Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 221
Filter
1.
Differentiation ; 137: 100765, 2024.
Article in English | MEDLINE | ID: mdl-38522217

ABSTRACT

The acquisition of the post-mitotic state is crucial for the execution of many terminally differentiated cell behaviors during organismal development. However, the mechanisms that maintain the post-mitotic state in this context remain poorly understood. To gain insight into these mechanisms, we used the genetically and visually accessible model of C. elegans anchor cell (AC) invasion into the vulval epithelium. The AC is a terminally differentiated uterine cell that normally exits the cell cycle and enters a post-mitotic state before initiating contact between the uterus and vulva through a cell invasion event. Here, we set out to identify the set of negative cell cycle regulators that maintain the AC in this post-mitotic, invasive state. Our findings revealed a critical role for CKI-1 (p21CIP1/p27KIP1) in redundantly maintaining the post-mitotic state of the AC, as loss of CKI-1 in combination with other negative cell cycle regulators-including CKI-2 (p21CIP1/p27KIP1), LIN-35 (pRb/p107/p130), FZR-1 (Cdh1/Hct1), and LIN-23 (ß-TrCP)-resulted in proliferating ACs. Remarkably, time-lapse imaging revealed that these ACs retain their ability to invade. Upon examination of a node in the gene regulatory network controlling AC invasion, we determined that proliferating, invasive ACs do so by maintaining aspects of pro-invasive gene expression. We therefore report that the requirement for a post-mitotic state for invasive cell behavior can be bypassed following direct cell cycle perturbation.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Mitosis , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Mitosis/genetics , Female , Cell Cycle/genetics , Vulva/cytology , Vulva/growth & development , Vulva/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cell Movement/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism
2.
PLoS Biol ; 20(2): e3001317, 2022 02.
Article in English | MEDLINE | ID: mdl-35192608

ABSTRACT

Cell invasion is an initiating event during tumor cell metastasis and an essential process during development. A screen of C. elegans orthologs of genes overexpressed in invasive human melanoma cells has identified several components of the conserved DNA pre-replication complex (pre-RC) as positive regulators of anchor cell (AC) invasion. The pre-RC genes function cell-autonomously in the G1-arrested AC to promote invasion, independently of their role in licensing DNA replication origins in proliferating cells. While the helicase activity of the pre-RC is necessary for AC invasion, the downstream acting DNA replication initiation factors are not required. The pre-RC promotes the invasive fate by regulating the expression of extracellular matrix genes and components of the PI3K signaling pathway. Increasing PI3K pathway activity partially suppressed the AC invasion defects caused by pre-RC depletion, suggesting that the PI3K pathway is one critical pre-RC target. We propose that the pre-RC, or a part of it, acts in the postmitotic AC as a transcriptional regulator that facilitates the switch to an invasive phenotype.


Subject(s)
Caenorhabditis elegans/genetics , Cell Cycle/genetics , Cell Movement/genetics , DNA Replication/genetics , Replication Origin/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Female , Gene Expression Profiling/methods , Gene Ontology , Larva/cytology , Larva/genetics , Larva/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics , Vulva/cytology , Vulva/metabolism
3.
Development ; 149(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34982813

ABSTRACT

During Caenorhabditis elegans vulval development, the uterine anchor cell (AC) first secretes an epidermal growth factor (EGF) to specify the vulval cell fates and then invades the underlying vulval epithelium. By doing so, the AC establishes direct contact with the invaginating primary vulF cells and attaches the developing uterus to the vulva. The signals involved and the exact sequence of events joining these two organs are not fully understood. Using a conditional let-23 EGF receptor (EGFR) allele along with novel microfluidic short- and long-term imaging methods, we discovered a specific function of the EGFR in the AC during vulval lumen morphogenesis. Tissue-specific inactivation of let-23 in the AC resulted in imprecise alignment of the AC with the primary vulval cells, delayed AC invasion and disorganized adherens junctions at the contact site forming between the AC and the dorsal vulF toroid. We propose that EGFR signaling, activated by a reciprocal EGF cue from the primary vulval cells, positions the AC at the vulval midline, guides it during invasion and assembles a cytoskeletal scaffold organizing the adherens junctions that connect the developing uterus to the dorsal vulF toroid. Thus, EGFR signaling in the AC ensures the precise alignment of the two developing organs.


Subject(s)
ErbB Receptors/metabolism , Morphogenesis , Signal Transduction , Vulva/metabolism , Adherens Junctions/metabolism , Animals , Caenorhabditis elegans , Cytoskeleton/metabolism , Epidermal Growth Factor/metabolism , Female , Vulva/cytology , Vulva/growth & development
4.
Genetics ; 218(4)2021 08 09.
Article in English | MEDLINE | ID: mdl-34037773

ABSTRACT

Egg laying in the nematode worm Caenorhabditis elegans is a two-state behavior modulated by internal and external sensory input. We have previously shown that homeostatic feedback of embryo accumulation in the uterus regulates bursting activity of the serotonergic HSN command neurons that sustains the egg-laying active state. How sensory feedback of egg release signals to terminate the egg-laying active state is less understood. We find that Gαo, a conserved Pertussis Toxin-sensitive G protein, signals within HSN to inhibit egg-laying circuit activity and prevent entry into the active state. Gαo signaling hyperpolarizes HSN, reducing HSN Ca2+ activity and input onto the postsynaptic vulval muscles. Loss of inhibitory Gαo signaling uncouples presynaptic HSN activity from a postsynaptic, stretch-dependent homeostat, causing precocious entry into the egg-laying active state when only a few eggs are present in the uterus. Feedback of vulval opening and egg release activates the uv1 neuroendocrine cells which release NLP-7 neuropeptides which signal to inhibit egg laying through Gαo-independent mechanisms in the HSNs and Gαo-dependent mechanisms in cells other than the HSNs. Thus, neuropeptide and inhibitory Gαo signaling maintain a bi-stable state of electrical excitability that dynamically controls circuit activity in response to both external and internal sensory input to drive a two-state behavior output.


Subject(s)
Action Potentials , Caenorhabditis elegans Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Neurons/metabolism , Oviposition , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Calcium/metabolism , Female , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Muscle Contraction , Neurons/physiology , Neuropeptides/genetics , Neuropeptides/metabolism , Signal Transduction , Vulva/cytology , Vulva/innervation , Vulva/physiology
5.
Development ; 148(5)2021 03 05.
Article in English | MEDLINE | ID: mdl-33526581

ABSTRACT

The evolutionarily conserved LIN-2 (CASK)/LIN-7 (Lin7A-C)/LIN-10 (APBA1) complex plays an important role in regulating spatial organization of membrane proteins and signaling components. In Caenorhabditiselegans, the complex is essential for the development of the vulva by promoting the localization of the sole Epidermal growth factor receptor (EGFR) ortholog LET-23 to the basolateral membrane of the vulva precursor cells where it can specify the vulval cell fate. To understand how the LIN-2/7/10 complex regulates receptor localization, we determined its expression and localization during vulva development. We found that LIN-7 colocalizes with LET-23 EGFR at the basolateral membrane, whereas the LIN-2/7/10 complex colocalizes with LET-23 EGFR at cytoplasmic punctae that mostly overlap with the Golgi. Furthermore, LIN-10 recruits LIN-2, which in turn recruits LIN-7. We demonstrate that the complex forms in vivo with a particularly strong interaction and colocalization between LIN-2 and LIN-7, consistent with them forming a subcomplex. Thus, the LIN-2/7/10 complex forms on the Golgi on which it likely targets LET-23 EGFR trafficking to the basolateral membrane rather than functioning as a tether.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , ErbB Receptors/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Vulva/metabolism , Animals , Animals, Genetically Modified/metabolism , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Cell Lineage , Cell Membrane/metabolism , ErbB Receptors/genetics , Female , Helminth Proteins/genetics , Helminth Proteins/metabolism , Larva/metabolism , Membrane Proteins/genetics , Neurons/metabolism , Protein Binding , Stem Cells/cytology , Stem Cells/metabolism , Vulva/cytology , Vulva/growth & development
7.
Int J Mol Sci ; 21(20)2020 Oct 15.
Article in English | MEDLINE | ID: mdl-33076222

ABSTRACT

The C. elegans vulva is an excellent model for the study of developmental biology and cell-cell signaling. The developmental induction of vulval precursor cells (VPCs) to assume the 3°-3°-2°-1°-2°-3° patterning of cell fates occurs with 99.8% accuracy. During C. elegans vulval development, an EGF signal from the anchor cell initiates the activation of RasLET-60 > RafLIN-45 > MEKMEK-2 > ERKMPK-1 signaling cascade to induce the 1° cell. The presumptive 1° cell signals its two neighboring cells via NotchLIN-12 to develop 2° cells. In addition, RasLET-60 switches effectors to RalGEFRGL-1 > RalRAL-1 to promote 2° fate. Shin et al. (2019) showed that RalGEFRGL-1 is a dual-function protein in VPCs fate patterning. RalGEFRGL-1 functions as a scaffold for PDKPDK-1 > AktAKT-1/2 modulatory signaling to promote 1° fate in addition to propagating the RasLET-60 modulatory signal through RalRAL-1 to promote 2° fate. The deletion of RalGEFRGL-1 increases the frequency of VPC patterning errors 15-fold compared to the wild-type control. We speculate that RalGEFRGL-1 represents an "insulated switch", whereby the promotion of one signaling activity curtails the promotion of the opposing activity. This property might increase the impact of the switch on fidelity more than two separately encoded proteins could. Understanding how developmental fidelity is controlled will help us to better understand the origins of cancer and birth defects, which occur in part due to the misspecification of cell fates.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Gene Expression Regulation, Developmental , Signal Transduction , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Cell Lineage , Female , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Vulva/cytology , Vulva/growth & development , Vulva/metabolism , ral GTP-Binding Proteins/genetics , ral GTP-Binding Proteins/metabolism , ras Proteins/genetics , ras Proteins/metabolism
8.
Development ; 147(11)2020 06 04.
Article in English | MEDLINE | ID: mdl-32439759

ABSTRACT

The anchor cell (AC) in C. elegans secretes an epidermal growth factor (EGF) homolog that induces adjacent vulval precursor cells (VPCs) to differentiate. The EGF receptor in the nearest VPC sequesters the limiting EGF amounts released by the AC to prevent EGF from spreading to distal VPCs. Here, we show that not only EGFR localization in the VPCs but also EGF polarity in the AC is necessary for robust fate specification. The AC secretes EGF in a directional manner towards the nearest VPC. Loss of AC polarity causes signal spreading and, when combined with MAPK pathway hyperactivation, the ectopic induction of distal VPCs. In a screen for genes preventing distal VPC induction, we identified sra-9 and nlp-26 as genes specifically required for polarized EGF secretion. sra-9(lf) and nlp-26(lf) mutants exhibit errors in vulval fate specification, reduced precision in VPC to AC alignment and increased variability in MAPK activation. sra-9 encodes a seven-pass transmembrane receptor acting in the AC and nlp-26 a neuropeptide-like protein expressed in the VPCs. SRA-9 and NLP-26 may transduce a feedback signal to channel EGF secretion towards the nearest VPC.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Epidermal Growth Factor/metabolism , Vulva/metabolism , Animals , Animals, Genetically Modified/growth & development , Animals, Genetically Modified/metabolism , CRISPR-Cas Systems/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Epidermal Growth Factor/antagonists & inhibitors , Epidermal Growth Factor/genetics , ErbB Receptors/metabolism , Female , Gene Editing , Larva/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis , Netrins/genetics , Netrins/metabolism , RNA Interference , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Vulva/cytology , Vulva/growth & development , ras GTPase-Activating Proteins/antagonists & inhibitors , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
9.
Dev Biol ; 461(2): 110-123, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32032579

ABSTRACT

During development, cell fate decisions are often highly stochastic, but with the frequency of the different possible fates tightly controlled. To understand how signaling networks control the cell fate frequency of such random decisions, we studied the stochastic decision of the Caenorhabditis elegans P3.p cell to either fuse to the hypodermis or assume vulva precursor cell fate. Using time-lapse microscopy to measure the single-cell dynamics of two key inhibitors of cell fusion, the Hox gene LIN-39 and Wnt signaling through the ß-catenin BAR-1, we uncovered significant variability in the dynamics of LIN-39 and BAR-1 levels. Most strikingly, we observed that BAR-1 accumulated in a single, 1-4 â€‹h pulse at the time of the P3.p cell fate decision, with strong variability both in pulse slope and time of pulse onset. We found that the time of BAR-1 pulse onset was delayed relative to the time of cell fusion in mutants with low cell fusion frequency, linking BAR-1 pulse timing to cell fate outcome. Overall, a model emerged where animal-to-animal variability in LIN-39 levels and BAR-1 pulse dynamics biases cell fate by modulating their absolute level at the time cell fusion is induced. Our results highlight that timing of cell signaling dynamics, rather than its average level or amplitude, could play an instructive role in determining cell fate.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , beta Catenin/metabolism , Animals , CRISPR-Cas Systems , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/biosynthesis , Caenorhabditis elegans Proteins/genetics , Cell Differentiation , Cell Fusion , Cell Lineage , Cytoskeletal Proteins/metabolism , Female , Gene Expression Regulation, Developmental , Genotype , Homeodomain Proteins/metabolism , In Situ Hybridization, Fluorescence , Integumentary System/anatomy & histology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Recombinant Fusion Proteins/metabolism , Single-Cell Analysis , Stochastic Processes , Time-Lapse Imaging , Vulva/cytology , Wnt Signaling Pathway
10.
Forensic Sci Int Genet ; 43: 102153, 2019 11.
Article in English | MEDLINE | ID: mdl-31505370

ABSTRACT

Data from all sexual assault cases analysed at the Section of Forensic Biology at Oslo University Hospital in the period 2013-2015 were reviewed to study transfer and persistence of cells deposited on the body. Data were recorded on detection of both sperm and epithelial cells. The final dataset consist of 2141 samples from 765 cases. In this study "positive findings" refer to evidence to support the proposition that the DNA profile was contributed by the POI and do not only correspond to detection of cell type, e.g. sperm cells. Positive findings from analysis of sperm cells could be detected in samples collected up to 72 h after deposition, and was less frequently detected in oral swabs were the longest observed persistence time was 12 h. Positive findings from analysis of epithelial cells were observed up to 43 h after deposition. A high success rate was observed from penile swabs collected within 24 h of the incidence demonstrating the importance of collecting and analysing such samples in cases where no semen is detected.


Subject(s)
DNA Fingerprinting , DNA/isolation & purification , Epithelial Cells/cytology , Sex Offenses , Spermatozoa/cytology , Epithelial Cells/chemistry , Female , Forensic Genetics/methods , Humans , Male , Mouth/cytology , Polymerase Chain Reaction , Rectum/cytology , Retrospective Studies , Skin/cytology , Specimen Handling , Spermatozoa/chemistry , Time Factors , Vagina/cytology , Vulva/cytology
11.
G3 (Bethesda) ; 9(11): 3567-3574, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31519743

ABSTRACT

Genetic analysis of LIN-12/Notch signaling in C. elegans has provided many insights into human biology. Activating missense mutations in the Negative Regulatory Region (NRR) of the ectodomain of LIN-12/Notch were first described in C. elegans, and similar mutations in human Notch were later found to cause T-cell acute lymphoblastic leukemia (T-ALL). The ubiquitin ligase sel-10/Fbw7 is the prototype of a conserved negative regulator of lin-12/Notch that was first defined by loss-of-function mutations that enhance lin-12 NRR-missense activity in C. elegans, and then demonstrated to regulate Notch activity in mammalian cells and to be a bona fide tumor suppressor in T-ALL. Here, we report the results of an RNAi screen of 248 C. elegans protein kinase-encoding genes with human orthologs for enhancement of a weakly activating NRR-missense mutation of lin-12 in the Vulval Precursor Cells. We identified, and validated, thirteen kinase genes whose loss led to increase lin-12 activity; eleven of these genes have never been implicated previously in regulating Notch activity in any system. Depleting the activity of five kinase genes (cdk-8, wnk-1, kin-3, hpo-11, and mig-15) also significantly enhanced the activity of a transgene in which heterologous sequences drive expression of the untethered intracellular domain of LIN-12, suggesting that they increase the activity or stability of the signal-transducing form of LIN-12/Notch. Precedents set by other regulators of lin-12/Notch defined through genetic interactions in C. elegans suggest that this new set of genes may include negative regulators that are functionally relevant to mammalian development and cancer.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Receptors, Notch/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Animals , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Female , Mutation, Missense , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Double-Stranded/metabolism , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/genetics , Signal Transduction/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Vulva/cytology , Vulva/metabolism
12.
PLoS Genet ; 15(5): e1008056, 2019 05.
Article in English | MEDLINE | ID: mdl-31086367

ABSTRACT

The six C. elegans vulval precursor cells (VPCs) are induced to form the 3°-3°-2°-1°-2°-3° pattern of cell fates with high fidelity. In response to EGF signal, the LET-60/Ras-LIN-45/Raf-MEK-2/MEK-MPK-1/ERK canonical MAP kinase cascade is necessary to induce 1° fate and synthesis of DSL ligands for the lateral Notch signal. In turn, LIN-12/Notch receptor is necessary to induce neighboring cells to become 2°. We previously showed that, in response to graded EGF signal, the modulatory LET-60/Ras-RGL-1/RalGEF-RAL-1/Ral signal promotes 2° fate in support of LIN-12. In this study, we identify two key differences between RGL-1 and RAL-1. First, deletion of RGL-1 confers no overt developmental defects, while previous studies showed RAL-1 to be essential for viability and fertility. From this observation, we hypothesize that the essential functions of RAL-1 are independent of upstream activation. Second, RGL-1 plays opposing and genetically separable roles in VPC fate patterning. RGL-1 promotes 2° fate via canonical GEF-dependent activation of RAL-1. Conversely, RGL-1 promotes 1° fate via a non-canonical GEF-independent activity. Our genetic epistasis experiments are consistent with RGL-1 functioning in the modulatory 1°-promoting AGE-1/PI3-Kinase-PDK-1-AKT-1 cascade. Additionally, animals lacking RGL-1 experience 15-fold higher rates of VPC patterning errors compared to the wild type. Yet VPC patterning in RGL-1 deletion mutants is not more sensitive to environmental perturbations. We propose that RGL-1 functions to orchestrate opposing 1°- and 2°-promoting modulatory cascades to decrease developmental stochasticity. We speculate that such switches are broadly conserved but mostly masked by paralog redundancy or essential functions.


Subject(s)
Caenorhabditis elegans/genetics , Epidermal Growth Factor/genetics , Gene Expression Regulation, Developmental , Guanine Nucleotide Exchange Factors/genetics , Vulva/metabolism , Animals , Body Patterning/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Epidermal Growth Factor/metabolism , Epistasis, Genetic , Female , Fertility/genetics , Guanine Nucleotide Exchange Factors/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Vulva/cytology , Vulva/growth & development , raf Kinases/genetics , raf Kinases/metabolism , ral GTP-Binding Proteins/genetics , ral GTP-Binding Proteins/metabolism , ras Proteins/genetics , ras Proteins/metabolism
13.
Dev Cell ; 42(3): 271-285.e3, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28787593

ABSTRACT

During epithelial tube morphogenesis, linear arrays of cells are converted into tubular structures through actomyosin-generated intracellular forces that induce tissue invagination and lumen formation. We have investigated lumen morphogenesis in the C. elegans vulva. The first discernible event initiating lumen formation is the apical constriction of the two innermost primary cells (VulF). The VulF cells thereafter constrict their lateral membranes along the apicobasal axis to extend the lumen dorsally. Lateral, but not apical, VulF constriction requires the prior invasion of the anchor cell (AC). The invading AC extends actin-rich protrusions toward VulF, resulting in the formation of a direct AC-VulF interface. The recruitment of the F-BAR-domain protein TOCA-1 to the AC-VulF interface induces the accumulation of force-generating actomyosin, causing a switch from apical to lateral membrane constriction and the dorsal extension of the lumen. Invasive cells may induce shape changes in adjacent cells to penetrate their target tissues.


Subject(s)
Caenorhabditis elegans/embryology , Morphogenesis , Vulva/embryology , Actomyosin/genetics , Actomyosin/metabolism , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Movement , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Female , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Vulva/cytology , Vulva/metabolism
14.
Dev Cell ; 42(5): 542-553.e4, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28826819

ABSTRACT

Kinase translocation reporters (KTRs) are genetically encoded fluorescent activity sensors that convert kinase activity into a nucleocytoplasmic shuttling equilibrium for visualizing single-cell signaling dynamics. Here, we adapt the first-generation KTR for extracellular signal-regulated kinase (ERK) to allow easy implementation in vivo. This sensor, "ERK-nKTR," allows quantitative and qualitative assessment of ERK activity by analysis of individual nuclei and faithfully reports ERK activity during development and neural function in diverse cell contexts in Caenorhabditis elegans. Analysis of ERK activity over time in the vulval precursor cells, a well-characterized paradigm of epidermal growth factor receptor (EGFR)-Ras-ERK signaling, has identified dynamic features not evident from analysis of developmental endpoints alone, including pulsatile frequency-modulated signaling associated with proximity to the EGF source. The toolkit described here will facilitate studies of ERK signaling in other C. elegans contexts, and the design features will enable implementation of this technology in other multicellular organisms.


Subject(s)
Biosensing Techniques/methods , Caenorhabditis elegans/cytology , Caenorhabditis elegans/enzymology , Cell Lineage , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Animals , Cell Line , Cell Movement , Cell Nucleus/metabolism , Female , Genes, Reporter , Germ Cells/cytology , Mammals , Mutation/genetics , Myoblasts/cytology , Neurons/cytology , Phosphorylation , Reproducibility of Results , Stem Cells/cytology , Stem Cells/metabolism , Subcellular Fractions/metabolism , Vulva/cytology
15.
Prog. obstet. ginecol. (Ed. impr.) ; 60(4): 347-350, jul.-ago. 2017. tab, ilus
Article in Spanish | IBECS | ID: ibc-165800

ABSTRACT

Presentamos el caso de una paciente adolescente, estudiada desde los 11 años por episodios de úlceras genitales asociados a aftas bucales. Tras llevar a cabo un complejo proceso de diagnóstico diferencial, se descartó cualquier tipo de agente causal y se etiquetó el cuadro como una aftosis compleja idiopática, que se resolvió espontáneamente aplicando tratamiento sintomático. A pesar de haberse descartado por el momento una enfermedad de Behçet, la paciente es controlada periódicamente, dada la dificultad diagnóstica de esta entidad sobre todo durante los primeros años de evolución (AU)


We report the case of a teenage patient studied since she was 11 years old by episodes of genital ulcers associated with aphthous stomatitis. After carrying out a complex process of differential diagnosis, any kind of causal agent was ruled out and we got the diagnostic of complex aphthosis idiopathic, which resolved spontaneously applying symptomatic treatment. Although having ruled out Behcet’s disease, the patient is monitored periodically, given the difficulty in diagnosing this entity especially during the first years of evolution (AU)


Subject(s)
Humans , Female , Adolescent , Stomatitis, Aphthous/diagnosis , Stomatitis, Aphthous/drug therapy , Recurrence , Behcet Syndrome/diagnosis , Behcet Syndrome/drug therapy , Vulva/injuries , Biopsy , Diagnosis, Differential , Vulva/cytology , Anti-Bacterial Agents/therapeutic use , Vulva , Vulvar Diseases/diagnosis , Ciprofloxacin/therapeutic use , Anesthetics/therapeutic use , Anti-Infective Agents, Local/therapeutic use
16.
Cell Syst ; 4(2): 219-230.e6, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28215526

ABSTRACT

It is a fundamental open question as to how embryos develop into complex adult organisms with astounding reproducibility, particularly because cells are inherently variable on the molecular level. During C. elegans vulva induction, the anchor cell induces cell fate in the vulva precursor cells in a distance-dependent manner. Surprisingly, we found that initial anchor cell position was highly variable and caused variability in cell fate induction. However, we observed that vulva induction was "canalized," i.e., the variability in anchor cell position and cell fate was progressively reduced, resulting in an invariant spatial pattern of cell fates at the end of induction. To understand the mechanism of canalization, we quantified induction dynamics as a function of anchor cell position during the canalization process. Our experiments, combined with mathematical modeling, showed that canalization required a specific combination of long-range induction, lateral inhibition, and cell migration that is also found in other developmental systems.


Subject(s)
Caenorhabditis elegans/genetics , Vulva/metabolism , Animals , Body Patterning/physiology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Cell Movement , Embryonic Induction , Female , Ligands , Models, Theoretical , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Signal Transduction , Vulva/cytology , Vulva/embryology , Vulva/growth & development
17.
Dev Cell ; 40(2): 202-214, 2017 01 23.
Article in English | MEDLINE | ID: mdl-28041904

ABSTRACT

Long-term studies of Caenorhabditis elegans larval development traditionally require tedious manual observations because larvae must move to develop, and existing immobilization techniques either perturb development or are unsuited for young larvae. Here, we present a simple microfluidic device to simultaneously follow development of ten C. elegans larvae at high spatiotemporal resolution from hatching to adulthood (∼3 days). Animals grown in microchambers are periodically immobilized by compression to allow high-quality imaging of even weak fluorescence signals. Using the device, we obtain cell-cycle statistics for C. elegans vulval development, a paradigm for organogenesis. We combine Nomarski and multichannel fluorescence microscopy to study processes such as cell-fate specification, cell death, and transdifferentiation throughout post-embryonic development. Finally, we generate time-lapse movies of complex neural arborization through automated image registration. Our technique opens the door to quantitative analysis of time-dependent phenomena governing cellular behavior during C. elegans larval development.


Subject(s)
Caenorhabditis elegans/metabolism , Imaging, Three-Dimensional , Microfluidics/methods , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Cell Cycle , Cell Death , Cell Division , Cell Tracking , Cell Transdifferentiation , Female , Gene Expression Regulation, Developmental , Larva/metabolism , Male , Neurites/metabolism , Time Factors , Time-Lapse Imaging , Vulva/cytology , Vulva/growth & development
18.
PLoS Genet ; 12(9): e1006278, 2016 09.
Article in English | MEDLINE | ID: mdl-27588814

ABSTRACT

Patterning of C. elegans vulval cell fates relies on inductive signaling. In this induction event, a single cell, the gonadal anchor cell, secretes LIN-3/EGF and induces three out of six competent precursor cells to acquire a vulval fate. We previously showed that this developmental system is robust to a four-fold variation in lin-3/EGF genetic dose. Here using single-molecule FISH, we find that the mean level of expression of lin-3 in the anchor cell is remarkably conserved. No change in lin-3 expression level could be detected among C. elegans wild isolates and only a low level of change-less than 30%-in the Caenorhabditis genus and in Oscheius tipulae. In C. elegans, lin-3 expression in the anchor cell is known to require three transcription factor binding sites, specifically two E-boxes and a nuclear-hormone-receptor (NHR) binding site. Mutation of any of these three elements in C. elegans results in a dramatic decrease in lin-3 expression. Yet only a single E-box is found in the Drosophilae supergroup of Caenorhabditis species, including C. angaria, while the NHR-binding site likely only evolved at the base of the Elegans group. We find that a transgene from C. angaria bearing a single E-box is sufficient for normal expression in C. elegans. Even a short 58 bp cis-regulatory fragment from C. angaria with this single E-box is able to replace the three transcription factor binding sites at the endogenous C. elegans lin-3 locus, resulting in the wild-type expression level. Thus, regulatory evolution occurring in cis within a 58 bp lin-3 fragment, results in a strict requirement for the NHR binding site and a second E-box in C. elegans. This single-cell, single-molecule, quantitative and functional evo-devo study demonstrates that conserved expression levels can hide extensive change in cis-regulatory site requirements and highlights the evolution of new cis-regulatory elements required for cell-specific gene expression.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , E-Box Elements/genetics , Epidermal Growth Factor/genetics , Vulva/growth & development , Animals , Body Patterning/genetics , Caenorhabditis elegans/growth & development , Cell Differentiation/genetics , Drosophila/genetics , Drosophila/growth & development , Female , Gene Expression Regulation, Developmental , Protein Binding/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Single-Cell Analysis , Vulva/cytology
19.
Dev Biol ; 416(1): 123-135, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27288708

ABSTRACT

How cells coordinate their spatial positioning through intercellular signaling events is poorly understood. Here we address this topic using Caenorhabditis elegans vulval patterning during which hypodermal vulval precursor cells (VPCs) adopt distinct cell fates determined by their relative positions to the gonadal anchor cell (AC). LIN-3/EGF signaling by the AC induces the central VPC, P6.p, to adopt a 1° vulval fate. Exact alignment of AC and VPCs is thus critical for correct fate patterning, yet, as we show here, the initial AC-VPC positioning is both highly variable and asymmetric among individuals, with AC and P6.p only becoming aligned at the early L3 stage. Cell ablations and mutant analysis indicate that VPCs, most prominently 1° cells, move towards the AC. We identify AC-released LIN-3/EGF as a major attractive signal, which therefore plays a dual role in vulval patterning (cell alignment and fate induction). Additionally, compromising Wnt pathway components also induces AC-VPC alignment errors, with loss of posterior Wnt signaling increasing stochastic vulval centering on P5.p. Our results illustrate how intercellular signaling reduces initial spatial variability in cell positioning to generate reproducible interactions across tissues.


Subject(s)
Embryonic Induction , Signal Transduction , Stem Cells , Vulva/embryology , Animals , Body Patterning , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Cell Lineage , Cell Movement , Female , Vulva/cytology , Wnt Proteins/metabolism
20.
Arch Gynecol Obstet ; 294(3): 549-54, 2016 09.
Article in English | MEDLINE | ID: mdl-27067433

ABSTRACT

OBJECTIVES: To determine whether there is any new anatomical structure present within the labia majora. STUDY DESIGN: A case serial study was executed on eleven consecutive fresh human female cadavers. Stratum-by-stratum dissections of the labia majora were performed. Twenty-two anatomic dissections of labia majora were completed. Eosin and Hematoxylin agents were used to stain newly discovered adipose sac's tissues of the labia majora and the cylinder-like structures, which cover condensed adipose tissues. The histology of these two structures was compared. RESULTS: All dissected labia majora demonstrated the presence of the anatomic existence of the adipose sac structure. Just under the dermis of the labia majora, the adipose sac was located, which was filled with lobules containing condensed fatty tissues in the form of cylinders. The histological investigation established that the well-organized fibro-connective-adipose tissues represented the adipose sac. The absence of descriptions of the adipose sac within the labia majora in traditional anatomic and gynecologic textbooks was noted. CONCLUSIONS: In this study group, the newly discovered adipose sac is consistently present within the anatomical structure of the labia majora. The well-organized fibro-connective-adipose tissue represents microscopic characteristic features of the adipose sac.


Subject(s)
Adipose Tissue/anatomy & histology , Vulva/anatomy & histology , Adipose Tissue/cytology , Adult , Aged , Aged, 80 and over , Biomedical Research , Cadaver , Female , Humans , Middle Aged , Vulva/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...