Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
PLoS Genet ; 19(9): e1010944, 2023 09.
Article in English | MEDLINE | ID: mdl-37721936

ABSTRACT

Some types of collagens, including transmembrane MACIT collagens and C. elegans cuticle collagens, are N-terminally cleaved at a dibasic site that resembles the consensus for furin or other proprotein convertases of the subtilisin/kexin (PCSK) family. Such cleavage may release transmembrane collagens from the plasma membrane and affect extracellular matrix assembly or structure. However, the functional consequences of such cleavage are unclear and evidence for the role of specific PCSKs is lacking. Here, we used endogenous collagen fusions to fluorescent proteins to visualize the secretion and assembly of the first collagen-based cuticle in C. elegans and then tested the role of the PCSK BLI-4 in these processes. Unexpectedly, we found that cuticle collagens SQT-3 and DPY-17 are secreted into the extraembryonic space several hours before cuticle matrix assembly. Furthermore, this early secretion depends on BLI-4/PCSK; in bli-4 and cleavage-site mutants, SQT-3 and DPY-17 are not efficiently secreted and instead form large intracellular puncta. Their later assembly into cuticle matrix is reduced but not entirely blocked. These data reveal a role for collagen N-terminal processing in intracellular trafficking and the control of matrix assembly in vivo. Our observations also prompt a revision of the classic model for C. elegans cuticle matrix assembly and the pre-cuticle-to-cuticle transition, suggesting that cuticle layer assembly proceeds via a series of regulated steps and not simply by sequential secretion and deposition.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Subtilisin , Animals , Amino Acid Sequence , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Collagen/genetics , Collagen/metabolism , Proprotein Convertases/genetics , Proprotein Convertases/metabolism , Subtilisin/genetics , Subtilisin/metabolism
2.
PLoS Genet ; 18(8): e1010348, 2022 08.
Article in English | MEDLINE | ID: mdl-35960773

ABSTRACT

Epithelial cells secrete apical extracellular matrices to form protruding structures such as denticles, ridges, scales, or teeth. The mechanisms that shape these structures remain poorly understood. Here, we show how the actin cytoskeleton and a provisional matrix work together to sculpt acellular longitudinal alae ridges in the cuticle of adult C. elegans. Transient assembly of longitudinal actomyosin filaments in the underlying lateral epidermis accompanies deposition of the provisional matrix at the earliest stages of alae formation. Actin is required to pattern the provisional matrix into longitudinal bands that are initially offset from the pattern of longitudinal actin filaments. These bands appear ultrastructurally as alternating regions of adhesion and separation within laminated provisional matrix layers. The provisional matrix is required to establish these demarcated zones of adhesion and separation, which ultimately give rise to alae ridges and their intervening valleys, respectively. Provisional matrix proteins shape the alae ridges and valleys but are not present within the final structure. We propose a morphogenetic mechanism wherein cortical actin patterns are relayed to the laminated provisional matrix to set up distinct zones of matrix layer separation and accretion that shape a permanent and acellular matrix structure.


Subject(s)
Actins , Caenorhabditis elegans , Actins/metabolism , Animals , Caenorhabditis elegans/metabolism , Cytoskeleton/genetics , Extracellular Matrix/metabolism , Morphogenesis
3.
PLoS Genet ; 16(11): e1009188, 2020 11.
Article in English | MEDLINE | ID: mdl-33141826

ABSTRACT

Zona Pellucida domain (ZP) proteins are critical components of the body's external-most protective layers, apical extracellular matrices (aECMs). Although their loss or dysfunction is associated with many diseases, it remains unclear how ZP proteins assemble in aECMs. Current models suggest that ZP proteins polymerize via their ZPn subdomains, while ZPc subdomains modulate ZPn behavior. Using the model organism C. elegans, we investigated the aECM assembly of one ZP protein, LET-653, which shapes several tubes. Contrary to prevailing models, we find that LET-653 localizes and functions via its ZPc domain. Furthermore, we show that ZPc domain function requires cleavage at the LET-653 C-terminus, likely in part to relieve inhibition of the ZPc by the ZPn domain, but also to promote some other aspect of ZPc domain function. In vitro, the ZPc, but not ZPn, domain bound crystalline aggregates. These data offer a new model for ZP function whereby the ZPc domain is primarily responsible for matrix incorporation and tissue shaping.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Extracellular Matrix/metabolism , Mucins/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Cell Line , Drosophila , Embryo, Nonmammalian , Models, Animal , Mucins/genetics , Mutation , Protein Aggregates/genetics , Protein Domains/genetics
4.
Semin Cell Dev Biol ; 67: 123-131, 2017 07.
Article in English | MEDLINE | ID: mdl-27178486

ABSTRACT

A seamless tube is a very narrow-bore tube that is composed of a single cell with an intracellular lumen and no adherens or tight junctions along its length. Many capillaries in the vertebrate vascular system are seamless tubes. Seamless tubes also are found in invertebrate organs, including the Drosophila trachea and the Caenorhabditis elegans excretory system. Seamless tube cells can be less than a micron in diameter, and they can adopt very simple "doughnut-like" shapes or very complex, branched shapes comparable to those of neurons. The unusual topology and varied shapes of seamless tubes raise many basic cell biological questions about how cells form and maintain such structures. The prevalence of seamless tubes in the vascular system means that answering such questions has significant relevance to human health. In this review, we describe selected examples of seamless tubes in animals and discuss current models for how seamless tubes develop and are shaped, focusing particularly on insights that have come from recent studies in Drosophila and C. elegans.


Subject(s)
Capillaries/cytology , Endothelial Cells/ultrastructure , Epithelial Cells/ultrastructure , Morphogenesis/genetics , Trachea/cytology , Vascular Diseases/pathology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Capillaries/anatomy & histology , Capillaries/metabolism , Cell Polarity , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Exocytosis , Gene Expression Regulation , Humans , Models, Biological , Pinocytosis , Trachea/anatomy & histology , Trachea/metabolism , Vascular Diseases/genetics , Vascular Diseases/metabolism
5.
PLoS Genet ; 12(8): e1006205, 2016 08.
Article in English | MEDLINE | ID: mdl-27482894

ABSTRACT

Most epithelial cells secrete a glycoprotein-rich apical extracellular matrix that can have diverse but still poorly understood roles in development and physiology. Zona Pellucida (ZP) domain glycoproteins are common constituents of these matrices, and their loss in humans is associated with a number of diseases. Understanding of the functions, organization and regulation of apical matrices has been hampered by difficulties in imaging them both in vivo and ex vivo. We identified the PAN-Apple, mucin and ZP domain glycoprotein LET-653 as an early and transient apical matrix component that shapes developing epithelia in C. elegans. LET-653 has modest effects on shaping of the vulva and epidermis, but is essential to prevent lumen fragmentation in the very narrow, unicellular excretory duct tube. We were able to image the transient LET-653 matrix by both live confocal imaging and transmission electron microscopy. Structure/function and fluorescence recovery after photobleaching studies revealed that LET-653 exists in two separate luminal matrix pools, a loose fibrillar matrix in the central core of the lumen, to which it binds dynamically via its PAN domains, and an apical-membrane-associated matrix, to which it binds stably via its ZP domain. The PAN domains are both necessary and sufficient to confer a cyclic pattern of duct lumen localization that precedes each molt, while the ZP domain is required for lumen integrity. Ectopic expression of full-length LET-653, but not the PAN domains alone, could expand lumen diameter in the developing gut tube, where LET-653 is not normally expressed. Together, these data support a model in which the PAN domains regulate the ability of the LET-653 ZP domain to interact with other factors at the apical membrane, and this ZP domain interaction promotes expansion and maintenance of lumen diameter. These data identify a transient apical matrix component present prior to cuticle secretion in C. elegans, demonstrate critical roles for this matrix component in supporting lumen integrity within narrow bore tubes such as those found in the mammalian microvasculature, and reveal functional importance of the evolutionarily conserved ZP domain in this tube protecting activity.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Extracellular Matrix/genetics , Glycoproteins/genetics , Mucins/genetics , Animals , Caenorhabditis elegans/chemistry , Caenorhabditis elegans/ultrastructure , Caenorhabditis elegans Proteins/biosynthesis , Caenorhabditis elegans Proteins/chemistry , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Gene Expression Regulation , Glycoproteins/biosynthesis , Glycoproteins/chemistry , Microscopy, Electron, Transmission , Mucins/biosynthesis , Mucins/chemistry , Protein Domains/genetics , Structure-Activity Relationship , Zona Pellucida/chemistry , Zona Pellucida/metabolism , Zona Pellucida/ultrastructure
6.
Mol Carcinog ; 55(8): 1243-50, 2016 08.
Article in English | MEDLINE | ID: mdl-26333016

ABSTRACT

Increased activity of B-Raf has been identified in approximately 7% of human cancers. Treatment of Eker rats (Tsc-2(EK/+) ), bearing a mutation in one allele of the tuberous sclerosis-2 (Tsc-2) gene, with the nephrocarcinogen 2,3,5-tris-(glutathion-S-yl) hydroquinone (TGHQ) results in loss of the wild-type allele of Tsc-2 in renal preneoplastic lesions and tumors. These tumors have increased protein expression of B-Raf, C-Raf (Raf-1), and increased expression and activity of ERK kinase. Similar changes are observed in Raf kinases following TGHQ-mediated transformation of primary renal epithelial cells derived from Tsc-2(EK/+) rats (QTRRE cells), cells that are also null for tuberin. Herein, we utilized LC-MS/MS to identify constitutive phosphorylation of S345 and S483 in both 100- and 95-kDa forms of B-Raf in QTRRE cells. Using microRotofor liquid-phase isoelectric focusing, we identified four fractions of B-Raf that contain different post-translational modification profiles in QTRRE cells. Amplification of the kinase domain of B-Raf from QTRRE cells, outer-stripe of the outer medulla of 8-month TGHQ- or vehicle-treated Tsc-2(+/+) and Tsc-2(EK/+) rats, as well as tumors excised from 8-month TGHQ-treated Tsc-2(EK/+) rats revealed three splice variants of B-Raf within the kinase domain. These splice variants differed by approximately 340, 544, and 600 bp; confirmed by sequencing. No point mutations within the kinase domain of B-Raf were identified. In addition, B-Raf/Raf-1/14-3-3 complex formation in the QTRRE cells was decreased by sorafenib, with concomitant selective decreases in p-ERK levels. Transcriptional and post-translational characterization of critical kinases, such as B-Raf, may contribute to the progression of tuberous sclerosis RCC. (246/250) © 2015 Wiley Periodicals, Inc.


Subject(s)
Carcinoma, Renal Cell/metabolism , Glutathione/analogs & derivatives , Hydroquinones/toxicity , Kidney Neoplasms/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Tuberous Sclerosis/metabolism , Animals , Carcinoma, Renal Cell/chemically induced , Carcinoma, Renal Cell/genetics , Cell Line , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Glutathione/toxicity , Humans , Kidney Neoplasms/chemically induced , Kidney Neoplasms/genetics , Male , Neoplasms, Experimental , Phosphorylation , Protein Domains , Protein Processing, Post-Translational , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins c-raf , RNA Splicing/drug effects , Rats , Tuberous Sclerosis/chemically induced , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency
7.
bioRxiv ; 2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37333289

ABSTRACT

Some types of collagens, including transmembrane MACIT collagens and C. elegans cuticle collagens, are N-terminally cleaved at a dibasic site that resembles the consensus for furin or other proprotein convertases of the subtilisin/kexin (PCSK) family. Such cleavage may release transmembrane collagens from the plasma membrane and affect extracellular matrix assembly or structure. However, the functional consequences of such cleavage are unclear and evidence for the role of specific PCSKs is lacking. Here, we used endogenous collagen fusions to fluorescent proteins to visualize the secretion and assembly of the first collagen-based cuticle in C. elegans and then tested the role of the PCSK BLI-4 in these processes. Unexpectedly, we found that cuticle collagens SQT-3 and DPY-17 are secreted into the extraembryonic space several hours before cuticle matrix assembly. Furthermore, this early secretion depends on BLI-4/PCSK; in bli-4 and cleavage-site mutants, SQT-3 and DPY-17 are not efficiently secreted and instead form large intracellular aggregates. Their later assembly into cuticle matrix is reduced but not entirely blocked. These data reveal a role for collagen N-terminal processing in intracellular trafficking and in the spatial and temporal restriction of matrix assembly in vivo . Our observations also prompt a revision of the classic model for C. elegans cuticle matrix assembly and the pre-cuticle-to-cuticle transition, suggesting that cuticle layer assembly proceeds via a series of regulated steps and not simply by sequential secretion and deposition.

8.
Toxicol Sci ; 186(2): 221-241, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35134991

ABSTRACT

Elucidation of predictive fluidic biochemical markers to detect and monitor chemical-induced neurodegeneration has been a major challenge due to a lack of understanding of molecular mechanisms driving altered neuronal morphology and function, as well as poor sensitivity in methods to quantify low-level biomarkers in bodily fluids. Here, we evaluated 5 neurotoxicants (acetaminophen [negative control], bisindolylmaleimide-1, colchicine, doxorubicin, paclitaxel, and rotenone) in human-induced pluripotent stem cell-derived neurons to profile secreted microRNAs (miRNAs) at early and late stages of decline in neuronal cell morphology and viability. Based on evaluation of these morphological (neurite outgrowth parameters) and viability (adenosine triphosphate) changes, 2 concentrations of each chemical were selected for analysis in a human 754 miRNA panel: a low concentration with no/minimal effect on cell viability but a significant decrease in neurite outgrowth, and a high concentration with a significant decrease in both endpoints. A total of 39 miRNAs demonstrated significant changes (fold-change ≥ 1.5 or ≤ 0.67, p value < .01) with at least 1 exposure. Further analyses of targets modulated by these miRNAs revealed 38 key messenger RNA targets with roles in neurological dysfunctions, and identified transforming growth factor-beta (TGF-ß) signaling as a commonly enriched pathway. Of the 39 miRNAs, 5 miRNAs, 3 downregulated (miR-20a, miR-30b, and miR-30d) and 3 upregulated (miR-1243 and miR-1305), correlated well with morphological changes induced by multiple neurotoxicants and were notable based on their relationship to various neurodegenerative conditions and/or key pathways, such as TGF-ß signaling. These datasets reveal miRNA candidates that warrant further evaluation as potential safety biomarkers of chemical-induced neurodegeneration.


Subject(s)
Induced Pluripotent Stem Cells , MicroRNAs , Biomarkers/metabolism , Gene Expression Profiling/methods , Humans , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/metabolism , Neurons , Transforming Growth Factor beta/metabolism
9.
ALTEX ; 39(4): 560-582, 2022.
Article in English | MEDLINE | ID: mdl-35502629

ABSTRACT

Drug-induced neurotoxicity is a leading cause of safety-related attrition for therapeutics in clinical trials, often driven by poor predictivity of preclinical in vitro and in vivo models of neurotoxicity. Over a dozen different iPSC-derived 3D spheroids have been described in recent years, but their ability to predict neurotoxicity in patients has not been evaluated nor compared with the predictive power of nonclinical species. To assess the predictive capabilities of human iPSC-derived neural spheroids (microBrains), we used 84 structurally diverse pharmaceuticals with robust clinical and pre-clinical datasets with varying degrees of seizurogenic and neurodegenerative liability. Drug-induced changes in neural viability and phenotypic calcium bursts were assessed using 7 endpoints based on calcium oscillation profiles and cel-lular ATP levels. These endpoints, normalized by therapeutic exposure, were used to build logistic regression models to establish endpoint cutoffs and evaluate probability for clinical neurotoxicity. The neurotoxicity score calculated from the logistic regression model could distinguish neurotoxic from non-neurotoxic clinical molecules with a specificity as high as 93.33% and a sensitivity of 53.49%, demonstrating a very low false positive rate for the prediction of seizures, convulsions, and neurodegeneration. In contrast, nonclinical species showed a higher sensitivity (75%) but much lower specificity (30.4%). The neural spheroids demonstrated higher likelihood ratio positive and inverse likelihood ratio neg-ative values compared with nonclinical safety studies. This assay has the potential to be used as a predictive assay to detect neurotoxicity in early drug discovery, aiding in the early identification of compounds that eventually may fail due to neurotoxicity.


Subject(s)
Induced Pluripotent Stem Cells , Neurotoxicity Syndromes , Humans , Neurotoxicity Syndromes/etiology , Seizures/chemically induced , Calcium Signaling , Pharmaceutical Preparations
10.
Genetics ; 219(3)2021 11 05.
Article in English | MEDLINE | ID: mdl-34740248

ABSTRACT

The Patched-related superfamily of transmembrane proteins can transport lipids or other hydrophobic molecules across cell membranes. While the Hedgehog receptor Patched has been intensively studied, much less is known about the biological roles of other Patched-related family members. Caenorhabditis elegans has a large number of Patched-related proteins, despite lacking a canonical Hedgehog pathway. Here, we show that PTR-4 promotes the assembly of the precuticle apical extracellular matrix, a transient and molecularly distinct matrix that precedes and patterns the later collagenous cuticle or exoskeleton. ptr-4 mutants share many phenotypes with precuticle mutants, including defects in eggshell dissolution, tube shaping, alae (cuticle ridge) structure, molting, and cuticle barrier function. PTR-4 localizes to the apical side of a subset of outward-facing epithelia, in a cyclical manner that peaks when precuticle matrix is present. Finally, PTR-4 is required to limit the accumulation of the lipocalin LPR-3 and to properly localize the Zona Pellucida domain protein LET-653 within the precuticle. We propose that PTR-4 transports lipids or other hydrophobic components that help to organize the precuticle and that the cuticle and molting defects seen in ptr-4 mutants result at least in part from earlier disorganization of the precuticle.


Subject(s)
Extracellular Matrix , Membrane Proteins , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , CRISPR-Cas Systems/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microscopy, Electron, Transmission , Molting/genetics , Mucins/metabolism , Mutation , Protein Domains/genetics
11.
J Dev Biol ; 8(4)2020 Oct 06.
Article in English | MEDLINE | ID: mdl-33036165

ABSTRACT

Apical extracellular matrices (aECMs) coat exposed surfaces of epithelia to shape developing tissues and protect them from environmental insults. Despite their widespread importance for human health, aECMs are poorly understood compared to basal and stromal ECMs. The nematode Caenorhabditis elegans contains a variety of distinct aECMs, some of which share many of the same types of components (lipids, lipoproteins, collagens, zona pellucida domain proteins, chondroitin glycosaminoglycans and proteoglycans) with mammalian aECMs. These aECMs include the eggshell, a glycocalyx-like pre-cuticle, both collagenous and chitin-based cuticles, and other understudied aECMs of internal epithelia. C. elegans allows rapid genetic manipulations and live imaging of fluorescently-tagged aECM components, and is therefore providing new insights into aECM structure, trafficking, assembly, and functions in tissue shaping.

12.
Elife ; 92020 09 25.
Article in English | MEDLINE | ID: mdl-32975517

ABSTRACT

Biological tubes must develop and maintain their proper diameter to transport materials efficiently. These tubes are molded and protected in part by apical extracellular matrices (aECMs) that line their lumens. Despite their importance, aECMs are difficult to image in vivo and therefore poorly understood. The Caenorhabditis elegans vulva has been a paradigm for understanding many aspects of organogenesis. Here we describe the vulva luminal matrix, which contains chondroitin proteoglycans, Zona Pellucida (ZP) domain proteins, and other glycoproteins and lipid transporters related to those in mammals. Confocal and transmission electron microscopy revealed, with unprecedented detail, a complex and dynamic aECM. Different matrix factors assemble on the apical surfaces of each vulva cell type, with clear distinctions seen between Ras-dependent (1°) and Notch-dependent (2°) cell types. Genetic perturbations suggest that chondroitin and other aECM factors together generate a structured scaffold that both expands and constricts lumen shape.


Subject(s)
Caenorhabditis elegans/embryology , Extracellular Matrix/metabolism , Glycoproteins/metabolism , Organogenesis , Animals , Embryo, Nonmammalian/embryology , Female , Vulva/embryology
13.
Genetics ; 211(1): 185-200, 2019 01.
Article in English | MEDLINE | ID: mdl-30409789

ABSTRACT

The body's external surfaces and the insides of biological tubes, like the vascular system, are lined by a lipid-, glycoprotein-, and glycosaminoglycan-rich apical extracellular matrix (aECM). aECMs are the body's first line of defense against infectious agents and promote tissue integrity and morphogenesis, but are poorly described relative to basement membranes and stromal ECMs. While some aECM components, such as zona pellucida (ZP) domain proteins, have been identified, little is known regarding the overall composition of the aECM or the mechanisms by which different aECM components work together to shape epithelial tissues. In Caenorhabditis elegans, external epithelia develop in the context of an ill-defined ZP-containing aECM that precedes secretion of the collagenous cuticle. C. elegans has 43 genes that encode at least 65 unique ZP proteins, and we show that some of these comprise distinct precuticle aECMs in the embryo. Previously, the nidogen- and EGF-domain protein DEX-1 was shown to anchor dendrites to the C. elegans nose tip in concert with the ZP protein DYF-7 Here, we identified a new, strong loss-of-function allele of dex-1, cs201dex-1 mutants die as L1 larvae and have a variety of tissue distortion phenotypes, including excretory defects, pharyngeal ingression, alae defects, and a short and fat body shape, that strongly resemble those of genes encoding ZP proteins. DEX-1 localizes to ZP-containing aECMs in the tissues that show defects in dex-1 mutants. Our studies suggest that DEX-1 is a component of multiple distinct embryonic aECMs that shape developing epithelia, and a potential partner of multiple ZP proteins.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Animals , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/metabolism , Cell Differentiation , Epithelial Cells/cytology , Loss of Function Mutation , Morphogenesis
14.
Genetics ; 211(1): 169-183, 2019 01.
Article in English | MEDLINE | ID: mdl-30409788

ABSTRACT

Phenotypic plasticity is a critical component of an organism's ability to thrive in a changing environment. The free-living nematode Caenorhabditis elegans adapts to unfavorable environmental conditions by pausing reproductive development and entering a stress-resistant larval stage known as dauer. The transition into dauer is marked by vast morphological changes, including remodeling of epidermis, neurons, and muscle. Although many of these dauer-specific traits have been described, the molecular basis of dauer-specific remodeling is still poorly understood. Here we show that the nidogen domain-containing protein DEX-1 facilitates stage-specific tissue remodeling during dauer morphogenesis. DEX-1 was previously shown to regulate sensory dendrite formation during embryogenesis. We find that DEX-1 is also required for proper remodeling of the stem cell-like epidermal seam cells. dex-1 mutant dauers lack distinct lateral cuticular alae during dauer and have increased sensitivity to sodium dodecyl sulfate. Furthermore, we find that DEX-1 is required for proper dauer mobility. We show that DEX-1 is secreted from the seam cells during dauer, but acts locally in a cell-autonomous manner. We find that dex-1 expression during dauer is regulated through DAF-16/FOXO-mediated transcriptional activation. Finally, we show that dex-1 acts with a family of zona pellucida domain-encoding genes to regulate dauer-specific epidermal remodeling. Taken together, our data indicate that DEX-1 is an extracellular matrix component that plays a central role in C. elegans epidermal remodeling during dauer.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Calcium-Binding Proteins/metabolism , Epidermis/growth & development , Animals , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Calcium-Binding Proteins/genetics , Epidermis/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Larva/genetics , Larva/growth & development , Morphogenesis , Stem Cells/cytology , Stem Cells/metabolism
15.
Genetics ; 207(2): 625-642, 2017 10.
Article in English | MEDLINE | ID: mdl-28842397

ABSTRACT

A lipid and glycoprotein-rich apical extracellular matrix (aECM) or glycocalyx lines exposed membranes in the body, and is particularly important to protect narrow tube integrity. Lipocalins ("fat cups") are small, secreted, cup-shaped proteins that bind and transport lipophilic cargo and are often found in luminal or aECM compartments such as mammalian plasma, urine, or tear film. Although some lipocalins can bind known aECM lipids and/or matrix metalloproteinases, it is not known if and how lipocalins affect aECM structure due to challenges in visualizing the aECM in most systems. Here we show that two Caenorhabditiselegans lipocalins, LPR-1 and LPR-3, have distinct functions in the precuticular glycocalyx of developing external epithelia. LPR-1 moves freely through luminal compartments, while LPR-3 stably localizes to a central layer of the membrane-anchored glycocalyx, adjacent to the transient zona pellucida domain protein LET-653 Like LET-653 and other C. elegans glycocalyx components, these lipocalins are required to maintain the patency of the narrow excretory duct tube, and also affect multiple aspects of later cuticle organization. lpr-1 mutants cannot maintain a continuous excretory duct apical domain and have misshapen cuticle ridges (alae) and abnormal patterns of cuticular surface lipid staining. lpr-3 mutants cannot maintain a passable excretory duct lumen, properly degrade the eggshell, or shed old cuticle during molting, and they lack cuticle barrier function. Based on these phenotypes, we infer that both LPR-1 and LPR-3 are required to build a properly organized aECM, while LPR-3 additionally is needed for aECM clearance and remodeling. The C. elegans glycocalyx provides a powerful system, amenable to both genetic analysis and live imaging, for investigating how lipocalins and lipids affect aECM structure.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Extracellular Matrix/metabolism , Lipocalins/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Epithelial Cells/metabolism , Glycocalyx/genetics , Glycocalyx/metabolism , Lipocalins/genetics , Mucins/genetics , Mucins/metabolism
17.
Circulation ; 106(22): 2793-9, 2002 Nov 26.
Article in English | MEDLINE | ID: mdl-12451005

ABSTRACT

BACKGROUND: We previously proposed that adenosine has mechanism-specific effects on atrial tachycardia (AT), such that adenosine terminates AT attributable to triggered activity, transiently suppresses automatic rhythms, and has no effect on macroreentrant AT. This, however, remains controversial, because other studies have reported that adenosine terminates reentrant AT. To clarify this issue, we used 3D electroanatomic mapping to delineate the tachycardia circuit and thereby determine whether the response to adenosine differentiates focal from macroreentrant AT. METHODS AND RESULTS: We examined the effect of adenosine on 43 ATs in 42 consecutive patients (59+/-15 years of age; 26 female) who received adenosine during tachycardia and whose mechanism of AT was characterized by pharmacological perturbation, entrainment, 3D electroanatomic mapping, and results of radiofrequency ablation. Eight tachycardias were macroreentrant (noncavotricuspid isthmus-dependent), and 35 ATs were focal (either triggered or automatic). Adenosine administered during AT (at doses sufficient to result in AV block) terminated or transiently suppressed focal AT in 33 of 35 cases, whereas 8 of 8 macroreentrant ATs were adenosine insensitive (P<0.001). Twenty-eight of 35 focal ATs were located along the crista terminalis or tricuspid annulus. CONCLUSIONS: The response of AT to adenosine can immediately differentiate atrial tachycardia arising from a focal source from that attributable to macroreentry. This finding can be exploited to facilitate developing a focused, strategic ablative approach at the onset of a procedure.


Subject(s)
Adenosine , Body Surface Potential Mapping , Electrophysiologic Techniques, Cardiac , Heart Atria/physiopathology , Imaging, Three-Dimensional , Tachycardia/diagnosis , Tachycardia/physiopathology , Adrenergic beta-Agonists/administration & dosage , Body Surface Potential Mapping/methods , Cardiac Pacing, Artificial , Catheter Ablation , Diagnosis, Differential , Ebstein Anomaly/diagnosis , Ebstein Anomaly/physiopathology , Electrocardiography , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Tachycardia/classification , Tachycardia/surgery , Treatment Outcome
18.
Am J Cardiol ; 110(11): 1631-6, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22921995

ABSTRACT

Conventional pacemaker and implantable cardioverter-defibrillator product labeling currently cautions against exposure to magnetic resonance imaging (MRI). However, there is a growing clinical need for MRI, without an acceptable alternative imaging modality in many patients with cardiac devices. The purpose of this study was to determine the risk of MRI at 1.5 T for patients with cardiac devices by measuring the frequency of device failures and clinically relevant device parameter changes. Data from a single-center retrospective review of 109 patients with pacemakers and implantable cardioverter-defibrillators (the MRI group) who underwent 125 clinically indicated MRI studies were compared to data from a prospective cohort of 50 patients with cardiac devices who did not undergo MRI (the control group). In the MRI group, there were no deaths, device failures requiring generator or lead replacement, induced arrhythmias, losses of capture, or electrical reset episodes. Decreases in battery voltage of ≥0.04 V occurred in 4%, pacing threshold increases of ≥0.5 V in 3%, and pacing lead impedance changes of ≥50 Ω in 6%. Although there were statistically significant differences between the MRI and control groups for the mean change in pacing lead impedance (-6.2 ± 23.9 vs 3.0 ± 22.1 Ω) and left ventricular pacing threshold (-0.1 ± 0.3 vs 0.1 ± 0.2 V), these differences were not clinically important. In conclusion, MRI in patients with cardiac devices resulted in no device or lead failures. A small number of clinically relevant changes in device parameter measurements were noted. However, these changes were similar to those in a control group of patients who did not undergo MRI.


Subject(s)
Arrhythmias, Cardiac/diagnosis , Defibrillators, Implantable , Equipment Failure/statistics & numerical data , Magnetic Resonance Imaging, Cine , Pacemaker, Artificial , Risk Assessment/methods , Aged , Arrhythmias, Cardiac/therapy , Contraindications , Equipment Design , Equipment Safety , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging, Cine/adverse effects , Magnetic Resonance Imaging, Cine/instrumentation , Male , Retrospective Studies , Risk Factors
19.
Toxicol Sci ; 122(2): 361-71, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21693435

ABSTRACT

The loss of tuberin, the tuberous sclerosis-2 (Tsc-2) gene product, is associated with cytoplasmic mislocalization of p27 in uterine leiomyomas derived from Eker rats (Tsc-2(EK/+)) and in human metastatic renal cell carcinoma tissue. Signaling associated with cytoplasmic mislocalization of p27 in renal cancer is relatively unknown. Renal tumors derived from 2,3,5-tris-(glutathion-S-yl)hydroquinone (TGHQ)-treated Tsc-2(EK/+) rats, and null for tuberin, display elevated nuclear and cytosolic p27, with parallel increases in cytosolic cyclin D1 levels. Similar changes are observed in TGHQ-transformed renal epithelial cells derived from Tsc-2(EK/+) rats (QTRRE cells), which, in addition to the cytoplasmic mislocalization of p27 and cyclin D1, exhibit high ERK, B-Raf, and Raf-1 kinase activity. Renal tumor xenografts, derived from subcutaneous injection of QTRRE cells into nude mice, also display increases in cytosolic mislocalization of p27 and cyclin D1. Dibutyryl cAMP and/or phosphodiesterase inhibitors (PIs; pentoxifylline or theophylline) increase Rap1B activation, B-Raf kinase activity, and cytosolic p27/cyclin D1 protein levels in QTRRE cells. Inhibition of Raf kinases with either sorafenib or B-Raf small interfering RNA (siRNA) caused a mitogen-activated protein kinase-mediated downregulation of p27. Moreover, decreases in cyclin D1 were also associated with p27 siRNA knockdown in QTRRE cells. Finally, theophylline-mediated increases in p27 and cyclin D1 were attenuated by sorafenib, which modulated Raf/MEK/ERK signaling. Collectively, these data suggest that the cAMP/Rap1B/B-Raf pathway modulates the expression of p27 and the cytoplasmic mislocalization of p27-cyclin D1 in tuberous sclerosis gene-regulated-renal cancer. Therefore, the loss of tuberin and engagement of the cAMP pathway may independently direct p27-cyclin D1 cytosolic stabilization during renal tumor formation.


Subject(s)
Bucladesine/pharmacology , Carcinoma, Renal Cell/chemically induced , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Kidney Neoplasms/chemically induced , Tuberous Sclerosis/metabolism , Animals , Benzenesulfonates/metabolism , Cell Line , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p27/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cytosol/metabolism , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Glutathione/analogs & derivatives , Glutathione/pharmacology , Humans , Hydroquinones/pharmacology , Male , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Niacinamide/analogs & derivatives , Pentoxifylline/metabolism , Phenylurea Compounds , Phosphodiesterase Inhibitors/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Pyridines/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction , Sorafenib , Theophylline/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
20.
Toxicol Sci ; 124(1): 75-87, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21813464

ABSTRACT

The mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase signaling cascades have been implicated in a number of human cancers. The tumor suppressor gene tuberous sclerosis-2 (Tsc-2) functions as a negative regulator of mTOR. Critical proteins in both pathways are activated following treatment of Eker rats (Tsc-2(EK/+)) with the nephrocarcinogen 2,3,5-tris-(glutathion-S-yl)hydroquinone (TGHQ), which also results in loss of the wild-type allele of Tsc-2 in renal preneoplastic lesions and tumors. Western blot analysis of kidney tumors formed following treatment of Tsc-2(EK/+) rats with TGHQ for 8 months revealed increases in B-Raf, Raf-1, pERK, cyclin D1, 4EBP1, and p-4EBP1-Ser65, -Thr70, and -Thr37/46 expression. Similar changes are observed following TGHQ-mediated transformation of primary renal epithelial cells derived from Tsc-2(EK/+) rats (quinol-thioether rat renal epithelial [QTRRE] cells) that are also null for tuberin. These cells exhibit high ERK, B-Raf, and Raf-1 kinase activity and increased expression of all p-4EBP1s and cyclin D1. Treatment of the QTRRE cells with the Raf kinase inhibitor, sorafenib, or the MEK1/2 kinase inhibitor, PD 98059, produced a significant decrease in the protein expression of all p-4EBP1s and cyclin D1. Following siRNA knockdown of Raf-1, Western blot analysis revealed a significant decrease in Raf-1, cyclin D1, and all p-4EBP1 forms noted above. In contrast, siRNA knockdown of B-Raf resulted in a nominal change in these proteins. The data indicate that Raf-1/MEK/ERK participates in crosstalk with 4EBP1, which represents a novel pathway interaction leading to increased protein synthesis, cell growth, and kidney tumor formation.


Subject(s)
Carcinoma, Renal Cell/chemically induced , Carrier Proteins/metabolism , Cyclin D1/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Glutathione/analogs & derivatives , Hydroquinones/toxicity , Kidney Neoplasms/chemically induced , Phosphoproteins/metabolism , Receptor Cross-Talk , Tumor Suppressor Proteins/genetics , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Carrier Proteins/genetics , Cell Culture Techniques , Cell Line , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cyclin D1/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression Regulation, Neoplastic , Glutathione/toxicity , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Loss of Heterozygosity , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Male , Phosphoproteins/genetics , Protein Biosynthesis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf , RNA, Small Interfering/genetics , Rats , Rats, Mutant Strains , Receptor Cross-Talk/drug effects , Signal Transduction , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL