteins that regulate RNA splicing. Our combined results indicate that the three classes of chemical have distinct effects on the proteome of normal mouse mammary cultures, some estrogen-like but most estrogen independent, that influence diverse biological processes including apoptosis, cell adhesion, and proliferation.


Subject(s)
Environmental Pollutants/toxicity , Mammary Glands, Animal/drug effects , Organoids/drug effects , Proteome/metabolism , Proteomics/methods , Animals , Benzhydryl Compounds/toxicity , Chromatography, High Pressure Liquid , Cluster Analysis , Estrogens, Non-Steroidal/toxicity , Female , Humans , Mammary Glands, Animal/metabolism , Mass Spectrometry , Mice , Organoids/metabolism , Phenols/toxicity , Phthalic Acids/toxicity , Polychlorinated Biphenyls/toxicity , Proteome/classification
10.
Annu Rev Genet ; 49: 413-38, 2015.
Article in English | MEDLINE | ID: mdl-26473379

ABSTRACT

The compact nervous system of Caenorhabditis elegans and its genetic tractability are features that make this organism highly suitable for investigating energy balance in an animal system. Here, we focus on molecular components and organizational principles emerging from the investigation of pathways that largely originate in the nervous system and regulate feeding behavior but also peripheral fat regulation through neuroendocrine signaling. We provide an overview of studies aimed at understanding how C. elegans integrate internal and external cues in feeding behavior. We highlight some of the similarities and differences in energy balance between C. elegans and mammals. We also provide our perspective on unresolved issues, both conceptual and technical, that we believe have hampered critical evaluation of findings relevant to fat regulation in C. elegans.


Subject(s)
Adipose Tissue/physiology , Caenorhabditis elegans/physiology , Feeding Behavior/physiology , Nervous System Physiological Phenomena , Animals , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Energy Metabolism , Feedback, Physiological , Neurosecretory Systems/physiology , Octopamine/metabolism , Serotonin/metabolism , Signal Transduction , Tyramine/metabolism
11.
Elife ; 42015 Aug 19.
Article in English | MEDLINE | ID: mdl-26287524
12.
Cell ; 160(1-2): 119-31, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25594177

ABSTRACT

The kynurenine pathway of tryptophan metabolism is involved in the pathogenesis of several brain diseases, but its physiological functions remain unclear. We report that kynurenic acid, a metabolite in this pathway, functions as a regulator of food-dependent behavioral plasticity in C. elegans. The experience of fasting in C. elegans alters a variety of behaviors, including feeding rate, when food is encountered post-fast. Levels of neurally produced kynurenic acid are depleted by fasting, leading to activation of NMDA-receptor-expressing interneurons and initiation of a neuropeptide-y-like signaling axis that promotes elevated feeding through enhanced serotonin release when animals re-encounter food. Upon refeeding, kynurenic acid levels are eventually replenished, ending the elevated feeding period. Because tryptophan is an essential amino acid, these findings suggest that a physiological role of kynurenic acid is in directly linking metabolism to activity of NMDA and serotonergic circuits, which regulate a broad range of behaviors and physiologies.


Subject(s)
Behavior, Animal , Caenorhabditis elegans/metabolism , Feeding Behavior , Kynurenic Acid/metabolism , Animals , Cues , Fasting , Interneurons/metabolism , Kynurenine/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Serotonin , Signal Transduction , Transaminases/metabolism , Tryptophan/metabolism
13.
Crit Rev Biochem Mol Biol ; 50(1): 69-84, 2015.
Article in English | MEDLINE | ID: mdl-25228063

ABSTRACT

C. elegans provides a genetically tractable system for deciphering the homeostatic mechanisms that underlie fat regulation in intact organisms. Here, we provide an overview of the recent advances in the C. elegans fat field with particular attention to studies of C. elegans lipid droplets, the complex links between lipases, autophagy, and lifespan, and analyses of key transcriptional regulatory mechanisms that coordinate lipid homeostasis. These studies demonstrate the ancient origins of mammalian and C. elegans fat regulatory pathways and highlight how C. elegans is being used to identify and analyze novel lipid pathways that are then shown to function similarly in mammals. Despite its many advantages, study of fat regulation in C. elegans is currently faced with a number of conceptual and methodological challenges. We critically evaluate some of the assumptions in the field and highlight issues that we believe should be taken into consideration when interpreting lipid content data in C. elegans.


Subject(s)
Caenorhabditis elegans/metabolism , Lipid Metabolism , Lipids/analysis , Animals , Autophagy , Caenorhabditis elegans/physiology , Gene Expression Regulation , Intestinal Mucosa/metabolism , Lipase/metabolism
14.
PLoS Biol ; 11(11): e1001712, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24260022

ABSTRACT

Phenotypic screens can identify molecules that are at once penetrant and active on the integrated circuitry of a whole cell or organism. These advantages are offset by the need to identify the targets underlying the phenotypes. Additionally, logistical considerations limit screening for certain physiological and behavioral phenotypes to organisms such as zebrafish and C. elegans. This further raises the challenge of elucidating whether compound-target relationships found in model organisms are preserved in humans. To address these challenges we searched for compounds that affect feeding behavior in C. elegans and sought to identify their molecular mechanisms of action. Here, we applied predictive chemoinformatics to small molecules previously identified in a C. elegans phenotypic screen likely to be enriched for feeding regulatory compounds. Based on the predictions, 16 of these compounds were tested in vitro against 20 mammalian targets. Of these, nine were active, with affinities ranging from 9 nM to 10 µM. Four of these nine compounds were found to alter feeding. We then verified the in vitro findings in vivo through genetic knockdowns, the use of previously characterized compounds with high affinity for the four targets, and chemical genetic epistasis, which is the effect of combined chemical and genetic perturbations on a phenotype relative to that of each perturbation in isolation. Our findings reveal four previously unrecognized pathways that regulate feeding in C. elegans with strong parallels in mammals. Together, our study addresses three inherent challenges in phenotypic screening: the identification of the molecular targets from a phenotypic screen, the confirmation of the in vivo relevance of these targets, and the evolutionary conservation and relevance of these targets to their human orthologs.


Subject(s)
Caenorhabditis elegans/drug effects , Feeding Behavior/drug effects , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/metabolism , Computer Simulation , Drug Evaluation, Preclinical , Humans , Peristalsis/drug effects , Pharynx/drug effects , Phenotype , Quinolines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Small Molecule Libraries
15.
Methods Cell Biol ; 107: 383-407, 2012.
Article in English | MEDLINE | ID: mdl-22226531

ABSTRACT

In Caenorhabdatis elegans as in other animals, fat regulation reflects the outcome of behavioral, physiological, and metabolic processes. The amenability of C. elegans to experimentation has led to utilization of this organism for elucidating the complex homeostatic mechanisms that underlie energy balance in intact organisms. The optical advantages of C. elegans further offer the possibility of studying cell biological mechanisms of fat uptake, transport, storage, and utilization, perhaps in real time. Here, we discuss the rationale as well as advantages and potential pitfalls of methods used thus far to study metabolism and fat regulation, specifically triglyceride metabolism, in C. elegans. We provide detailed methods for visualization of fat depots in fixed animals using histochemical stains and in live animals by vital dyes. Protocols are provided and discussed for chloroform-based extraction of total lipids from C. elegans homogenates used to assess total triglyceride or phospholipid content by methods such as thin-layer chromatography or used to obtain fatty acid profiles by methods such as gas chromatography/mass spectrometry. Additionally, protocols are provided for the determination of rates of intestinal fatty acid uptake and fatty acid breakdown by ß-oxidation. Finally, we discuss methods for determining rates of de novo fat synthesis and Raman scattering approaches that have recently been employed to investigate C. elegans lipids without reliance on invasive techniques. As the C. elegans fat field is relatively new, we anticipate that the indicated methods will likely be improved upon and expanded as additional researchers enter this field.


Subject(s)
Caenorhabditis elegans/physiology , Energy Metabolism/physiology , Fatty Acids/metabolism , Lipid Metabolism/physiology , Spectrum Analysis, Raman/methods , Staining and Labeling/methods , Triglycerides/metabolism , Animals , Chromatography, Thin Layer , Fluorescent Dyes , Gas Chromatography-Mass Spectrometry , Homeostasis , Intestinal Mucosa/metabolism , Oxazines , Tissue Extracts/chemistry , Tissue Fixation
16.
Nat Chem Biol ; 7(4): 206-13, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21390037

ABSTRACT

The regulation of energy homeostasis integrates diverse biological processes ranging from behavior to metabolism and is linked fundamentally to numerous disease states. To identify new molecules that can bypass homeostatic compensatory mechanisms of energy balance in intact animals, we screened for small-molecule modulators of Caenorhabditis elegans fat content. We report on several molecules that modulate fat storage without obvious deleterious effects on feeding, growth and reproduction. A subset of these compounds also altered fat storage in mammalian and insect cell culture. We found that one of the newly identified compounds exerts its effects in C. elegans through a pathway that requires previously undescribed functions of an AMP-activated kinase catalytic subunit and a transcription factor previously unassociated with fat regulation. Thus, our strategy identifies small molecules that are effective within the context of intact animals and reveals relationships between new pathways that operate across phyla to influence energy homeostasis.


Subject(s)
Caenorhabditis elegans/metabolism , Fats/metabolism , Lipid Metabolism , AMP-Activated Protein Kinases/metabolism , Adipose Tissue/metabolism , Animals , Caenorhabditis elegans Proteins/metabolism , Catalysis , Energy Metabolism , Homeostasis
17.
Cell Metab ; 12(4): 398-410, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20889131

ABSTRACT

Acyl-CoA synthases are important for lipid synthesis and breakdown, generation of signaling molecules, and lipid modification of proteins, highlighting the challenge of understanding metabolic pathways within intact organisms. From a C. elegans mutagenesis screen, we found that loss of ACS-3, a long-chain acyl-CoA synthase, causes enhanced intestinal lipid uptake, de novo fat synthesis, and accumulation of enlarged, neutral lipid-rich intestinal depots. Here, we show that ACS-3 functions in seam cells, epidermal cells anatomically distinct from sites of fat uptake and storage, and that acs-3 mutant phenotypes require the nuclear hormone receptor NHR-25, a key regulator of C. elegans molting. Our findings suggest that ACS-3-derived long-chain fatty acyl-CoAs, perhaps incorporated into complex ligands such as phosphoinositides, modulate NHR-25 function, which in turn regulates an endocrine program of lipid uptake and synthesis. These results reveal a link between acyl-CoA synthase function and an NR5A family nuclear receptor in C. elegans.


Subject(s)
Caenorhabditis elegans/metabolism , Coenzyme A Ligases/physiology , DNA-Binding Proteins/physiology , Fats/metabolism , Transcription Factors/physiology , Animals , Coenzyme A Ligases/genetics , Intestinal Mucosa/metabolism , Lipids/biosynthesis , Mutagenesis, Site-Directed , Receptors, Cytoplasmic and Nuclear
18.
J Biol Chem ; 282(20): 14836-44, 2007 May 18.
Article in English | MEDLINE | ID: mdl-17389606

ABSTRACT

The low affinity IgE receptor, FcepsilonRII (CD23), is both a positive and negative regulator of IgE synthesis. The proteinase activity that converts the membrane-bound form of CD23 into a soluble species (sCD23) is an important regulator of the function of CD23 and may be an important therapeutic target for the control of allergy and inflammation. We have characterized the catalytic activity of ADAM (a disintegrin and metalloproteinase) 10 toward human CD23. We found that ADAM10 efficiently catalyzes the cleavage of peptides derived from two distinct cleavage sites in the CD23 backbone. Tissue inhibitors of metalloproteinases and a specific prodomain-based inhibitor of ADAM10 perturb the release of endogenously produced CD23 from human leukemia cell lines as well as primary cultures of human B-cells. Expression of a mutant metalloproteinase-deficient construct of ADAM10 partially inhibited the production of sCD23. Similarly, small inhibitory RNA knockdown of ADAM10 partially inhibited CD23 release and resulted in the accumulation of the membrane-bound form of CD23 on the cells. ADAM10 contributes to CD23 shedding and thus could be considered a potential therapeutic target for the treatment of allergic disease.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , B-Lymphocytes/metabolism , Membrane Proteins/metabolism , Receptors, IgE/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , ADAM10 Protein , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Animals , Humans , Hypersensitivity/genetics , Hypersensitivity/metabolism , Inflammation/genetics , Inflammation/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Receptors, IgE/genetics , Tissue Inhibitor of Metalloproteinases/metabolism , U937 Cells
19.
J Am Chem Soc ; 125(16): 4708-9, 2003 Apr 23.
Article in English | MEDLINE | ID: mdl-12696879

ABSTRACT

Specific labeling of biomolecules with biochemical and biophysical probes is a central element of proteomics research. Here we describe a coumarin-phosphine dye that undergoes activation of coumarin fluorescence upon Staudinger ligation with azides. Since azides can be metabolically incorporated into cellular proteins and oligosaccharides, this dye may be a useful tool for profiling proteins and their posttranslational modifications.


Subject(s)
Azides/chemistry , Coumarins/chemistry , Fluorescent Dyes/chemistry , Phosphines/chemistry , Animals , Fluorescence , Kinetics , Mice , Tetrahydrofolate Dehydrogenase/analysis , Tetrahydrofolate Dehydrogenase/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...