Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Bacteriol ; 194(12): 3189-98, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22505682

ABSTRACT

The tubulin homolog FtsZ forms a polymeric membrane-associated ring structure (Z ring) at midcell that establishes the site of division and provides an essential framework for the localization of a multiprotein molecular machine that promotes division in Escherichia coli. A number of regulatory proteins interact with FtsZ and modulate FtsZ assembly/disassembly processes, ensuring the spatiotemporal integrity of cytokinesis. The Z-associated proteins (ZapA, ZapB, and ZapC) belong to a group of FtsZ-regulatory proteins that exhibit functionally redundant roles in stabilizing FtsZ-ring assembly by binding and bundling polymeric FtsZ at midcell. In this study, we report the identification of ZapD (YacF) as a member of the E. coli midcell division machinery. Genetics and cell biological evidence indicate that ZapD requires FtsZ but not other downstream division proteins for localizing to midcell, where it promotes FtsZ-ring assembly via molecular mechanisms that overlap with ZapA. Biochemical evidence indicates that ZapD directly interacts with FtsZ and promotes bundling of FtsZ protofilaments. Similarly to ZapA, ZapB, and ZapC, ZapD is dispensable for division and therefore belongs to the growing group of FtsZ-associated proteins in E. coli that aid in the overall fitness of the division process.


Subject(s)
Bacterial Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Division , Cytoskeletal Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Cell Cycle Proteins/genetics , Escherichia coli Proteins/genetics , Genes, Essential , Protein Binding , Protein Interaction Mapping
2.
Dev Dyn ; 240(3): 723-36, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21337470

ABSTRACT

We describe the localization of the golgin GMAP210 and the intraflagellar protein IFT88 in the Golgi of spermatids and the participation of these two proteins in the development of the acrosome-acroplaxome complex, the head-tail coupling apparatus (HTCA) and the spermatid tail. Immunocytochemical experiments show that GMAP210 predominates in the cis-Golgi, whereas IFT88 prevails in the trans-Golgi network. Both proteins colocalize in proacrosomal vesicles, along acrosome membranes, the HTCA and the developing tail. IFT88 persists in the acrosome-acroplaxome region of the sperm head, whereas GMAP210 is no longer seen there. Spermatids of the Ift88 mouse mutant display abnormal head shaping and are tail-less. GMAP210 is visualized in the Ift88 mutant during acrosome-acroplaxome biogenesis. However, GMAP210-stained vesicles, mitochondria and outer dense fiber material build up in the manchette region and fail to reach the abortive tail stump in the mutant. In vitro disruption of the spermatid Golgi and microtubules with Brefeldin-A and nocodazole blocks the progression of GMAP210- and IFT88-stained proacrosomal vesicles to the acrosome-acroplaxome complex but F-actin distribution in the acroplaxome is not affected. We provide the first evidence that IFT88 is present in the Golgi of spermatids, that the microtubule-associated golgin GMAP210 and IFT88 participate in acrosome, HTCA, and tail biogenesis, and that defective intramanchette transport of cargos disrupts spermatid tail development.


Subject(s)
Acrosome/metabolism , Golgi Apparatus/metabolism , Nuclear Proteins/metabolism , Spermatids/metabolism , Tumor Suppressor Proteins/metabolism , Acrosome/ultrastructure , Actins/metabolism , Animals , Brefeldin A/pharmacology , Cytoskeletal Proteins , Fluorescent Antibody Technique, Indirect , Golgi Apparatus/ultrastructure , Immunoblotting , Immunohistochemistry , Male , Mice , Microscopy, Electron , Microtubules/metabolism , Microtubules/ultrastructure , Nocodazole/pharmacology , Nuclear Proteins/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Spermatids/ultrastructure , Tumor Suppressor Proteins/genetics
3.
Spermatogenesis ; 1(3): 221-230, 2011 Jul.
Article in English | MEDLINE | ID: mdl-22319670

ABSTRACT

Spermatids generate diverse and unusual actin and microtubule populations during spermiogenesis to fulfill mechanical and cargo transport functions assisted by motor and non-motor proteins. Disruption of cargo transport may lead to teratozoospermia and consequent male infertility. How motor and non-motor proteins utilize the cytoskeleton to transport cargos during sperm development is not clear. Filamentous actin (F-actin) and the associated motor protein myosin Va participate in the transport of Golgi-derived proacrosomal vesicles to the acrosome and along the manchette. The acrosome is stabilized by the acroplaxome, a cytoskeletal plate anchored to the nuclear envelope. The acroplaxome plate harbors F-actin and actin-like proteins as well as several other proteins, including keratin 5/Sak57, Ran GTPase, Hook1, dynactin p150Glued, cenexin-derived ODF2, testis-expressed profilin-3 and profilin-4, testis-expressed Fer tyrosine kinase (FerT), members of the ubiquitin-proteasome system and cortactin. Spermatids express transcripts encoding the non-spliced form of cortactin, a F-actin-regulatory protein. Tyrosine phosphorylated cortactin and FerT coexist in the acrosome-acroplaxome complex. Hook1 and p150Glued, known to participate in vesicle cargo transport, are sequentially seen from the acroplaxome to the manchette to the head-tail coupling apparatus (HTCA). The golgin Golgi-microtubule associated protein GMAP210 resides in the cis-Golgi whereas the intraflagellar protein IFT88 localizes in the trans-Golgi network. Like Hook1 and p150Glued, GMAP210 and IFT88 colocalize at the cytosolic side of proacrosomal vesicles and, following vesicle fusion, become part of the outer and inner acrosomal membranes before relocating to the acroplaxome, manchette and HTCA. A hallmark of the manchette and axoneme is microtubule heterogeneity, determined by the abundance of acetylated, tysosinated and glutamylated tubulin isoforms produced by post-translational modifications. We postulate that the construction of the male gamete requires microtubule and F-actin tracks and specific molecular motors and associated non-motor proteins for the directional positioning of vesicular and non-vesicular cargos at specific intracellular sites.

4.
Biol Reprod ; 81(6): 1196-205, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19710508

ABSTRACT

The hypodactylous (hd) locus impairs limb development and spermatogenesis, leading to male infertility in rats. We show that the hd mutation is caused by an insertion of an endogenous retrovirus into intron 10 of the Cntrob gene. The retroviral insertion in hd mutant rats disrupts the normal splicing of Cntrob transcripts and results in the expression of a truncated protein. During the final phase of spermiogenesis, centrobin localizes to the manchette, centrosome, and the marginal ring of the spermatid acroplaxome, where it interacts with keratin 5-containing intermediate filaments. Mutant spermatids show a defective acroplaxome marginal ring and separation of the centrosome from its normal attachment site of the nucleus. This separation correlates with a disruption of head-tail coupling apparatus, leading to spermatid decapitation during the final step of spermiogenesis and the absence of sperm in the epididymis. Cntrob may represent a novel candidate gene for presently unexplained hereditary forms of teratozoospermia and the "easily decapitated sperm syndrome" in humans.


Subject(s)
Cell Cycle Proteins/physiology , Genes, Homeobox/genetics , Homeodomain Proteins/genetics , Sperm Head/metabolism , Sperm Tail/metabolism , Spermatogenesis/genetics , Animals , Blotting, Far-Western , Centrosome/metabolism , Endogenous Retroviruses/genetics , Epididymis/metabolism , Fluorescent Antibody Technique , Homeodomain Proteins/metabolism , Infertility, Male/genetics , Infertility, Male/metabolism , Introns/genetics , Keratin-5/genetics , Keratin-5/metabolism , Male , Microscopy, Electron , Mutation/genetics , Protein Transport/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Spermatids/metabolism
5.
Dev Dyn ; 238(7): 1851-61, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19517565

ABSTRACT

We report the cDNA cloning of rat testis Rnf19a, a ubiquitin protein ligase, and show 98% and 93% protein sequence identity of testicular mouse and human Rnf19a, respectively. Rnf19a interacts with Psmc3, a protein component of the 19S regulatory cap of the 26S proteasome. During spermatid development, Rnf19a and Psmc3 are initially found in Golgi-derived proacrosomal vesicles. Later on, Rnf19a, Psmc3, and ubiquitin are seen along the cytosolic side of the acrosomal membranes and the acroplaxome, a cytoskeletal plate linking the acrosome to the spermatid nuclear envelope. Rnf19a and Psmc3 accumulate at the acroplaxome marginal ring-manchette perinuclear ring region during spermatid head shaping and in the developing sperm head-tail coupling apparatus and tail. Rnf19a and Psmc3 may interact directly or indirectly with each other, presumably pointing to the participation of the ubiquitin-proteasome system in acrosome biogenesis, spermatid head shaping, and development of the head-tail coupling apparatus and tail.


Subject(s)
Acrosome/metabolism , Adenosine Triphosphatases/physiology , Proteasome Endopeptidase Complex/physiology , Sperm Head/physiology , Spermatids/physiology , Ubiquitin-Protein Ligases/physiology , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Animals , Cell Differentiation/physiology , Cell Membrane/metabolism , Male , Models, Biological , Molecular Sequence Data , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Rats , Sequence Homology, Amino Acid , Sperm Head/metabolism , Spermatids/metabolism , Spermatogenesis/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
6.
Mol Reprod Dev ; 76(9): 832-43, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19441121

ABSTRACT

We report that full-length and truncated transcripts of Fyn tyrosine protein kinase are expressed during testicular development. Truncated Fyn (tr-Fyn) transcripts encode a 24 kDa protein with a N-terminal (NT) domain, a complete Src homology (SH) 3 domain and an incomplete SH2 domain. The kinase domain is missing in tr-Fyn. In contrast, full-length Fyn transcripts encode a 59-55 kDa protein. Fractionated spermatids by centrifugal elutriation express tr-Fyn transcripts and protein, but not full-length Fyn transcripts and protein. Neither full-length Fyn nor tr-Fyn transcripts and encoded proteins are detected in elutriated pachytene spermatocytes. Sertoli cells express full-length and truncated Fyn throughout testicular development. In contrast, sperm contain full-length Fyn transcripts and protein but not the truncated form. tr-Fyn protein is visualized at the cytosolic side of Golgi membranes, derived proacrosomal vesicles, along the outer acrosome membrane and the inner acrosome membrane-acroplaxome complex anchoring the acrosome to the spermatid nuclear envelope. Fyn and phosphotyrosine immunoreactivity coexist in the tail of capacitated sperm. During fertilization, the Fyn-containing acroplaxome seen in the egg-bound and egg-fused sperm is no longer detected upon decondensation of the sperm nucleus. tr-Fyn expands the catalog of truncated tyrosine protein kinases expressed during spermiogenesis. We suggest that the NT and SH3 domains of tr-Fyn may recruit adaptor and effector proteins, in particular GTPase activating proteins, required for acrosome-acroplaxome biogenesis, acroplaxome F-actin dynamics and Sertoli cell function. During fertilization, full-length Fyn in the acroplaxome may contribute to a transient local signaling burst during the early events of sperm-egg interaction.


Subject(s)
Acrosome/metabolism , Fertilization/physiology , Proto-Oncogene Proteins c-fyn/biosynthesis , Spermatogenesis/physiology , Animals , Immunohistochemistry , Male , Microscopy, Electron , Microscopy, Fluorescence , Proto-Oncogene Proteins c-fyn/chemistry , Proto-Oncogene Proteins c-fyn/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sertoli Cells/metabolism , Sperm Capacitation , Spermatids/metabolism , Spermatozoa/metabolism , Testis/metabolism
7.
Dev Dyn ; 237(12): 3882-91, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18985748

ABSTRACT

We report the association of testicular Fer, a non-receptor tyrosine kinase, with acrosome development and remodeling of the acrosome-associated acroplaxome plate during spermatid head shaping. A single gene expresses two forms of Fer tyrosine kinases in testis: a somatic form (FerS) and a truncated testis-type form (FerT). FerT transcript variants are seen in spermatocytes and spermatids. FerS transcripts are not detected in round spermatids but are moderately transcribed in spermatocytes. FerT protein is associated with the spermatid medial/trans-Golgi region, proacrosomal vesicles, the cytosolic side of the outer acrosome membrane and adjacent to the inner acrosome membrane facing the acroplaxome. FerT coexist in the acroplaxome with phosphorylated cortactin, a regulator of F-actin dynamics. We propose that FerT participates in acrosome development and that phosphorylated cortactin may contribute to structural changes in F-actin in the acroplaxome during spermatid head shaping.


Subject(s)
Gene Expression Regulation, Developmental , Protein-Tyrosine Kinases/metabolism , Sperm Head/enzymology , Spermatids/enzymology , Transcription, Genetic/genetics , Animals , Gene Expression Regulation, Enzymologic , Genetic Variation/genetics , Humans , Male , Mice , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/genetics , Rats , Sperm Head/metabolism , Sperm Head/ultrastructure , Spermatids/growth & development , Spermatids/metabolism , Spermatids/ultrastructure
8.
Mol Reprod Dev ; 75(11): 1591-606, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18398819

ABSTRACT

Outer dense fibers are a major constituent of the sperm tail and outer dense fiber 2 (ODF2) protein is one of their major components. ODF2 shares partial homology with cenexin 1 and cenexin 2, regarded as centriolar proteins. We show that ODF2 and cenexin 2 transcripts are the product of differential splicing of a single gene, designated Cenexin/ODF2 and that cenexin 1 is an incomplete clone of ODF2. ODF2 terminates in exon 20b whereas in cenexin 2 this exon is spliced out and translation terminates in exon 24. We demonstrate a transcriptional switch during rat testicular development, from somatic-type to testis-type ODF2 and cenexin transcripts during the onset of meiosis. The switch is completed when spermiogenesis is established. ODF2 immunoreactive sites were visualized in the acroplaxome, along the sperm tail and the centrosome-derived sperm head-to-tail coupling apparatus. An unexpected finding was the presence of ODF2 antigenic sites, but not cenexin antigenic sites, in the dense fibrillar component of the nucleolus of Sertoli cells, spermatogonia and primary spermatocytes. The characterization of the genomic origin, processing and developmental expression of ODF2 transcript isoforms and their protein products can help reconcile differences in the literature on the role of ODF2 and cenexin in the centrosome. Furthermore, the finding of ODF2 in the dense fibrillar component of the nucleolus suggests that this protein, in addition to its presence in sperm outer dense fibers and centrosome, highlights and adds to the nucleolar function during spermatogenesis and early embryogenesis.


Subject(s)
Cell Nucleolus/physiology , Gene Expression Regulation, Developmental/physiology , Genome/physiology , Heat-Shock Proteins/genetics , RNA Processing, Post-Transcriptional/physiology , Testis/metabolism , Transcription, Genetic/physiology , Amino Acid Sequence , Animals , Animals, Newborn , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/physiology , Male , Mice , Molecular Sequence Data , Organ Specificity/genetics , Rats , Sertoli Cells/metabolism , Sertoli Cells/physiology , Spermatogenesis/genetics , Spermatozoa/cytology , Spermatozoa/physiology , Testis/physiology
9.
Soc Reprod Fertil Suppl ; 65: 33-43, 2007.
Article in English | MEDLINE | ID: mdl-17644953

ABSTRACT

The shaping of the mammalian sperm involves the elongation and condensation of the spermatid nucleus, the development of the acrosome, and the transient appearance of the microtubular manchette. F-actin-containing ectoplasmic hoops of Sertoli cells embrace the upper third of the spermatid head during elongation. During acrosomal biogenesis, proacrosomal vesicles derived from the Golgi apparatus, dock and fuse along the acroplaxome, an F-actin/keratin 5-containing cytoskeletal plate. The acroplaxome consists of a bent plate and a marginal ring encircling the spermatid nucleus. It anchors the developing acrosome to the spermatid nucleus. The manchette, consisting of a perinuclear rings with inserted microtubules, lies subjacent to the marginal ring of the acroplaxome. During spermatid elongation, the two overlapping rings reduce their diameter to fit, in a sleeve-like fashion, the decreasing diameter of the spermatid nucleus. The acroplaxome may provide a planar scaffold to modulate exogenous constriction forces generated by Sertoli cell F-actin hoops during spermatid head elongation. The dynamics of the F-actin cytoskeleton, one of the components of the acroplaxome and Sertoli cell hoops, can be regulated by tyrosine kinases, which target cortactin, an F-actin-associated protein. Tyrosine phosphorylation of cortactin correlates with a reduction in the crosslinking properties of F-actin. Phosphorylated cortactin and tyrosine kinase Fer are present in the acroplaxome, thus supporting a role of this F-actin remodelling pathway during spermatid head shaping. Keratin 5, an additional component of the acroplaxome, may also undergo dynamic reorganization during spermatid head elongation. We postulate that the F-actin/keratin 5 cytoskeleton in the acroplaxome may undergo a dynamic reorganization to modulate exogenous shear forces exerted by Sertoli cell F-actin hoops during spermatid head shaping. The acroplaxome-manchette perinuclear rings may reduce their diameter to balance exogenous constriction forces generated by the embracing Sertoli cell F-actin hoops and guide nuclear elongation.


Subject(s)
Gene Expression Regulation , Mammals/physiology , Sperm Head/ultrastructure , Spermatogenesis/genetics , Actins/metabolism , Actins/ultrastructure , Animals , Gene Expression , Keratin-5/metabolism , Keratin-5/ultrastructure , Male , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Sertoli Cells/physiology , Sperm Head/metabolism
10.
Mol Reprod Dev ; 73(7): 906-17, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16557522

ABSTRACT

We have previously reported that Sertoli cell geometric changes induced by a Fas (CD95) agonist or by restricting Sertoli cell spreading can trigger spermatogenic cell detachment from Sertoli cell surfaces and initiate a programmed cell death sequence. Here, we have focused on ADAM proteins, tetraspanins CD9 and CD81, and the integrin beta1 subunit, which is co-expressed in testis with integrin alpha3 and integrin alpha6 subunits, to understand how these molecules may stabilize spermatogenic cell attachment to Sertoli cell surfaces. Like ADAM proteins, integrin beta1, alpha3, and alpha6 subunits, and CD9 and CD81 transcripts are expressed in the fetal testis and throughout testicular maturation, as well as, in Sertoli-spermatogenic cell co-cultures. Prespermatogonia (gonocytes) display CD9 and CD81 immunoreactive sites. Integrin alpha6 subunit transcripts have unusual developmental characteristics: fetal testis expresses the integrin alpha6B isoform exclusively. In contrast, the integrin alpha6B isoform co-exists with the integrin alpha6A isoform in prepubertal testes and Sertoli-spermatogenic cell co-cultures. A blocking anti body targeting the extracellular domain (N-terminal) of the integrin beta1 subunit causes rapid contraction of Sertoli cells leading to the gradual detachment of associated spermatogenic cells. In contrast, predicted active site peptides targeting the disintegrin domain of ADAM 1, ADAM 2, ADAM 3 (cyritestin), ADAM 4, ADAM 5, ADAM 6, and ADAM 15 (metragidin) do not disturb significantly the attachment of spermatogenic cells to Sertoli cell surfaces. Spermatogenic cells dislodged from their attachment sites by the integrin beta1 subunit blocking antibody display annexin V immunoreactivity, a sign of early apoptosis. Time-lapse videomicroscopy demonstrates that the removal by apoptosis of a single member of a spermatogenic cell cohort inter-connected by cytoplasmic bridges does not affect the remaining members of the cohort. During spermatogenic cell apoptosis, integrin beta1, alpha3, and alpha6 subunits, and tetraspanins CD9 and C81 become displaced away from the developing apoptotic bodies. In contrast, the intermediate filament protein Sak57, a keratin 5 ortholog, concentrates in the developing apoptotic bodies. We propose that the redistribution of integrin-tetraspanin complexes during spermatogenic cell apoptosis may be evidence of a signaling cascade initiated by Sertoli cell geometric changes. As a result, Sertoli cell reduction in surface area may be a limiting factor of spermatogenic cell survival and in the developmental regulation of spermatogenic cell progenies in the intact seminiferous epithelium.


Subject(s)
Antigens, CD/metabolism , Apoptosis/physiology , Integrins/metabolism , Spermatogonia/cytology , Spermatogonia/metabolism , ADAM Proteins/metabolism , Animals , Cells, Cultured , Fusion Regulatory Protein-1/metabolism , Integrases , Integrin alpha3/metabolism , Integrin alpha6/metabolism , Integrin beta1/metabolism , Keratin-5 , Keratins/metabolism , Male , Membrane Glycoproteins/metabolism , Protein Structure, Tertiary , Rats , Sertoli Cells/cytology , Sertoli Cells/metabolism , Tetraspanin 28 , Tetraspanin 29 , Viral Proteins
11.
Mol Reprod Dev ; 72(2): 259-71, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16015579

ABSTRACT

Keratin 9 (K9) is one of the components of the perinuclear ring of the manchette found in developing spermatids but is predominantly expressed in the epidermis of the footpad (palm and sole in human epidermis). As an initial step to determine the function of K9 protein in sperm development, we have generated a mutant mouse by homologous recombination of the targeting vector containing the disrupted K9 gene in which the neo(r) gene was inserted into the intron 6. This insertion resulted in the expression of two K9 mRNAs: a wild-type K9 mRNA, in which intron 6 with the neo(r) gene was completely spliced out, and a mutated form in which only a portion of the intron 6 between neo(r) gene and exon 7 was spliced out. While both heterozygous (K9(+/neo)) and homozygous (K9(neo/neo)) mutant mice expressed the wild-type form of K9 protein, the expression profile of the wild-type K9 in K9(neo/neo) mutants was modified. In addition, the open reading frame of the aberrant mRNA terminated at the exon 6/intron 6 splice site, resulting in a truncated K9 protein. Both K9(+neo) and K9(neo/neo) male mice displayed spermatids with ectopic manchette. Coiled tails were seen in maturing spermatids and epididymal sperm of mutant mice and sperm with deformed tails displayed forward motility. A predominant sperm anomaly was residual cytoplasm at the end of the mitochondria-containing middle piece tail segment. The residual cytoplasm displayed vesicles with random in situ motion, suggesting a transport impediment toward the distal end of the sperm tail. All mutant mice were fertile. Surprisingly, in oocyte nuclear injection experiments using K9(neo/neo) sperm donor, 76% of the resulting animals displayed a deletion of the neo(r) gene from the intron 6 of the mutated K9 allele. Results of this study support the view that intron 6 influences the transcriptional efficiency of the K9 gene by decreasing production of wild-type K9 and changing the expression of K9 proteins.


Subject(s)
Introns/genetics , Keratins/genetics , Mutagenesis, Insertional/genetics , Neomycin/pharmacology , Sperm Tail/metabolism , Animals , Base Sequence , Chromosomes, Mammalian/genetics , Cloning, Molecular , DNA, Complementary/genetics , Heterozygote , Homozygote , Keratins/metabolism , Male , Mice , Mice, Transgenic , Microscopy, Electron , Phenotype , Polymorphism, Genetic/genetics , Sperm Injections, Intracytoplasmic , Sperm Tail/ultrastructure , Testis/abnormalities , Testis/cytology , Testis/drug effects , Testis/metabolism
12.
Biol Reprod ; 70(5): 1400-10, 2004 May.
Article in English | MEDLINE | ID: mdl-14724135

ABSTRACT

Acrosome biogenesis involves the transport and fusion of Golgi-derived proacrosomal vesicles along the acroplaxome, an F-actin/keratin 5-containing cytoskeletal plate anchored to the spermatid nucleus. A significant issue is whether the acroplaxome develops in acrosomeless mutant mice. Male mice with a Hrb null mutation are infertile and both spermatids and sperm are round-headed and lack an acrosome. Hrb, a protein that contains several NPF motifs (Asn-Pro-Phe) and interacts with proteins with Eps15 homology domains, is regarded as critical for the docking and/or fusion of Golgi-derived proacrosomal vesicles. Here we report that the lack of an acrosome in Hrb mutant spermatids does not prevent the development of the acroplaxome. Yet the acroplaxome in the mutant contains F-actin but is deficient in keratin 5. We also show that the actin-based motor protein myosin Va and its receptor, Rab27a/b, known to be involved in vesicle transport, are present in the Golgi and Golgi-derived proacrosomal vesicles in wild-type and Hrb mutant mouse spermatids. In the Hrb mutant, myosin-Va-bound proacrosome vesicles tether to the acroplaxome, where they flatten and form a flat sac, designated pseudoacrosome. As spermiogenesis advances, round-shaped spermatid nuclei of the mutant display several nuclear protrusions, designated nucleopodes. Nucleopodes are consistently found at the acroplaxome- pseudoacrosome site. Our findings support the interpretation that the acroplaxome provides a focal point for myosin-Va/ Rab27a/b-driven proacrosomal vesicles to accumulate, coalesce, and form an acrosome in wild-type spermatids and a pseudoacrosome in Hrb mutant spermatids. We suggest that nucleopodes develop at a site where a keratin 5-deficient acroplaxome may not withstand tension forces operating during spermatid nuclear shaping.


Subject(s)
Acrosome/physiology , Carrier Proteins/genetics , Golgi Apparatus/metabolism , Mutation , Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , Spermatids/physiology , rab GTP-Binding Proteins/metabolism , Actins/metabolism , Adaptor Proteins, Vesicular Transport , Animals , Fluorescent Antibody Technique, Indirect , Immunohistochemistry , Keratin-15 , Keratin-5 , Keratins/deficiency , Male , Mice , Microscopy, Electron , Microscopy, Immunoelectron , Spermatids/ultrastructure , rab27 GTP-Binding Proteins
13.
Mol Biol Cell ; 14(11): 4628-40, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14551252

ABSTRACT

Nuclear shaping is a critical event during sperm development as demonstrated by the incidence of male infertility associated with abnormal sperm ad shaping. Herein, we demonstrate that mouse and rat spermatids assemble in the subacrosomal space a cytoskeletal scaffold containing F-actin and Sak57, a keratin ortholog. The cytoskeletal plate, designated acroplaxome, anchors the developing acrosome to the nuclear envelope. The acroplaxome consists of a marginal ring containing keratin 5 10-nm-thick filaments and F-actin. The ring is closely associated with the leading edge of the acrosome and to the nuclear envelope during the elongation of the spermatid head. Anchorage of the acroplaxome to the gradually shaping nucleus is not disrupted by hypotonic treatment and brief Triton X-100 extraction. By examining spermiogenesis in the azh mutant mouse, characterized by abnormal spermatid/sperm head shaping, we have determined that a deformity of the spermatid nucleus is restricted to the acroplaxome region. These findings lead to the suggestion that the acroplaxome nucleates an F-actin-keratin-containing assembly with the purpose of stabilizing and anchoring the developing acrosome during spermatid nuclear elongation. The acroplaxome may also provide a mechanical planar scaffold modulating external clutching forces generated by a stack of Sertoli cell F-actin-containing hoops encircling the elongating spermatid nucleus.


Subject(s)
Acrosome/metabolism , Actins/metabolism , Cell Nucleus/metabolism , Keratins/metabolism , Spermatids/metabolism , Acrosome/ultrastructure , Amino Acid Sequence , Animals , Cell Nucleus/ultrastructure , Cloning, Molecular , Fluorescent Antibody Technique , Gene Library , Humans , Keratin-15 , Keratin-5 , Male , Mice , Microscopy, Immunoelectron , Molecular Sequence Data , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Rats , Sequence Analysis, Protein , Skin/cytology , Skin/metabolism , Spermatids/ultrastructure , Spermatogenesis/physiology , Testis/cytology , Testis/metabolism
14.
Dev Dyn ; 227(3): 458-67, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12815633

ABSTRACT

Immediately after birth, primordial germinal cell-derived prespermatogonia (PSG), located in the center of the testicular cords, migrate between adjacent Sertoli cells to establish contact with the cord basal lamina. PSG migration suggests continued assembly and disassembly of cell-cell contacts by a molecular mechanism that may involve integrins and their ligands, the disintegrin domain of spermatogenic cell-specific plasma membrane proteins called ADAMs. We have analyzed the temporal gene expression of selected ADAMs in intact fetal, early postnatal, and pubertal rat testis and Sertoli-spermatogenic cell cocultures by reverse transcriptase-polymerase chain reaction, in situ hybridization, and immunocytochemistry. We report that several ADAM transcripts are expressed in fetal, neonatal, and prepubertal testes. Cyritestin (ADAM3), ADAM5, ADAM6, and ADAM15 are expressed in day 17 fetal testes. In contrast, no expression of fertilin alpha (ADAM1) and fertilin beta (ADAM 2) was detected in fetal testes. Fertilin beta gene expression starts after postnatal day 2, subsequent to the expression of fertilin alpha, which occurs on postnatal day 1. After postnatal day 2, all the indicated ADAMs, including the fertilin alpha and fertilin beta, continue to be expressed. Transcripts of spermatogenic cell-specific fertilin alpha, fertilin beta, ADAM3, and ADAM5 were detected during the coculture of PSG with Sertoli cells for up to 72 hr after plating. The presence of fertilin beta mRNA and protein in cocultured PSG was visualized by in situ hybridization and immunocytochemistry, respectively. These observations indicate that PSG in coculture with Sertoli cells provide a suitable approach for analyzing cell-cell adhesive responses involving spermatogenic cell-specific ADAMs.


Subject(s)
Gene Expression Regulation, Developmental , Membrane Glycoproteins/biosynthesis , Metalloendopeptidases/biosynthesis , Spermatogonia/metabolism , Testis/embryology , ADAM Proteins , Animals , Cell Adhesion , Cell Membrane/metabolism , Cell Movement , Coculture Techniques , Fertilins , Fluorescent Antibody Technique, Indirect , Immunohistochemistry , In Situ Hybridization , Ligands , Male , Membrane Glycoproteins/physiology , Metalloendopeptidases/physiology , Protein Structure, Tertiary , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sertoli Cells/cytology , Testis/cytology , Time Factors
15.
Mol Reprod Dev ; 63(1): 131-40, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12211070

ABSTRACT

Ran, a Ras-related GTPase, is required for transporting proteins in and out of the nucleus during interphase and for regulating the assembly of microtubules. cDNA cloning shows that rat testis, like mouse testis, expresses both somatic and testis-specific forms of Ran-GTPase. The presence of a homologous testis-specific form of Ran-GTPase in rodents implies that the Ran-GTPase pathway plays a significant role during sperm development. This suggestions is supported by distinct Ran-GTPase immunolocalization sites identified in developing spermatids. Confocal microscopy demonstrates that Ran-GTPase localizes in the nucleus of round spermatids and along the microtubules of the manchette in elongating spermatids. When the manchette disassembles, Ran-GTPase immunoreactivity is visualized in the centrosome region of maturing spermatids. The circumstantial observation that fractionated manchettes, containing copurified centrin-immunoreactive centrosomes, can organize a three-dimensional lattice in the presence of taxol and GTP, points to the role of Ran-GTPase and associated factors in microtubule nucleation as well as the potential nucleating function of spermatid centrosomes undergoing a reduction process. Electron microscopy demonstrates the presence in manchette preparations of spermatid centrosomes, recognized as such by their association with remnants of the implantation fossa, a dense plate observed only at the basal surface of developing spermatid and sperm nuclei. In addition, we have found importin beta1 immunoreactivity in the nucleus of elongating spermatids, a finding that, together with the presence of Ran-GTPase in the nucleus of round spermatids and the manchette, suggest a potential role of Ran-GTPase machinery in nucleocytoplasmic transport. Our expression and localization analysis, correlated with functional observations in other cell systems, suggest that Ran-GTPase may be involved in both nucleocytoplasmic transport and microtubules assembly, two critical events during the development of functional sperm. In addition, the manchette-to-centrosome Ran-GTPase relocation, together with the similar redistribution of various proteins associated to the manchette, suggest the existence of an intramanchette molecular transport mechanism, which may share molecular analogies with intraflagellar transport.


Subject(s)
Centrosome/enzymology , Microtubules/physiology , Spermatogenesis/physiology , Spermatozoa/enzymology , ran GTP-Binding Protein/physiology , Amino Acid Sequence , Animals , Cell Nucleus/enzymology , Cell Nucleus/ultrastructure , Centrosome/physiology , Centrosome/ultrastructure , Cloning, Molecular , DNA, Complementary/genetics , Guanosine Triphosphate/pharmacology , Isoenzymes/physiology , Male , Molecular Sequence Data , Paclitaxel/pharmacology , Protein Transport/physiology , Rats , Sequence Alignment , Sequence Homology, Amino Acid , Sperm Head/enzymology , Sperm Head/physiology , Sperm Head/ultrastructure , Spermatozoa/drug effects , Spermatozoa/physiology , Spermatozoa/ultrastructure , Testis/enzymology , beta Karyopherins/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...