Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Dev Biol ; 398(2): 206-17, 2015 02 15.
Article in English | MEDLINE | ID: mdl-25481758

ABSTRACT

Egg chambers from starved Drosophila females contain large aggregates of processing (P) bodies and cortically enriched microtubules. As this response to starvation is rapidly reversed upon re-feeding females or culturing egg chambers with exogenous bovine insulin, we examined the role of endogenous insulin signaling in mediating the starvation response. We found that systemic Drosophila insulin-like peptides (dILPs) activate the insulin pathway in follicle cells, which then regulate both microtubule and P body organization in the underlying germline cells. This organization is modulated by the motor proteins Dynein and Kinesin. Dynein activity is required for microtubule and P body organization during starvation, while Kinesin activity is required during nutrient-rich conditions. Blocking the ability of egg chambers to form P body aggregates in response to starvation correlated with reduced progeny survival. These data suggest a potential mechanism to maximize fecundity even during periods of poor nutrient availability, by mounting a protective response in immature egg chambers.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Food , Germ Cells/metabolism , Insulin/metabolism , Ovum/cytology , Signal Transduction , Animals , Apoptosis , Cattle , Cytoplasmic Structures/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Embryo, Nonmammalian/metabolism , Female , Microtubules/metabolism , Models, Biological , Oocytes/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Ovum/metabolism , Peptides/metabolism , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism , TOR Serine-Threonine Kinases/metabolism
2.
BMC Genomics ; 11: 285, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20459611

ABSTRACT

BACKGROUND: The central metabolic pathway of glycolysis converts glucose to pyruvate, with the net production of 2 ATP and 2 NADH per glucose molecule. Each of the ten reactions in this pathway is typically catalyzed by multiple isozymes encoded by a multigene family. Several isozymes in this pathway are expressed only during spermatogenesis, and gene targeting studies indicate that they are essential for sperm function and male fertility in mouse. At least three of the novel glycolytic isozymes are encoded by retrogenes (Pgk2, Aldoart1, and Aldoart2). Their restricted expression profile suggests that retrotransposition may play a significant role in the evolution of sperm glycolytic enzymes. RESULTS: We conducted a comprehensive genomic analysis of glycolytic enzymes in the human and mouse genomes and identified several intronless copies for all enzymes in the pathway, except Pfk. Within each gene family, a single orthologous gene was typically retrotransposed frequently and independently in both species. Several retroposed sequences maintained open reading frames (ORFs) and/or provided evidence of alternatively spliced exons. We analyzed expression of sequences with ORFs and <99% sequence identity in the coding region and obtained evidence for the expression of an alternative Gpi1 transcript in mouse spermatogenic cells. CONCLUSIONS: Our analysis detected frequent, recent, and lineage-specific retrotransposition of orthologous glycolytic enzymes in the human and mouse genomes. Retrotransposition events are associated with LINE/LTR and genomic integration is random. We found evidence for the alternative splicing of parent genes. Many retroposed sequences have maintained ORFs, suggesting a functional role for these genes.


Subject(s)
Glycolysis , Retroelements , Spermatozoa/enzymology , Alternative Splicing , Animals , Gene Expression , Humans , Long Interspersed Nucleotide Elements , Male , Membrane Proteins/genetics , Mice , Open Reading Frames , Spermatozoa/metabolism , Terminal Repeat Sequences , Testis/metabolism
3.
Sci Signal ; 2(102): ra86, 2009 Dec 22.
Article in English | MEDLINE | ID: mdl-20029031

ABSTRACT

Amino acid residues 1 to 434 of the E3 ubiquitin ligase Cbl control signaling of the epidermal growth factor receptor (EGFR) by enhancing its ubiquitination, down-regulation, and lysosomal degradation. This region of Cbl comprises a tyrosine kinase-binding domain, a linker region, a really interesting new gene finger (RF), and a subset of the residues of the RF tail. In experiments with full-length alanine substitution mutants, we demonstrated that the RF tail of Cbl regulated biochemically distinct checkpoints in the endocytosis of EGFR. The Cbl- and ubiquitin-dependent degradation of the regulator of internalization hSprouty2 was compromised by the Val(431)--> Ala mutation, whereas the Cbl- and EGFR-dependent dephosphorylation or degradation of the endosomal trafficking regulator Hrs was compromised by the Phe(434)--> Ala mutation. Deregulated phosphorylation of Hrs correlated with inhibition of the fusion of early endosomes and of the degradation of EGFR. This study provides the first evidence that Cbl regulates receptor fate by controlling the fusion of sorting endosomes. We postulate that it does so by modulating the abundance of tyrosine-phosphorylated Hrs.


Subject(s)
Down-Regulation/physiology , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/metabolism , ErbB Receptors/metabolism , Membrane Fusion/physiology , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , COS Cells , Chlorocebus aethiops , Humans , Immunoprecipitation , Microscopy, Fluorescence , Phosphorylation
4.
Dev Biol ; 314(2): 300-16, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18191828

ABSTRACT

We report the cloning and characterization of MOEP19, a novel 19 kDa RNA binding protein that marks a defined cortical cytoplasmic domain in oocytes and provides evidence of mammalian oocyte polarity and a form of pre-patterning that persists in zygotes and early embryos through the morula stage. MOEP19 contains a eukaryotic type KH-domain, typical of the KH-domain type I superfamily of RNA binding proteins, and both recombinant and native MOEP19 bind polynucleotides. By immunofluorescence, MOEP19 protein was first detected in primary follicles throughout the ooplasm. As oocytes expanded in size during oogenesis, MOEP19 increased in concentration. MOEP19 localized in the ovulated egg and early zygote as a symmetrical spherical cortical domain underlying the oolemma, deep to the zone of cortical granules. MOEP19 remained restricted to a cortical cytoplasmic crescent in blastomeres of 2-, 4- and 8-cell embryos. The MOEP19 domain was absent in regions underlying cell contacts. In morulae, the MOEP19 domain was found at the apex of outer, polarized blastomeres but was undetectable in blastomeres of the inner cell mass. In early blastocysts, MOEP19 localized in both mural and polar trophectoderm and a subset of embryos showed inner cell mass localization. MOEP19 concentration dramatically declined in late blastocysts. When blastomeres of 4- to 8-cell stages were dissociated, the polarized MOEP19 domain assumed a symmetrically spherical localization, while overnight culture of dissociated blastomeres resulted in formation of re-aggregated embryos in which polarity of the MOEP19 domain was re-established at the blastomere apices. MOEP19 showed no evidence of translation in ovulated eggs, indicating that MOEP19 is a maternal effect gene. The persistence during early development of the MOEP19 cortical oocyte domain as a cortical crescent in blastomers suggests an intrinsic pre-patterning in the egg that is related to the apical-basolateral polarity of the embryo. Although the RNAs bound to MOEP19 are presently unknown, we predict that the MOEP19 domain directs RNAs essential for normal embryonic development to specific locations in the oocyte and early embryo.


Subject(s)
Blastomeres/physiology , Ectoderm/physiology , Oocytes/physiology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Trophoblasts/physiology , Amino Acid Sequence , Animals , Blastomeres/cytology , Cell Polarity , Cloning, Molecular , Conserved Sequence , Ectoderm/cytology , Egg Proteins/analysis , Embryo, Mammalian/physiology , Embryonic Development , Female , Fluorescent Antibody Technique, Indirect , Mass Spectrometry , Methionine/metabolism , Mice , Molecular Sequence Data , Oocytes/cytology , Phosphorylation , Polymerase Chain Reaction , RNA/genetics , RNA/isolation & purification , Recombinant Proteins/metabolism , Trophoblasts/cytology
5.
Dev Biol ; 309(1): 18-31, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17659271

ABSTRACT

Enzymes in the glycolytic pathway of mammalian sperm are modified extensively and are localized in the flagellum, where several are tightly bound to the fibrous sheath. This study provides the first evidence for three novel aldolase isozymes in mouse sperm, two encoded by Aldoart1 and Aldoart2 retrogenes on different chromosomes and another by Aldoa_v2, a splice variant of Aldoa. Phylogenetic analyses and comparative genomics indicate that the retrogenes and splice variant have remained functional and have been under positive selection for millions of years. Their expression is restricted to the male germline and is tightly regulated at both transcriptional and translational levels. All three isozymes are present only in spermatids and sperm and have distinctive features that may be important for localization in the flagellum and/or altered metabolic regulation. Both ALDOART1 and ALDOA_V2 have unusual N-terminal extensions not found in other aldolases. The N-terminal extension of ALDOA_V2 is highly conserved in mammals, suggesting a conserved function in sperm. We hypothesize that the N-terminal extensions are responsible for localizing components of the glycolytic pathway to the fibrous sheath and that this localization is required to provide sufficient ATP along the length of the flagellum to support sperm motility.


Subject(s)
Alternative Splicing , Fructose-Bisphosphate Aldolase/metabolism , Phylogeny , Retroelements/physiology , Spermatozoa/metabolism , Amino Acid Sequence , Animals , Fructose-Bisphosphate Aldolase/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Molecular Sequence Data , Retroelements/genetics , Testis/metabolism
6.
Mol Hum Reprod ; 10(6): 433-44, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15044604

ABSTRACT

Two members of the human testis-specific serine/threonine (Ser/Thr) kinase family, TSSK 1 and TSSK 2, were cloned and sequenced from a human testis adaptor-ligated cDNA library using a PCR strategy. Within the cDNA, open reading frames (ORF) were defined encoding proteins of 367 and 358 amino acids respectively, as well as conserved kinase domains typical of the superfamily of Ser/Thr kinases. Both genes were intronless and mapped to chromosomes 5 and 22 respectively. The human and mouse homologues of TSSK 1 and TSSK 2, together with TSSK 3 and SSTK/FKSG82, constitute a kinase subfamily closely related to the calmodulin kinases and SNF/nim 1 kinase subfamilies. Similar to the mouse, tissue expression by northern and dot blot analysis revealed that human TSSK 1 and 2 messages are expressed exclusively in the testis. However, mRNA for these kinases can be detected in other tissues using real-time PCR. In addition, TSKS, the human homologue of a putative substrate of TSSK 1 and 2, was cloned. TSKS had an ORF of 592 amino acids and was also expressed exclusively in the testis as demonstrated by northern and dot blot analyses; however, lower levels of expression in other tissues were detected using real-time PCR. Human TSSK 2 and TSKS interacted in a yeast two-hybrid system and also co-immunoprecipitated after in vitro translation. TSSK 2 expressed in yeast and bacteria was able to autophosphorylate and also phosphorylated recombinant TSKS in vitro. Antibodies against recombinant TSSK 2 demonstrated that a member of the TSSK family was present in human testis and localized to the equatorial segment of ejaculated human sperm. In contrast, TSKS was only found in the testis. The finding of a TSSK family member in mature sperm suggests that this family of kinases might play a role in sperm function.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Spermatozoa/cytology , Spermatozoa/enzymology , Testis/enzymology , Amino Acid Sequence , Animals , Cloning, Molecular , Cytoskeletal Proteins , Humans , Male , Mice , Molecular Sequence Data , Phosphoproteins , Phylogeny , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/classification , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Testis/cytology , Testis/physiology , Tissue Distribution , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL