Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
PLoS Genet ; 13(11): e1007078, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29136647

RESUMEN

The katanin microtubule-severing proteins are essential regulators of microtubule dynamics in a diverse range of species. Here we have defined critical roles for the poorly characterised katanin protein KATNAL2 in multiple aspects of spermatogenesis: the initiation of sperm tail growth from the basal body, sperm head shaping via the manchette, acrosome attachment, and ultimately sperm release. We present data suggesting that depending on context, KATNAL2 can partner with the regulatory protein KATNB1 or act autonomously. Moreover, our data indicate KATNAL2 may regulate δ- and ε-tubulin rather than classical α-ß-tubulin microtubule polymers, suggesting the katanin family has a greater diversity of function than previously realised. Together with our previous research, showing the essential requirement of katanin proteins KATNAL1 and KATNB1 during spermatogenesis, our data supports the concept that in higher order species the presence of multiple katanins has allowed for subspecialisation of function within complex cellular settings such as the seminiferous epithelium.


Asunto(s)
Katanina/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos/genética , Animales , Células Germinativas/metabolismo , Haploidia , Infertilidad Masculina/metabolismo , Katanina/genética , Masculino , Ratones , Microtúbulos/metabolismo , Isoformas de Proteínas , Epitelio Seminífero/metabolismo , Espermatogénesis/genética , Espermatozoides/metabolismo , Testículo/metabolismo , Tubulina (Proteína)/metabolismo
2.
FASEB J ; 31(3): 1141-1152, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28003339

RESUMEN

Infertility occurs in 1 in 20 young men and is idiopathic in origin in most. We have reported that the leucine-rich repeat (LRR) and guanylate kinase-like domain containing, isoform (LRGUK)-1 is essential for sperm head shaping, via the manchette, and the initiation of sperm tail growth from the centriole/basal body, and thus, male fertility. Within this study we have used a yeast 2-hybrid screen of an adult testis library to identify LRGUK1-binding partners, which were then validated with a range of techniques. The data indicate that LRGUK1 likely achieves its function in partnership with members of the HOOK family of proteins (HOOK-1-3), Rab3-interacting molecule binding protein (RIMBP)-3 and kinesin light chain (KLC)-3, all of which are associated with intracellular protein transport as cargo adaptor proteins and are localized to the manchette. LRGUK1 consists of 3 domains; an LRR, a guanylate kinase (GUK)-like and an unnamed domain. In the present study, we showed that the GUK-like domain is essential for binding to HOOK2 and RIMBP3, and the LRR domain is essential for binding to KLC3. These findings establish LRGUK1 as a key component of a multiprotein complex with an essential role in microtubule dynamics within haploid male germ cells.-Okuda, H., DeBoer, K., O'Connor, A. E., Merriner, D. J., Jamsai, D., O'Bryan, M. K. LRGUK1 is part of a multiprotein complex required for manchette function and male fertility.


Asunto(s)
Guanilato-Quinasas/metabolismo , Infertilidad Masculina/metabolismo , Espermátides/metabolismo , Animales , Sitios de Unión , Línea Celular , Células Cultivadas , Proteínas de Unión al GTP/metabolismo , Guanilato-Quinasas/química , Células HEK293 , Humanos , Infertilidad Masculina/genética , Cinesinas , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Unión Proteica , Transporte de Proteínas , Ratas
3.
PLoS Genet ; 11(10): e1005620, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26496356

RESUMEN

piRNAs are critical for transposable element (TE) repression and germ cell survival during the early phases of spermatogenesis, however, their role in adult germ cells and the relative importance of piRNA methylation is poorly defined in mammals. Using a mouse model of HEN methyltransferase 1 (HENMT1) loss-of-function, RNA-Seq and a range of RNA assays we show that HENMT1 is required for the 2' O-methylation of mammalian piRNAs. HENMT1 loss leads to piRNA instability, reduced piRNA bulk and length, and ultimately male sterility characterized by a germ cell arrest at the elongating germ cell phase of spermatogenesis. HENMT1 loss-of-function, and the concomitant loss of piRNAs, resulted in TE de-repression in adult meiotic and haploid germ cells, and the precocious, and selective, expression of many haploid-transcripts in meiotic cells. Precocious expression was associated with a more active chromatin state in meiotic cells, elevated levels of DNA damage and a catastrophic deregulation of the haploid germ cell gene expression. Collectively these results define a critical role for HENMT1 and piRNAs in the maintenance of TE repression in adult germ cells and setting the spermatogenic program.


Asunto(s)
Infertilidad Masculina/genética , Metiltransferasas/genética , Estabilidad del ARN/genética , ARN Interferente Pequeño/genética , Espermatogénesis/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cromatina/genética , Elementos Transponibles de ADN/genética , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/crecimiento & desarrollo , Humanos , Infertilidad Masculina/patología , Masculino , Ratones
4.
PLoS Genet ; 11(3): e1005090, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25781171

RESUMEN

Male infertility affects at least 5% of reproductive age males. The most common pathology is a complex presentation of decreased sperm output and abnormal sperm shape and motility referred to as oligoasthenoteratospermia (OAT). For the majority of OAT men a precise diagnosis cannot be provided. Here we demonstrate that leucine-rich repeats and guanylate kinase-domain containing isoform 1 (LRGUK-1) is required for multiple aspects of sperm assembly, including acrosome attachment, sperm head shaping and the initiation of the axoneme growth to form the core of the sperm tail. Specifically, LRGUK-1 is required for basal body attachment to the plasma membrane, the appropriate formation of the sub-distal appendages, the extension of axoneme microtubules and for microtubule movement and organisation within the manchette. Manchette dysfunction leads to abnormal sperm head shaping. Several of these functions may be achieved in association with the LRGUK-1 binding partner HOOK2. Collectively, these data establish LRGUK-1 as a major determinant of microtubule structure within the male germ line.


Asunto(s)
Guanilato-Quinasas/metabolismo , Infertilidad Masculina/metabolismo , Espermatogénesis , Espermatozoides/metabolismo , Secuencia de Aminoácidos , Animales , Cuerpos Basales/metabolismo , Membrana Celular/metabolismo , Guanilato-Quinasas/química , Guanilato-Quinasas/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Datos de Secuencia Molecular , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Alineación de Secuencia , Espermatozoides/citología , Testículo/citología , Testículo/metabolismo
5.
Dev Dyn ; 246(12): 1027-1035, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28791777

RESUMEN

BACKGROUND: The microtubule-severing protein complex katanin is composed two subunits, the ATPase subunit, KATNA1, and the noncatalytic regulatory subunit, KATNB1. Recently, the Katnb1 gene has been linked to infertility, regulation of centriole and cilia formation in fish and mammals, as well as neocortical brain development. KATNB1 protein is expressed in germ cells in humans and mouse, mitotic/meiotic spindles and cilia, although the full expression pattern of the Katnb1 gene has not been described. RESULTS: Using a knockin-knockout mouse model of Katnb1 dysfunction we demonstrate that Katnb1 is ubiquitously expressed during embryonic development, although a stronger expression is seen in the crown cells of the gastrulation organizer, the murine node. Furthermore, null and hypomorphic Katnb1 gene mutations show a novel correlation between Katnb1 dysregulation and the development of impaired left-right signaling, including cardiac malformations. CONCLUSIONS: Katanin function is a critical regulator of heart development in mice. These findings are potentially relevant to human cardiac development. Developmental Dynamics 246:1027-1035, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Cardiopatías Congénitas , Katanina , Mutación , Transducción de Señal/genética , Animales , Técnicas de Sustitución del Gen , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Katanina/genética , Katanina/metabolismo , Ratones , Ratones Noqueados
6.
FASEB J ; 30(8): 2777-91, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27105888

RESUMEN

Spermatozoa require the process of capacitation to enable them to fertilize an egg. PKA is crucial to capacitation and the development of hyperactivated motility. Sperm PKA is activated by cAMP generated by the germ cell-enriched adenylyl cyclase encoded by Adcy10 Male mice lacking Adcy10 are sterile, because their spermatozoa are immotile. The current study was designed to identify binding partners of the sperm-specific (Cα2) catalytic subunit of PKA (PRKACA) by using it as the "bait" in a yeast 2-hybrid system. This approach was used to identify a novel germ cell-enriched protein, sperm PKA interacting factor (SPIF), in 25% of the positive clones. Homozygous Spif-null mice were embryonically lethal. SPIF was coexpressed and coregulated with PRKACA and with t-complex protein (TCP)-11, a protein associated with PKA signaling. We established that these 3 proteins form part of a novel complex in mouse spermatozoa. Upon capacitation, the SPIF protein becomes tyrosine phosphorylated in >95% of sperm. An apparent molecular rearrangement in the complex occurs, bringing PRKACA and TCP11 into proximity. Taken together, these results suggest a role for the novel complex of SPIF, PRKACA, and TCP11 during sperm capacitation, fertilization, and embryogenesis.-Stanger, S. J., Law, E. A., Jamsai, D., O'Bryan, M. K., Nixon, B., McLaughlin, E. A., Aitken, R. J., Roman, S. D. A novel germ cell protein, SPIF (sperm PKA interacting factor), is essential for the formation of a PKA/TCP11 complex that undergoes conformational and phosphorylation changes upon capacitation.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de la Membrana/metabolismo , Capacitación Espermática/fisiología , Espermatozoides/fisiología , Animales , Proteínas Portadoras/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Femenino , Regulación de la Expresión Génica/fisiología , Heterocigoto , Masculino , Proteínas de la Membrana/genética , Ratones , Fosforilación , Conformación Proteica , Isoformas de Proteínas , Subunidades de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Razón de Masculinidad , Técnicas del Sistema de Dos Híbridos
7.
PLoS Genet ; 9(7): e1003628, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935508

RESUMEN

Alternative splicing of precursor messenger RNA (pre-mRNA) is common in mammalian cells and enables the production of multiple gene products from a single gene, thus increasing transcriptome and proteome diversity. Disturbance of splicing regulation is associated with many human diseases; however, key splicing factors that control tissue-specific alternative splicing remain largely undefined. In an unbiased genetic screen for essential male fertility genes in the mouse, we identified the RNA binding protein RBM5 (RNA binding motif 5) as an essential regulator of haploid male germ cell pre-mRNA splicing and fertility. Mice carrying a missense mutation (R263P) in the second RNA recognition motif (RRM) of RBM5 exhibited spermatid differentiation arrest, germ cell sloughing and apoptosis, which ultimately led to azoospermia (no sperm in the ejaculate) and male sterility. Molecular modelling suggested that the R263P mutation resulted in compromised mRNA binding. Within the adult mouse testis, RBM5 localises to somatic and germ cells including spermatogonia, spermatocytes and round spermatids. Through the use of RNA pull down coupled with microarrays, we identified 11 round spermatid-expressed mRNAs as putative RBM5 targets. Importantly, the R263P mutation affected pre-mRNA splicing and resulted in a shift in the isoform ratios, or the production of novel spliced transcripts, of most targets. Microarray analysis of isolated round spermatids suggests that altered splicing of RBM5 target pre-mRNAs affected expression of genes in several pathways, including those implicated in germ cell adhesion, spermatid head shaping, and acrosome and tail formation. In summary, our findings reveal a critical role for RBM5 as a pre-mRNA splicing regulator in round spermatids and male fertility. Our findings also suggest that the second RRM of RBM5 is pivotal for appropriate pre-mRNA splicing.


Asunto(s)
Empalme Alternativo/genética , Diferenciación Celular/genética , Infertilidad Masculina/genética , Motivos de Nucleótidos/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Animales , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Células Germinativas/patología , Humanos , Infertilidad Masculina/patología , Masculino , Ratones , Modelos Moleculares , Mutación , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química , Espermátides/metabolismo , Espermátides/patología , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética
8.
Biol Reprod ; 92(4): 99, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25715794

RESUMEN

The endometrium (the mucosal lining of the uterus) is a dynamic tissue that undergoes extensive remodeling, secretory transformation in preparation for implantation of an embryo, inflammatory and proteolytic activity during menstruation, and rapid postmenstrual repair. A plethora of local factors influence these processes. Recently, a cysteine-rich protein, CRISP3, a clade of the CRISP, antigen 5, pathogenesis-related (CAP) protein superfamily, has been implicated in uterine function. The localization, regulation, and potential function of CRISP3 in both the human and mouse endometrium is described. CRISP3 localizes to the luminal and glandular epithelium of the endometrium within both species, with increased immunoreactivity during the proliferative phase of the human cycle. CRISP3 also localizes to neutrophils, particularly within the premenstrual human endometrium and during the postbreakdown repair phase of a mouse model of endometrial breakdown and repair. Endometrial CRISP3 is produced by primary human endometrial epithelial cells and secreted in vivo to accumulate in the uterine cavity. Secreted CRISP3 is more abundant in uterine lavage fluid during the proliferative phase of the menstrual cycle. Human endometrial epithelial CRISP3 is present in both a glycosylated and a nonglycosylated form in vitro and in vivo. Treatment of endometrial epithelial cells in vitro with recombinant CRISP3 enhances both adhesion and proliferation. These data suggest roles for epithelial and neutrophil-derived CRISP3 in postmenstrual endometrial repair and regeneration.


Asunto(s)
Adhesión Celular/fisiología , Endometrio/fisiología , Células Epiteliales/fisiología , Ciclo Estral/fisiología , Ciclo Menstrual/fisiología , Proteínas y Péptidos Salivales/biosíntesis , Proteínas de Plasma Seminal/biosíntesis , Adulto , Animales , Proliferación Celular , Endometrio/citología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Neutrófilos/metabolismo , Embarazo , Cultivo Primario de Células , Proteínas y Péptidos Salivales/genética , Proteínas de Plasma Seminal/genética
9.
PLoS Genet ; 8(10): e1002969, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23055941

RESUMEN

A significant percentage of young men are infertile and, for the majority, the underlying cause remains unknown. Male infertility is, however, frequently associated with defective sperm motility, wherein the sperm tail is a modified flagella/cilia. Conversely, a greater understanding of essential mechanisms involved in tail formation may offer contraceptive opportunities, or more broadly, therapeutic strategies for global cilia defects. Here we have identified Rab-like 2 (RABL2) as an essential requirement for sperm tail assembly and function. RABL2 is a member of a poorly characterized clade of the RAS GTPase superfamily. RABL2 is highly enriched within developing male germ cells, where it localizes to the mid-piece of the sperm tail. Lesser amounts of Rabl2 mRNA were observed in other tissues containing motile cilia. Using a co-immunoprecipitation approach and RABL2 affinity columns followed by immunochemistry, we demonstrated that within developing haploid germ cells RABL2 interacts with intra-flagella transport (IFT) proteins and delivers a specific set of effector (cargo) proteins, including key members of the glycolytic pathway, to the sperm tail. RABL2 binding to effector proteins is regulated by GTP. Perturbed RABL2 function, as exemplified by the Mot mouse line that contains a mutation in a critical protein-protein interaction domain, results in male sterility characterized by reduced sperm output, and sperm with aberrant motility and short tails. Our data demonstrate a novel function for the RABL protein family, an essential role for RABL2 in male fertility and a previously uncharacterised mechanism for protein delivery to the flagellum.


Asunto(s)
Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Espermatozoides/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Animales , Secuencia de Bases , Secuencia Conservada , Fertilidad/genética , Expresión Génica , Orden Génico , Células Germinativas/metabolismo , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutación , Fenotipo , Unión Proteica , Transporte de Proteínas , Alineación de Secuencia , Motilidad Espermática/genética , Cola del Espermatozoide/metabolismo
10.
PLoS Genet ; 8(5): e1002698, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22654669

RESUMEN

Katanin is an evolutionarily conserved microtubule-severing complex implicated in multiple aspects of microtubule dynamics. Katanin consists of a p60 severing enzyme and a p80 regulatory subunit. The p80 subunit is thought to regulate complex targeting and severing activity, but its precise role remains elusive. In lower-order species, the katanin complex has been shown to modulate mitotic and female meiotic spindle dynamics and flagella development. The in vivo function of katanin p80 in mammals is unknown. Here we show that katanin p80 is essential for male fertility. Specifically, through an analysis of a mouse loss-of-function allele (the Taily line), we demonstrate that katanin p80, most likely in association with p60, has an essential role in male meiotic spindle assembly and dissolution and the removal of midbody microtubules and, thus, cytokinesis. Katanin p80 also controls the formation, function, and dissolution of a microtubule structure intimately involved in defining sperm head shaping and sperm tail formation, the manchette, and plays a role in the formation of axoneme microtubules. Perturbed katanin p80 function, as evidenced in the Taily mouse, results in male sterility characterized by decreased sperm production, sperm with abnormal head shape, and a virtual absence of progressive motility. Collectively these data demonstrate that katanin p80 serves an essential and evolutionarily conserved role in several aspects of male germ cell development.


Asunto(s)
Adenosina Trifosfatasas , Células Germinativas , Infertilidad Masculina/genética , Microtúbulos , Espermatogénesis/genética , Espermatozoides , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Animales , Astenozoospermia/genética , Expresión Génica , Células Germinativas/citología , Células Germinativas/metabolismo , Katanina , Masculino , Meiosis/genética , Ratones , Microtúbulos/genética , Microtúbulos/metabolismo , Datos de Secuencia Molecular , Mutación Missense , Oligospermia/genética , Subunidades de Proteína/genética , Motilidad Espermática/genética , Espermatozoides/citología , Espermatozoides/metabolismo , Espermatozoides/patología , Huso Acromático/genética , Testículo/metabolismo
11.
Proc Natl Acad Sci U S A ; 108(17): 7034-9, 2011 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-21482758

RESUMEN

The cysteine-rich secretory proteins (CRISPs) are a group of four proteins in the mouse that are expressed abundantly in the male reproductive tract, and to a lesser extent in other tissues. Analysis of reptile CRISPs and mouse CRISP2 has shown that CRISPs can regulate cellular homeostasis via ion channels. With the exception of the ability of CRISP2 to regulate ryanodine receptors, the in vivo targets of mammalian CRISPs function are unknown. In this study, we have characterized the ion channel regulatory activity of epididymal CRISP4 using electrophysiology, cell assays, and mouse models. Through patch-clamping of testicular sperm, the CRISP4 CRISP domain was shown to inhibit the transient receptor potential (TRP) ion channel TRPM8. These data were confirmed using a stably transfected CHO cell line. TRPM8 is a major cold receptor in the body, but is found in other tissues, including the testis and on the tail and head of mouse and human sperm. Functional assays using sperm from wild-type mice showed that TRPM8 activation significantly reduced the number of sperm undergoing the progesterone-induced acrosome reaction following capacitation, and that this response was reversed by the coaddition of CRISP4. In accordance, sperm from Crisp4 null mice had a compromised ability to undergo to the progesterone-induced acrosome reaction. Collectively, these data identify CRISP4 as an endogenous regulator of TRPM8 with a role in normal sperm function.


Asunto(s)
Reacción Acrosómica/fisiología , Proteínas de Plasma Seminal/metabolismo , Espermatozoides/metabolismo , Canales Catiónicos TRPM/metabolismo , Reacción Acrosómica/efectos de los fármacos , Animales , Células CHO , Cricetinae , Cricetulus , Humanos , Masculino , Ratones , Ratones Noqueados , Progesterona/farmacología , Progestinas/farmacología , Proteínas de Plasma Seminal/genética , Espermatozoides/citología , Canales Catiónicos TRPM/genética
13.
Biol Cell ; 102(2): 93-106, 2009 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-19686095

RESUMEN

BACKGROUND INFORMATION: CRISP2 (cysteine-rich secretory protein 2) is a sperm acrosome and tail protein with the ability to regulate Ca2+ flow through ryanodine receptors. Based on these properties, CRISP2 has a potential role in fertilization through the regulation of ion signalling in the acrosome reaction and sperm motility. The purpose of the present study was to determine the expression, subcellular localization and the role in spermatogenesis of a novel CRISP2-binding partner, which we have designated SHTAP (sperm head and tail associated protein). RESULTS: Using yeast two-hybrid screens of an adult testis expression library, we identified SHTAP as a novel mouse CRISP2-binding partner. Sequence analysis of all Shtap cDNA clones revealed that the mouse Shtap gene is embedded within a gene encoding the unrelated protein NSUN4 (NOL1/NOP2/Sun domain family member 4). Five orthologues of the Shtap gene have been annotated in public databases. SHTAP and its orthologues showed no significant sequence similarity to any known protein or functional motifs, including NSUN4. Using an SHTAP antiserum, multiple SHTAP isoforms (approximately 20-87 kDa) were detected in the testis, sperm, and various somatic tissues. Interestingly, only the approximately 26 kDa isoform of SHTAP was able to interact with CRISP2. Furthermore, yeast two-hybrid assays showed that both the CAP (CRISP/antigen 5/pathogenesis related-1) and CRISP domains of CRISP2 were required for maximal binding to SHTAP. SHTAP protein was localized to the peri-acrosomal region of round spermatids, and the head and tail of the elongated spermatids and sperm tail where it co-localized with CRISP2. During sperm capacitation, SHTAP and the SHTAP-CRISP2 complex appeared to be redistributed within the head. CONCLUSIONS: The present study is the first report of the identification, annotation and expression analysis of the mouse Shtap gene. The redistribution observed during sperm capacitation raises the possibility that SHTAP and the SHTAP-CRISP2 complex play a role in the attainment of sperm functional competence.


Asunto(s)
Proteínas Portadoras/metabolismo , Glicoproteínas/metabolismo , Espermatogénesis , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Proteínas Portadoras/genética , Moléculas de Adhesión Celular , Secuencia Conservada , Humanos , Masculino , Proteínas de la Membrana , Metiltransferasas , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Alineación de Secuencia
14.
Handb Exp Pharmacol ; (198): 179-94, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20839092

RESUMEN

The production of functional spermatozoa is a complex process requiring the coordinated expression of thousands of genes. It is likely that the intricate nature of these interactions contributes to the large number of idiopathic male infertility cases seen in humans. Conversely, the complexity of the highly regulated and interconnected processes of spermatogenesis and posttesticular sperm maturation events offers opportunities for the development of male-based contraceptive targets. The recent advances in genetic manipulation technologies and the completion of the human and mouse genome sequencing programs have provided scientists with sophisticated ways to generate mouse models for the study of basic biological mechanisms, in order to understand disease pathology and develop novel therapeutic approaches. The three common types of mouse model used for medical research are transgenic, knockout/knockin, and chemical-induced point mutant mice. Each type has relative strengths and weaknesses with respect to its fidelity to the disease processes in humans. In this chapter, we focus on the utility of the different types of mouse model in obtaining a better understanding of the mechanisms that control spermatogenesis and developing male-based contraceptive regimens.


Asunto(s)
Anticonceptivos Masculinos/farmacología , Fertilidad/efectos de los fármacos , Fertilidad/genética , Ratones/fisiología , Animales , Humanos , Masculino , Ratones Noqueados , Ratones Transgénicos , Mutagénesis , Mutación Puntual
15.
Reproduction ; 135(6): 751-9, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18502891

RESUMEN

Cysteine-rich secretory protein 2 (CRISP2) is a testis-enriched protein localized to the sperm acrosome and tail. CRISP2 has been proposed to play a critical role in spermatogenesis and male fertility, although the precise function(s) of CRISP2 remains to be determined. Recent data have shown that the CRISP domain of the mouse CRISP2 has the ability to regulate Ca(2+) flow through ryanodine receptors (RyR) and to bind to MAP kinase kinase kinase 11 (MAP3K11). To further define the biochemical pathways within which CRISP2 is involved, we screened an adult mouse testis cDNA library using a yeast two-hybrid assay to identify CRISP2 interacting partners. One of the most frequently identified CRISP2-binding proteins was gametogenetin 1 (GGN1). Interactions occur between the ion channel regulatory region within the CRISP2 CRISP domain and the carboxyl-most 158 amino acids of GGN1. CRISP2 does not bind to the GGN2 or GGN3 isoforms. Furthermore, we showed that Ggn1 is a testis-enriched mRNA and the protein first appeared in late pachytene spermatocytes and was up-regulated in round spermatids before being incorporated into the principal piece of the sperm tail where it co-localized with CRISP2. These data along with data on RyR and MAP3K11 binding define the CRISP2 CRISP domain as a protein interaction motif and suggest a role for the GGN1-CRISP2 complex in sperm tail development and/or motility.


Asunto(s)
Glicoproteínas/análisis , Cola del Espermatozoide/química , Hormonas Testiculares/análisis , Testículo/química , Acrosoma/química , Acrosoma/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Northern Blotting/métodos , Western Blotting/métodos , Moléculas de Adhesión Celular , Clonación Molecular , Ingeniería Genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Inmunohistoquímica , Masculino , Proteínas de la Membrana , Ratones , Datos de Secuencia Molecular , Unión Proteica , Motilidad Espermática/fisiología , Cola del Espermatozoide/metabolismo , Espermátides/química , Espermátides/metabolismo , Espermatocitos/química , Espermatocitos/metabolismo , Espermatogénesis/fisiología , Hormonas Testiculares/genética , Hormonas Testiculares/metabolismo , Testículo/metabolismo , Técnicas del Sistema de Dos Híbridos
16.
Sci Rep ; 7(1): 16323, 2017 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-29176597

RESUMEN

Cigarette smoking is undoubtedly a risk factor for lung cancer. Moreover, smokers with genetic mutations on chromosome 3p21.3, a region frequently deleted in cancer and notably in lung cancer, have a dramatically higher risk of aggressive lung cancer. The RNA binding motif 5 (RBM5) is one of the component genes in the 3p21.3 tumour suppressor region. Studies using human cancer specimens and cell lines suggest a role for RBM5 as a tumour suppressor. Here we demonstrate, for the first time, an in vivo role for RBM5 as a tumour suppressor in the mouse lung. We generated Rbm5 loss-of-function mice and exposed them to a tobacco carcinogen NNK. Upon exposure to NNK, Rbm5 loss-of-function mice developed lung cancer at similar rates to wild type mice. As tumourigenesis progressed, however, reduced Rbm5 expression lead to significantly more aggressive lung cancer i.e. increased adenocarcinoma nodule numbers and tumour size. Our data provide in vivo evidence that reduced RBM5 function, as occurs in a large number of patients, coupled with exposure to tobacco carcinogens is a risk factor for an aggressive lung cancer phenotype. These data suggest that RBM5 loss-of-function likely underpins at least part of the pro-tumourigenic consequences of 3p21.3 deletion in humans.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Neoplasias Pulmonares/metabolismo , Pulmón/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Genes Supresores de Tumor/fisiología , Humanos , Pulmón/patología , Neoplasias Pulmonares/patología , Masculino , Ratones , Proteínas de Unión al ARN/genética , Proteínas Supresoras de Tumor/genética
17.
Biochim Biophys Acta ; 1728(3): 150-62, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15820143

RESUMEN

We report the generation and characterisation of the first transgenic mice exclusively expressing normal human beta-globin ((hu)beta-globin) from a 183 kb genomic fragment. Four independent lines were generated, each containing 2-6 copies of the (hu)beta-globin locus at a single integration site. Steady state levels of (hu)beta-globin protein were dependent on transgene copy number, but independent of the site of integration. Hemizygosity for the transgene on a heterozygous knockout background ((hu)beta(+/0), (mu)beta(th-3/+)) complemented fully the hematological abnormalities associated with the heterozygous knockout mutation in all four lines. Importantly, the rescue of the embryonic lethal phenotype that is characteristic of homozygosity for the knockout mutation was also demonstrated in two transgenic lines that were homozygous for two copies of the (hu)beta-globin locus, and in one transgenic line, which was hemizygous for six copies of the (hu)beta-globin locus. Our results illustrate the importance of transgene copy number determination and of the hemizygosity/homozygosity status in phenotypic complementation studies of transgenic mice containing large heterologous transgenes. Transgenic mouse colonies with 100% (hu)beta-globin production from the intact (hu)beta-globin locus have been established and will be invaluable in comparative and gene therapy studies with mouse models containing specific beta-thalassemia mutations in the (hu)beta-globin locus.


Asunto(s)
Cromosomas Artificiales Bacterianos/genética , Modelos Animales de Enfermedad , Globinas/genética , Globinas/metabolismo , Ratones/genética , Fenotipo , Transgenes/genética , Animales , Southern Blotting , Cromatografía Líquida de Alta Presión , Cartilla de ADN , Electroforesis en Acetato de Celulosa , Componentes del Gen , Prueba de Complementación Genética , Hemoglobinas/genética , Heterocigoto , Humanos , Hibridación Fluorescente in Situ , Ratones Noqueados , Ratones Transgénicos , Microinyecciones , Talasemia beta/genética
18.
Int J Hematol ; 83(3): 229-37, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16720553

RESUMEN

Hereditary persistence of fetal hemoglobin (HPFH) is associated with a high level of hemoglobin F (HbF) synthesis in adult heterozygotes. In this study, 2 of 6 unrelated HPFH Thai families were found to be Southeast Asian-type HPFH (SEA-HPFH) by analyses of the hematologic data and Southern blot hybridization with polymerase chain reaction-amplified DNA probes. DNA mapping with a probe for a delta-globin fragment showed a 27-kb deletion of DNA that included the beta-globin gene and the 3' deoxyribonuclease I hypersensitive site 1 (3'HS1) sequence downstream. Deletion of the insulator, 3'HS1, and the juxta-position of the HPFH-3 core enhancer downstream to the 3' breakpoint have been postulated to be the cause of high HbF production in these individuals. To test this hypothesis, we transfected K562 cells with 4 different bacterial artificial chromosome constructs containing the enhanced green fluorescent protein (EGFP) gene at the position of the Agamma-globin gene (pEBAC/148beta:EGFP). Flow cytometry was used to compare EGFP expression from the pEBAC/148beta:EGFP construct with the HPFH-3 core enhancer immediately 5' to the SEA-HPFH breakpoint (pEnH), from the pEBAC/148beta:EGFP construct with 8 kb of the breakpoint sequence and the HPFH-3 core enhancer (pSEA-HPFH), and from the construct with 3'HS1 followed by the pSEA-HPFH sequence (pSEA-HPFH_3pHS1). The results show that high HbF production in SEA-HPFH occurs from a deletion of the 3'HS1 sequence and the juxtaposition of the HPFH-3 enhancer downstream to the delta-globin gene.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Hemoglobina Fetal/genética , Eliminación de Gen , Globinas/genética , Hemoglobinopatías/genética , Adolescente , Adulto , Cromosomas Artificiales Bacterianos , Análisis Mutacional de ADN , Femenino , Hemoglobina Fetal/biosíntesis , Hemoglobinopatías/metabolismo , Humanos , Células K562 , Masculino , Persona de Mediana Edad , Embarazo , Tailandia , Transfección
19.
Fertil Steril ; 103(1): 214-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25439847

RESUMEN

OBJECTIVE: To assess the association between copy number variations (CNVs) and meiotic arrest and azoospermic men. DESIGN: Genetic association study. SETTING: University. PATIENT(S): Australian men: 19 with histologically confirmed meiotic arrest, 110 men with azoospermia in the absence of histologic data, and 97 fertile men (controls). INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): The identification of CNV by microarray and/or multiplex ligation-dependent probe amplification (MLPA), and the localization of unique CNV encoded proteins to the human testis. RESULT(S): Microarray identified two CNVs unique to meiosis arrest patients. One containing the MYRIP gene and a second containing LRRC4C and the long noncoding RNA LOC100507205. All three genes are transcribed in the human testis, and MYRIP and LRRC4C localize to meiotic cells. The reverse genetic screen for CNVs in meiosis genes identified in mouse models further identified CNVs including HSPA2 as being associated with azoospermia. CONCLUSION(S): These data raise the possibility that, while relatively rare, CNVs may contribute to human male infertility and that CNV screening should be incorporated into long-term plans for genome profiling as a diagnostic tool.


Asunto(s)
Azoospermia/genética , Azoospermia/patología , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Variaciones en el Número de Copia de ADN/genética , Testículo/patología , Testículo/fisiopatología , Estudios de Asociación Genética , Humanos , Masculino
20.
Spermatogenesis ; 5(1): e979061, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26413409

RESUMEN

Male germ cell genome integrity is critical for spermatogenesis, fertility and normal development of the offspring. Several DNA repair pathways exist in male germ cells. One such important pathway is the Fanconi anemia (FANC) pathway. Unlike in somatic cells, expression profiles and the role of the FANC pathway in germ cells remain largely unknown. In this study, we undertook an extensive expression analyses at both mRNA and protein levels of key components of the FANC pathway during spermatogenesis in the mouse. Herein we show that Fanc mRNAs and proteins displayed developmental enrichment within particular male germ cell types. Spermatogonia and pre-leptotene spermatocytes contained the majority of the FANC components examined i.e. complex I members FANCB, FANCG and FANCM, complex II members FANCD2 and FANCI, and complex III member FANCJ. Leptotene, zygotene and early pachytene spermatocytes contained FANCB, FANCG, FANCM and FANCD2. With the exception of FANCL, all FANC proteins examined were not detected in round spermatids. Elongating and elongated spermatids contained FANCB, FANCG, FANCL and FANCJ. qPCR analysis on isolated spermatocytes and round spermatids showed that Fancg, Fancl, Fancm, Fancd2, Fanci and Fancj mRNAs were expressed in both of these germ cell types, indicating that some degree of translational repression of these FANC proteins occurs during the transition from meiosis to spermiogenesis. Taken together, our findings raise the possibility that the assembly of FANC protein complexes in each of the male germ cell type is unique and may be distinct from the proposed model in mitotic cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA