Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Dev Dyn ; 252(9): 1149-1161, 2023 09.
Article in English | MEDLINE | ID: mdl-37204056

ABSTRACT

BACKGROUND: p24/transmembrane Emp24 domain (TMED) proteins are a set of evolutionarily conserved, single pass transmembrane proteins that have been shown to facilitate protein secretion and selection of cargo proteins to transport vesicles in the cellular secretion pathway. However, their functions in animal development are incompletely understood. RESULTS: The C. elegans genome encodes eight identified TMED genes, with at least one member from each defined subfamily (α, ß, γ, δ). TMED gene mutants exhibit a shared set of defects in embryonic viability, animal movement, and vulval morphology. Two γ subfamily genes, tmed-1 and tmed-3, exhibit the ability to compensate for each other, as defects in movement and vulva morphology are only apparent in double mutants. TMED mutants also exhibit a delay in breakdown of basement membrane during vulva development. CONCLUSIONS: The results establish a genetic and experimental framework for the study of TMED gene function in C. elegans, and argue that a functional protein from each subfamily is important for a shared set of developmental processes. A specific function for TMED genes is to facilitate breakdown of the basement membrane between the somatic gonad and vulval epithelial cells, suggesting a role for TMED proteins in tissue reorganization during animal development.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Female , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Membrane Proteins/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Phenotype , Biomarkers , Vulva/metabolism
2.
Genes Dev ; 29(16): 1707-20, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26302789

ABSTRACT

Inactivation of phosphatase and tensin homology deleted on chromosome 10 (PTEN) is linked to increased PI3K-AKT signaling, enhanced organismal growth, and cancer development. Here we generated and analyzed Pten knock-in mice harboring a C2 domain missense mutation at phenylalanine 341 (Pten(FV)), found in human cancer. Despite having reduced levels of PTEN protein, homozygous Pten(FV/FV) embryos have intact AKT signaling, develop normally, and are carried to term. Heterozygous Pten(FV/+) mice develop carcinoma in the thymus, stomach, adrenal medulla, and mammary gland but not in other organs typically sensitive to Pten deficiency, including the thyroid, prostate, and uterus. Progression to carcinoma in sensitive organs ensues in the absence of overt AKT activation. Carcinoma in the uterus, a cancer-resistant organ, requires a second clonal event associated with the spontaneous activation of AKT and downstream signaling. In summary, this PTEN noncatalytic missense mutation exposes a core tumor suppressor function distinct from inhibition of canonical AKT signaling that predisposes to organ-selective cancer development in vivo.


Subject(s)
Carcinoma/genetics , Mutation, Missense/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Signal Transduction , Animals , Carcinoma/enzymology , Carcinoma/physiopathology , Cell Nucleus/metabolism , Cells, Cultured , Embryo, Mammalian , Enzyme Activation , Female , Gene Knock-In Techniques , Mice , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Protein Stability
3.
Mol Biol Evol ; 37(5): 1350-1361, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31960924

ABSTRACT

Comparative genomic sequence analysis has found that the genes for many chromatin-associated proteins are poorly conserved, but the biological consequences of these sequence changes are not understood. Here, we show that four genes identified for an Inappropriate Vulval cell Proliferation (ivp) phenotype in the nematode Caenorhabditis briggsae exhibit distinct functions and genetic interactions when compared with their orthologs in C. elegans. Specifically, we show that the four C. briggsae ivp genes encode the noncanonical histone HTZ-1/H2A.z and three nematode-specific proteins predicted to function in the nucleus. The mutants exhibit ectopic vulval precursor cell proliferation (the multivulva [Muv] phenotype) due to inappropriate expression of the lin-3/EGF gene, and RNAseq analysis suggests a broad role for these ivp genes in transcriptional repression. Importantly, although the C. briggsae phenotypes have parallels with those seen in the C. elegans synMuv system, except for the highly conserved HTZ-1/H2A.z, comparable mutations in C. elegans ivp orthologs do not exhibit synMuv gene interactions or phenotypes. These results demonstrate the evolutionary changes that can underlie conserved biological outputs and argue that proteins critical to repress inappropriate expression from the genome participate in a rapidly evolving functional landscape.


Subject(s)
Caenorhabditis/genetics , Evolution, Molecular , Gene Expression Regulation, Developmental , Animals , Caenorhabditis/growth & development , Caenorhabditis/metabolism , Female , Histones/metabolism , Nuclear Proteins/genetics , Vulva/growth & development
4.
J Theor Biol ; 517: 110596, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33508328

ABSTRACT

The development of multicellular organisms relies on correct patterns of cell fates to produce functional tissues in the mature organism. A commonly observed developmental pattern consists of alternating cell fates, where neighboring cells take on distinct cell fates characterized by contrasting gene and protein expression levels, and this cell fate pattern repeats over two or more cells. The patterns produced by these fate decisions are regulated by a small number of highly conserved signaling networks, some of which are mediated by long range diffusible signals and others mediated by local contact-dependent signals. However, it is not completely understood how local and long range signals associated with these networks interact to produce fate patterns that are both robust and flexible. Here we analyze mathematical models to investigate the patterning of cell fates in an array of cells, focusing on a two cell repeating pattern. Bifurcation analysis of a multicellular ODE model, where we consider the cells as discrete compartments, suggests that cells must balance sensitivity to external signals with robustness to perturbations. To focus on the patterning dynamics close to the bifurcation point, we derive a continuum PDE model that integrates local and long range signaling. For those cells with dynamics close to the bifurcation point, sensitivity to long range signals determines how far a pattern extends in space, while the number of local signaling connections determines the type of pattern produced. This investigation provides a general framework for understanding developmental patterning, and how both long range and local signals play a role in generating features observed across biology, such as species differences in nematode vulval development and insect bristle patterning, as well as medically relevant processes such as control of stem cell fate in the intestinal crypt.


Subject(s)
Signal Transduction , Stem Cells , Body Patterning , Cell Differentiation
5.
Dev Biol ; 444(2): 71-82, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30336114

ABSTRACT

FACT (facilitates chromatin transcription) is a histone chaperone complex important in genomic processes including transcription, DNA replication, and DNA repair. FACT is composed of two proteins, SSRP1 and SPT16, which are highly conserved across eukaryotes. While the mechanisms for FACT in nucleosome reorganization and its relationship to DNA processes is well established, how these roles impact coordination in multicellular animal development are less well understood. Here we characterize the genes encoding FACT complex proteins in the nematode C. elegans. We show that whereas C. elegans includes one SPT16 gene (spt-16), two genes (hmg-3 and hmg-4) encode SSRP1 proteins. Depletion of FACT complex genes interferes with embryonic cell division and cell cycle timing generally, with anterior pharynx development especially sensitive to these defects. hmg-3 and hmg-4 exhibit redundancy for these maternally-provided embryonic functions, but are each uniquely required zygotically for normal germline development. This work provides a framework to study FACT gene function in developmental processes, and identifies that distinct functional requirements for gene duplicates can be manifest within a single tissue.


Subject(s)
DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Carrier Proteins , Cell Cycle , Cell Cycle Proteins/metabolism , Chromatin , DNA Repair , DNA Replication , Genes, Duplicate/genetics , Genes, Duplicate/physiology , High Mobility Group Proteins/genetics , Histones/metabolism , Nucleosomes , Transcription Factors/metabolism
6.
Dev Genes Evol ; 227(3): 213-218, 2017 06.
Article in English | MEDLINE | ID: mdl-28220250

ABSTRACT

Normal vulval development in the nematode Caenorhabditis briggsae is identical to that in the related Caenorhabditis elegans. However, several experiments suggest that there are differences between the two species with respect to the contribution of EGF/Ras signaling. To investigate these differences genetically, we have characterized a C. briggsae mutant strain that phenocopies the effect observed when C. briggsae animals are treated with U0126, an inhibitor of the EGF pathway component MEK. We identify that the gene affected in the mutant strain is Cbr-sur-2, which encodes a MED23 mediator complex protein that acts downstream of EGF signaling in C. elegans and other organisms, such as mammals. When Cbr-sur-2 and Cel-sur-2 mutants are compared, we find that the production of additional vulval cells from P5.p and P7.p in C. elegans is dependent on proper development of P6.p, while C. briggsae does not have a similar requirement. Combined chemical and genetic interference with the EGF pathway completely eliminates vulval development in C. elegans but not in C. briggsae. Our results provide genetic evidence for the differing requirements for EGF signaling in the two species.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/growth & development , Caenorhabditis/growth & development , Transcription Factors/metabolism , Animals , Caenorhabditis/classification , Epidermal Growth Factor/metabolism , Female , Signal Transduction , Vulva/growth & development
7.
Evol Dev ; 17(1): 34-48, 2015.
Article in English | MEDLINE | ID: mdl-25627712

ABSTRACT

Studies of vulval development in the nematode C. elegans have identified many genes that are involved in cell division and differentiation processes. Some of these encode components of conserved signal transduction pathways mediated by EGF, Notch, and Wnt. To understand how developmental mechanisms change during evolution, we are doing a comparative analysis of vulva formation in C. briggsae, a species that is closely related to C. elegans. Here, we report 14 mutations in 7 Multivulva (Muv) genes in C. briggsae that inhibit inappropriate division of vulval precursors. We have developed a new efficient and cost-effective gene mapping method to localize Muv mutations to small genetic intervals on chromosomes, thus facilitating cloning and functional studies. We demonstrate the utility of our method by determining molecular identities of three of the Muv genes that include orthologs of Cel-lin-1 (ETS) and Cel-lin-31 (Winged-Helix) of the EGF-Ras pathway and Cel-pry-1 (Axin), of the Wnt pathway. The remaining four genes reside in regions that lack orthologs of known C. elegans Muv genes. Inhibitor studies demonstrate that the Muv phenotype of all four new genes is dependent on the activity of the EGF pathway kinase, MEK. One of these, Cbr-lin(gu167), shows modest increase in the expression of Cbr-lin-3/EGF compared to wild type. These results argue that while Cbr-lin(gu167) may act upstream of Cbr-lin-3/EGF, the other three genes influence the EGF pathway downstream or in parallel to Cbr-lin-3. Overall, our findings demonstrate that the genetic program underlying a conserved developmental process includes both conserved and divergent functional contributions.


Subject(s)
Caenorhabditis/embryology , Caenorhabditis/genetics , Epidermal Growth Factor/metabolism , Signal Transduction , Animals , Caenorhabditis/classification , Caenorhabditis/metabolism , Female , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Vulva/cytology , Vulva/embryology , Vulva/metabolism
8.
PLoS Genet ; 7(7): e1002174, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21779179

ABSTRACT

The nematode Caenorhabditis briggsae is an emerging model organism that allows evolutionary comparisons with C. elegans and exploration of its own unique biological attributes. To produce a high-resolution C. briggsae recombination map, recombinant inbred lines were generated from reciprocal crosses between two strains and genotyped at over 1,000 loci. A second set of recombinant inbred lines involving a third strain was also genotyped at lower resolution. The resulting recombination maps exhibit discrete domains of high and low recombination, as in C. elegans, indicating these are a general feature of Caenorhabditis species. The proportion of a chromosome's physical size occupied by the central, low-recombination domain is highly correlated between species. However, the C. briggsae intra-species comparison reveals striking variation in the distribution of recombination between domains. Hybrid lines made with the more divergent pair of strains also exhibit pervasive marker transmission ratio distortion, evidence of selection acting on hybrid genotypes. The strongest effect, on chromosome III, is explained by a developmental delay phenotype exhibited by some hybrid F2 animals. In addition, on chromosomes IV and V, cross direction-specific biases towards one parental genotype suggest the existence of cytonuclear epistatic interactions. These interactions are discussed in relation to surprising mitochondrial genome polymorphism in C. briggsae, evidence that the two strains diverged in allopatry, the potential for local adaptation, and the evolution of Dobzhansky-Muller incompatibilities. The genetic and genomic resources resulting from this work will support future efforts to understand inter-strain divergence as well as facilitate studies of gene function, natural variation, and the evolution of recombination in Caenorhabditis nematodes.


Subject(s)
Caenorhabditis/genetics , Evolution, Molecular , Inbreeding , Recombination, Genetic/genetics , Animals , Caenorhabditis/growth & development , Caenorhabditis elegans/genetics , Chromosome Mapping , Chromosomes/genetics , Crosses, Genetic , Databases, Genetic , Female , Gene Rearrangement/genetics , Genetic Variation , Genome/genetics , Genotype , Linkage Disequilibrium/genetics , Male , Phenotype , Polymorphism, Single Nucleotide/genetics , Species Specificity , Synteny/genetics
9.
Dis Model Mech ; 17(8)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38946472

ABSTRACT

Ras genes are important oncogenes that are frequently mutated in cancer. Human oncogenic variants exhibit functional distinctions in terms of their representation in different cancer types, impact on cellular targets and sensitivity to pharmacological treatments. However, how these distinct variants influence and respond to the cellular networks in which they are embedded is poorly understood. To identify novel participants in the complex interplay between Ras genotype and cell interaction networks in vivo, we have developed and tested an experimental framework using a simple vulva-development assay in the nematode C. elegans. Using this system, we evaluated a set of Ras oncogenic substitution changes at G12, G13 and Q61. We found that these variants fall into distinct groups based on phenotypic differences, sensitivity to gene dosage and inhibition of the downstream kinase MEK and their response to genetic modulators that influence Ras activity in a non-autonomous manner. Together, our results demonstrated that oncogenic C. elegans Ras variants exhibit clear distinctions in how they interface with the vulva-development network and showed that extracellular modulators yield variant-restricted effects in vivo.


Subject(s)
Caenorhabditis elegans , Vulva , ras Proteins , Caenorhabditis elegans/genetics , Animals , Vulva/pathology , Vulva/metabolism , ras Proteins/metabolism , ras Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Female , Phenotype , Mutation/genetics , Oncogenes/genetics , Humans
10.
Genetics ; 227(3)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38809718

ABSTRACT

Gene duplication is an important substrate for the evolution of new gene functions, but the impacts of gene duplicates on their own activities and on the developmental networks in which they act are poorly understood. Here, we use a natural experiment of lin-12/Notch gene duplication within the nematode genus Caenorhabditis, combined with characterization of loss- and gain-of-function mutations, to uncover functional distinctions between the duplicate genes in 1 species (Caenorhabditis briggsae) and their single-copy ortholog in Caenorhabditis elegans. First, using improved genomic sequence and gene model characterization, we confirm that the C. briggsae genome includes 2 complete lin-12 genes, whereas most other genes encoding proteins that participate in the LIN-12 signaling pathway retain a one-to-one orthology with C. elegans. We use CRISPR-mediated genome editing to introduce alleles predicted to cause gain-of-function (gf) or loss-of-function (lf) into each C. briggsae gene and find that the gf mutations uncover functional distinctions not apparent from the lf alleles. Specifically, Cbr-lin-12.1(gf), but not Cbr-lin-12.2(gf), causes developmental defects similar to those observed in Cel-lin-12(gf). In contrast to Cel-lin-12(gf), however, the Cbr-lin-12.1(gf) alleles do not cause dominant phenotypes as compared to the wild type, and the mutant phenotype is observed only when 2 gf alleles are present. Our results demonstrate that gene duplicates can exhibit differential capacities to compensate for each other and to interfere with normal development, and uncover coincident gene duplication and evolution of developmental sensitivity to LIN-12/Notch activity.


Subject(s)
Caenorhabditis elegans Proteins , Evolution, Molecular , Gene Duplication , Receptors, Notch , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis/genetics , Signal Transduction/genetics
11.
Nat Methods ; 7(9): 693-5, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20805797

ABSTRACT

The technical toolkit for Caenorhabditis elegans expands to include experimental selection using antibiotic resistance genes.


Subject(s)
Caenorhabditis elegans/genetics , Drug Resistance/genetics , Selection, Genetic , Animals , Animals, Genetically Modified , Caenorhabditis elegans/drug effects , Drug Resistance/drug effects , Gentamicins/pharmacology , Selection, Genetic/drug effects , Transgenes/genetics
13.
Nat Genet ; 36(3): 231-2, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14758362

ABSTRACT

The evolution of complexity relies on changes that result in new gene functions. Here we show that the unique morphological and functional features of the excretory duct cell in C. elegans result from the gain of expression of a single gene. Our results show that innovation can be achieved by altered expression of a transcription factor without coevolution of all target genes.


Subject(s)
Caenorhabditis elegans/anatomy & histology , Caenorhabditis elegans/genetics , Genes, Helminth , Animals , Base Sequence , Caenorhabditis elegans Proteins/genetics , Evolution, Molecular , Morphogenesis , Sequence Homology , Transcription Factors/genetics , Transgenes , Zinc Fingers/genetics
14.
Sci Rep ; 13(1): 16285, 2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37770498

ABSTRACT

Mathematical models of complex systems rely on parameter values to produce a desired behavior. As mathematical and computational models increase in complexity, it becomes correspondingly difficult to find parameter values that satisfy system constraints. We propose a Markov Chain Monte Carlo (MCMC) approach for the problem of constrained model parameter generation by designing a Markov chain that efficiently explores a model's parameter space. We demonstrate the use of our proposed methodology to analyze responses of a newly constructed bistability-constrained model of protein phosphorylation to perturbations in the underlying protein network. Our results suggest that parameter generation for constrained models using MCMC provides powerful tools for modeling-aided analysis of complex natural processes.

15.
G3 (Bethesda) ; 12(10)2022 09 30.
Article in English | MEDLINE | ID: mdl-35929788

ABSTRACT

Communication between mesodermal cells and epithelial cells is fundamental to normal animal development and is frequently disrupted in cancer. However, the genes and processes that mediate this communication are incompletely understood. To identify genes that mediate this communication and alter the proliferation of cells with an oncogenic Ras genotype, we carried out a tissue-specific genome-wide RNAi screen in Caenorhabditis elegans animals bearing a let-60(n1046gf) (RasG13E) allele. The screen identifies 24 genes that, when knocked down in adjacent mesodermal tissue, suppress the increased vulval epithelial cell proliferation defect associated with let-60(n1046gf). Importantly, gene knockdown reverts the mutant animals to a wild-type phenotype. Using chimeric animals, we genetically confirm that 2 of the genes function nonautonomously to revert the let-60(n1046gf) phenotype. The effect is genotype restricted, as knockdown does not alter development in a wild type (let-60(+)) or activated EGF receptor (let-23(sa62gf)) background. Although many of the genes identified encode proteins involved in essential cellular processes, including chromatin formation, ribosome function, and mitochondrial ATP metabolism, knockdown does not alter the normal development or function of targeted mesodermal tissues, indicating that the phenotype derives from specific functions performed by these cells. We show that the genes act in a manner distinct from 2 signal ligand classes (EGF and Wnt) known to influence the development of vulval epithelial cells. Altogether, the results identify genes with a novel function in mesodermal cells required for communicating with and promoting the proliferation of adjacent epithelial cells with an activated Ras genotype.


Subject(s)
Caenorhabditis elegans Proteins , Adenosine Triphosphate/metabolism , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Chromatin/metabolism , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , Female , Helminth Proteins/genetics , Ligands , Mutation , Signal Transduction/genetics , Vulva/metabolism , ras Proteins/genetics , ras Proteins/metabolism
16.
Dev Biol ; 332(1): 156-65, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19481074

ABSTRACT

Wnt signaling and HOM-C/Hox genes pattern cell fate along the anterior/posterior axis in many animals. In general, Wnt signaling participates in establishing the anterior/posterior axis, whereas HOM-C genes confer regional identities to cells along the axis. However, recent work in non-bilaterial metazoans suggests that the ancestral patterning system relied on Wnts, with a later co-option of HOM-C genes to replace Wnts in regional patterning. Here we provide direct experimental support for this model from C. elegans, where a regional Wnt patterning system is uncovered in HOM-C gene mutants. Anterior/posterior patterning of P11/P12 cell fate in the C. elegans tail is normally dependent on the HOM-C gene egl-5/Abdominal-B. If the HOM-C gene mab-5/fushi tarazu is also mutant, however, a Wnt signal can promote P12 fate in the absence of egl-5. Furthermore, transcription of egl-5 in the P12.pa cell is influenced by an autoregulatory element that is essential in wild type, but not in mab-5 egl-5 double mutants, identifying regulatory parallels between P12 cell fate specification and egl-5 transcriptional regulation in the P12 lineage. Together, our results identify complex regulatory relationships among signaling pathways and HOM-C genes, and uncover a layering of patterning systems that may reflect their evolutionary history.


Subject(s)
Body Patterning , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Epidermis/embryology , Genes, Homeobox , Signal Transduction , Wnt Proteins/metabolism , Animals , Base Sequence , Body Patterning/drug effects , Caenorhabditis elegans/cytology , Caenorhabditis elegans/drug effects , Caenorhabditis elegans Proteins/genetics , Cell Fusion , Cell Lineage/drug effects , Conserved Sequence , Enhancer Elements, Genetic/genetics , Epidermal Cells , Epidermal Growth Factor/pharmacology , Epidermis/drug effects , Epidermis/metabolism , Gene Expression Regulation, Developmental/drug effects , Genes, Reporter , Homeostasis/drug effects , Models, Biological , Molecular Sequence Data , Mutation/genetics , Signal Transduction/drug effects , Transgenes
17.
BMC Genomics ; 11: 236, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20385026

ABSTRACT

BACKGROUND: The nematode C. briggsae serves as a useful model organism for comparative analysis of developmental and behavioral processes. The amenability of C. briggsae to genetic manipulations and the availability of its genome sequence have prompted researchers to study evolutionary changes in gene function and signaling pathways. These studies rely on the availability of forward genetic tools such as mutants and mapping markers. RESULTS: We have computationally identified more than 30,000 polymorphisms (SNPs and indels) in C. briggsae strains AF16 and HK104. These include 1,363 SNPs that change restriction enzyme recognition sites (snip-SNPs) and 638 indels that range between 7 bp and 2 kb. We established bulk segregant and single animal-based PCR assay conditions and used these to test 107 polymorphisms. A total of 75 polymorphisms, consisting of 14 snip-SNPs and 61 indels, were experimentally confirmed with an overall success rate of 83%. The utility of polymorphisms in genetic studies was demonstrated by successful mapping of 12 mutations, including 5 that were localized to sub-chromosomal regions. Our mapping experiments have also revealed one case of a misassembled contig on chromosome 3. CONCLUSIONS: We report a comprehensive set of polymorphisms in C. briggsae wild-type strains and demonstrate their use in mapping mutations. We also show that molecular markers can be useful tools to improve the C. briggsae genome sequence assembly. Our polymorphism resource promises to accelerate genetic and functional studies of C. briggsae genes.


Subject(s)
Caenorhabditis/genetics , Chromosome Mapping/methods , Animals , INDEL Mutation , Polymorphism, Restriction Fragment Length , Polymorphism, Single Nucleotide
18.
Mol Genet Genomics ; 283(1): 73-87, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19921263

ABSTRACT

Excretory renal organs are critical in animals for osmoregulation and the elimination of waste. Renal organs across a range of species exhibit cellular and molecular similarities. For example, class III POU-homeodomain transcription factors are expressed in the renal organs of many invertebrates and vertebrates. However, the functional role for these factors is not well characterized. To better understand the role of class III POU-homeodomain proteins in animal excretory systems, we have characterized a set of genes expressed in the Caenorhabditis elegans excretory cell, and determined their regulation by the POU-III transcription factor CEH-6. Our molecular and biochemical studies show that CEH-6 regulates a subset of genes expressed in the excretory cell. Additionally, we find that the CEH-6-dependent genes share two molecular features: they contain at least one octamer regulatory element and they encode for transport and channel proteins. This work suggests that a role for POU-III factors in renal organs is to coordinate the expression of a set of functionally related genes.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Gene Expression Regulation , Homeodomain Proteins/metabolism , POU Domain Factors/metabolism , Water-Electrolyte Balance/genetics , Animals , Base Sequence , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Gene Regulatory Networks , Homeodomain Proteins/genetics , Organic Anion Transporters/genetics , POU Domain Factors/genetics
19.
Neoplasia ; 22(10): 484-496, 2020 10.
Article in English | MEDLINE | ID: mdl-32818842

ABSTRACT

Androgen Receptor (AR) signaling is a critical driver of hormone-dependent prostate cancer and has also been proposed to have biological activity in female hormone-dependent cancers, including type I endometrial carcinoma (EMC). In this study, we evaluated the preclinical efficacy of a third-generation AR antagonist, enzalutamide, in a genetic mouse model of EMC, Sprr2f-Cre;Ptenfl/fl. In this model, ablation of Pten in the uterine epithelium leads to localized and distant malignant disease as observed in human EMC. We hypothesized that administering enzalutamide through the diet would temporarily decrease the incidence of invasive and metastatic carcinoma, while prolonged administration would result in development of resistance and loss of efficacy. Short-term treatment with enzalutamide reduced overall tumor burden through increased apoptosis but failed to prevent progression of invasive and metastatic disease. These results suggest that AR signaling may have biphasic, oncogenic and tumor suppressive roles in EMC that are dependent on disease stage. Enzalutamide treatment increased Progesterone Receptor (PR) expression within both stromal and tumor cell compartments. Prolonged administration of enzalutamide decreased apoptosis, increased tumor burden and resulted in the clonal expansion of tumor cells expressing high levels of p53 protein, suggestive of acquired Trp53 mutations. In conclusion, we show that enzalutamide induces apoptosis in EMC but has limited efficacy overall as a single agent. Induction of PR, a negative regulator of endometrial proliferation, suggests that adding progestin therapy to enzalutamide administration may further decrease tumor burden and result in a prolonged response.


Subject(s)
Apoptosis , Benzamides/pharmacology , Cornified Envelope Proline-Rich Proteins/physiology , Disease Models, Animal , Drug Resistance, Neoplasm , Endometrial Neoplasms/drug therapy , Nitriles/pharmacology , PTEN Phosphohydrolase/physiology , Phenylthiohydantoin/pharmacology , Animals , Cell Proliferation , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Male , Mice , Mice, Knockout , Signal Transduction , Tumor Burden
SELECTION OF CITATIONS
SEARCH DETAIL