Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where V. Anne Westbrook is active.

Publication


Featured researches published by V. Anne Westbrook.


Journal of Biological Chemistry | 2003

Phosphoproteome Analysis of Capacitated Human Sperm EVIDENCE OF TYROSINE PHOSPHORYLATION OF A KINASE-ANCHORING PROTEIN 3 AND VALOSIN-CONTAINING PROTEIN/p97 DURING CAPACITATION

Scott B. Ficarro; Olga Chertihin; V. Anne Westbrook; Forest M. White; Friederike L. Jayes; Petr Kalab; Jarrod A. Marto; Jeffrey Shabanowitz; John C. Herr; Donald F. Hunt; Pablo E. Visconti

Before fertilization can occur, mammalian sperm must undergo capacitation, a process that requires a cyclic AMP-dependent increase in tyrosine phosphorylation. To identify proteins phosphorylated during capacitation, two-dimensional gel analysis coupled to anti-phosphotyrosine immunoblots and tandem mass spectrometry (MS/MS) was performed. Among the protein targets, valosin-containing protein (VCP), a homolog of the SNARE-interacting protein NSF, and two members of the A kinase-anchoring protein (AKAP) family were found to be tyrosine phosphorylated during capacitation. In addition, immobilized metal affinity chromatography was used to investigate phosphorylation sites in whole protein digests from capacitated human sperm. To increase this chromatographic selectivity for phosphopeptides, acidic residues in peptide digests were converted to their respective methyl esters before affinity chromatography. More than 60 phosphorylated sequences were then mapped by MS/MS, including precise sites of tyrosine and serine phosphorylation of the sperm tail proteins AKAP-3 and AKAP-4. Moreover, differential isotopic labeling was developed to quantify phosphorylation changes occurring during capacitation. The phosphopeptide enrichment and quantification methodology coupled to MS/MS, described here for the first time, can be employed to map and compare phosphorylation sites involved in multiple cellular processes. Although we were unable to determine the exact site of phosphorylation of VCP, we did confirm, using a cross-immunoprecipitation approach, that this protein is tyrosine phosphorylated during capacitation. Immunolocalization of VCP showed fluorescent staining in the neck of noncapacitated sperm. However, after capacitation, staining in the neck decreased, and most of the sperm showed fluorescent staining in the anterior head.


Biology of Reproduction | 2002

SAMP32, a Testis-Specific, Isoantigenic Sperm Acrosomal Membrane-Associated Protein

Zhonglin Hao; Michael J. Wolkowicz; Jagathpala Shetty; Kenneth L. Klotz; Laura C. Bolling; Buer Sen; V. Anne Westbrook; Scott Coonrod; Charles J. Flickinger; John C. Herr

Abstract To identify novel human sperm membrane antigens, we analyzed two-dimensional gels of sperm extracts containing hydrophobic proteins that partitioned into Triton X-114. Four protein spots with isoelectric points (pIs) ranging from 4.5 to 5.5 and apparent molecular weights from 32 to 34 kDa were sequenced by mass spectrometry and found to contain common peptide sequences. Cloning the corresponding cDNA revealed that these protein spots were products of a single gene (SAMP32), encoding a protein of 32 kDa with a predicted pI of 4.57. SAMP32 has a potential transmembrane domain in the carboxyl terminus and is phosphorylated in vivo on serine 256. Northern blotting of eight human tissues and RNA dot blotting of 76 human tissues showed that SAMP32 expression was testis specific. SAMP32 contained an amino terminal domain homologous to the major malarial circumsporozoite surface protein and a domain similar to that of Krp1 from Schizosaccharomyces pombe in its carboxyl terminus. The SAMP32 locus consists of seven exons on chromosome 6q15-16.2. Antiserum against recombinant SAMP32 recognized protein spots originally cored from a two-dimensional gel. This antiserum strongly stained the equatorial segment and faintly stained the acrosome cap of ejaculated human spermatozoa by immunofluorescence. Immunoelectron microscopy showed that SAMP32 was associated with the inner acrosomal membrane in the principal and the equatorial segments of the sperm acrosome. By immunostaining enzyme-dissociated testicular cells, SAMP32 was localized to Golgi phase round spermatids and subsequent stages of acrosome biogenesis. Recombinant SAMP32 reacted with serum from an infertile man, suggesting that it is isoantigenic. Antibodies against recombinant SAMP32 inhibited both the binding and the fusion of human sperm to zona-free hamster eggs.


Biology of Reproduction | 2000

Spermatid-Specific Expression of the Novel X-Linked Gene Product SPAN-X Localized to the Nucleus of Human Spermatozoa

V. Anne Westbrook; Alan B. Diekman; Ken Klotz; Vrinda V. Khole; Chris von Kap-Herr; Wendy L. Golden; Roger L. Eddy; Thomas B. Shows; Mark H. Stoler; Chi-Yu G. Lee; Charles J. Flickinger; John C. Herr

Abstract Formation of mature spermatozoa involves a series of dramatic molecular and morphological changes in the male germ cell lineage. These changes result from the temporally regulated transcription and translation of several testis-specific gene products. Here, we describe a novel, testis-specific protein designated SPAN-X for sperm protein associated with the nucleus on the X chromosome. SPAN-X sequences showed no significant similarity with known cDNA or peptide sequences. The SPAN-X peptide sequences contained three overlapping consensus nuclear localization signals, a high percentage (33%–37%) of charged amino acid residues, and a relatively acidic isoelectric point (pI; 4.88–6.05). Northern analysis of mRNA from multiple human tissues identified a SPAN-X transcript exclusively in the testis. In situ hybridization of human testes sections showed SPAN-X mRNA expression in haploid, round, and elongating spermatids. The SPANX gene was mapped to chromosome Xq27.1 by fluorescence in situ hybridization and by Southern blot analysis of human/mouse somatic cell hybrids. On Western blots of human sperm proteins, antirecombinant SPAN-X antibodies reacted with broad bands migrating between 15–20 kDa. Immunofluorescent labeling of human spermatozoa demonstrated SPAN-X localization to nuclear craters and cytoplasmic droplets. Expression of SPAN-X, an X-linked gene product, exclusively in haploid spermatids leads to interesting questions regarding the transcription of sex-linked genes during spermiogenesis.


Clinical Cancer Research | 2004

Genomic organization, incidence, and localization of the SPAN-x family of cancer-testis antigens in melanoma tumors and cell lines.

V. Anne Westbrook; Pamela D. Schoppee; Alan B. Diekman; Kenneth L. Klotz; Margaretta Allietta; Kevin T. Hogan; Craig L. Slingluff; James W. Patterson; Henry F. Frierson; William P. Irvin; Charles J. Flickinger; Michael A. Coppola; John C. Herr

Purpose: Members of the SPAN-X (sperm protein associated with the nucleus mapped to the X chromosome) family of cancer-testis antigens are promising targets for tumor immunotherapy because they are normally expressed exclusively during spermiogenesis on the adluminal side of the blood-testis barrier, an immune privileged compartment. Experimental Design and Results: This study analyzed the human SPANX genomic organization, as well as SPAN-X mRNA and protein expression in somatic and cancer cells. The SPANX family consists of five genes, one of which is duplicated, all located in a gene cluster at Xq27.1. From the centromere, the arrangement of the five SPANX genes mapped on one contiguous sequence is SPANXB, -C, -A1, -A2, and -D. Reverse transcription-PCR analyses demonstrated expression of SPAN-X mRNA in melanoma and ovarian cell lines, and virtual Northern analysis established SPANX gene expression in numerous cancer cell lines. Immunoblot analysis using polyclonal antisera raised against recombinant SPAN-X confirmed the translation of SPAN-X proteins in melanoma and ovarian tumor cell lines. The immunoreactive proteins migrated between Mr 15,000 and Mr 20,000 similar to those observed in spermatozoa. Immunoperoxidase labeling of melanoma cells and tissue sections demonstrated SPAN-X protein localization in the nucleus, cytoplasm, or both. Ultrastructurally, in melanoma cells with nuclear SPAN-X, the protein was associated with the nuclear envelope, a localization similar to that observed in human spermatids and spermatozoa. Significantly, the incidence of SPAN-X-positive immunostaining was greatest in the more aggressive skin tumors, particularly in distant, nonlymphatic metastatic melanomas. Conclusions: The data herein suggest that the SPAN-X protein may be a useful target in cancer immunotherapy.


Biology of Reproduction | 2001

Differential Nuclear Localization of the Cancer/Testis-Associated Protein, SPAN-X/CTp11, in Transfected Cells and in 50% of Human Spermatozoa

V. Anne Westbrook; Alan B. Diekman; Soren Naaby-Hansen; Kenneth L. Klotz; Theodore Thomas; Elizabeth J. Norton; Charles J. Flickinger; John C. Herr

Abstract Cancer-testis antigens (CTAs) represent potential targets for cancer immunotherapy because these proteins are widely distributed in tumors but not in normal tissues, except testes. In this paper, we identify homology of the CTA CTp11 with SPAN-X (sperm protein associated with the nucleus mapped to the X chromosome). On two-dimensional Western blots of human sperm extracts, SPAN-X antibodies recognized 19 spots ranging from 20 to 23 kDa with isoelectric points from 5.0 to 5.5. Differential extraction of spermatozoa demonstrated that the SPAN-X protein is highly insoluble. Only 50% of ejaculated spermatozoa exhibited SPAN-X immunofluorescent staining. Dual localization of the sex chromosomes and the SPAN-X protein demonstrated that an equal number of X- and Y-bearing spermatozoa exhibited SPAN-X staining. In transfected mammalian CV1 cells, the SPAN-Xa and SPAN-Xb proteins were localized to the nucleus and cytoplasm, respectively, by indirect immunofluorescence. On immunoblots of CV1 cells, the SPAN-Xa protein migrated at 15–20 kDa, whereas the SPAN-Xb protein migrated at a higher molecular weight of 21–22 kDa. The SPAN-X protein was ultrastructurally associated with nuclear vacuoles and the redundant nuclear envelope. SPAN-X is the first protein specifically localized to these poorly characterized structures of the mammalian sperm nucleus and provides a unique biochemical marker for investigation of their function in spermatozoa as well as the role of SPAN-X/CTp11 in human tumors.


PLOS ONE | 2007

Evolutionary Diversification of SPANX-N Sperm Protein Gene Structure and Expression

Natalay Kouprina; Vladimir N. Noskov; Adam Pavlicek; N. Keith Collins; Pamela D. Schoppee Bortz; Chris Ottolenghi; Dmitri Loukinov; Paul Goldsmith; John I. Risinger; Jung-Hyun Kim; V. Anne Westbrook; Gregory Solomon; Hanna Sounders; John C. Herr; Jerzy Jurka; Victor Lobanenkov; David Schlessinger; Vladimir Larionov

The sperm protein associated with nucleus in the X chromosome (SPANX) genes cluster at Xq27 in two subfamilies, SPANX-A/D and SPANX-N. SPANX-A/D is specific for hominoids and is fairly well characterized. The SPANX-N gave rise to SPANX-A/D in the hominoid lineage ∼7 MYA. Given the proposed role of SPANX genes in spermatogenesis, we have extended studies to SPANX-N gene evolution, variation, regulation of expression, and intra-sperm localization. By immunofluorescence analysis, SPANX-N proteins are localized in post-meiotic spermatids exclusively, like SPANX-A/D. But in contrast to SPANX-A/D, SPANX-N are found in all ejaculated spermatozoa rather than only in a subpopulation, are localized in the acrosome rather than in the nuclear envelope, and are expressed at a low level in several nongametogenic adult tissues as well as many cancers. Presence of a binding site for CTCF and its testis-specific paralogue BORIS in the SPANX promoters suggests, by analogy to MAGE-A1 and NY-ESO-1, that their activation in spermatogenesis is mediated by the programmed replacement of CTCF by BORIS. Based on the relative density of CpG, the more extended expression of SPANX-N compared to SPANX-A/D in nongametogenic tissues is likely attributed to differences in promoter methylation. Our findings suggest that the recent duplication of SPANX genes in hominoids was accompanied by different localization of SPANX-N proteins in post-meiotic sperm and additional expression in several nongonadal tissues. This suggests a corresponding functional diversification of SPANX gene families in hominoids. SPANX proteins thus provide unique targets to investigate their roles in the function of spermatozoa, selected malignancies, and for SPANX-N, in other tissues as well.


Biology of Reproduction | 2002

Analysis of a human sperm CD52 glycoform in primates: identification of an animal model for immunocontraceptive vaccine development

Tod C. McCauley; Barbara E. Kurth; Elizabeth J. Norton; Kenneth L. Klotz; V. Anne Westbrook; A. Jaganandha Rao; John C. Herr; Alan B. Diekman

Abstract Sperm agglutination antigen-1 (SAGA-1) is a human male reproductive tract glycoform of CD52. Unique modification of CD52 N-linked oligosaccharide chains in the epididymis and vas deferens results in the appearance of a carbohydrate epitope that is localized over the entire surface of human spermatozoa. SAGA-1 was characterized by the sperm-inhibitory murine monoclonal antibody (mAb) S19, and it is the target antigen of a human mAb (H6-3C4) associated with antibody-mediated infertility. Collectively, sperm surface localization, antibody inhibition of sperm function, and potential reproductive-tissue specificity identify SAGA-1 as an attractive candidate contraceptive immunogen. To establish an animal model for the study of SAGA-1 in immunologic infertility and immunocontraceptive development, we investigated the appearance of the S19 carbohydrate epitope in nonhuman primates. The S19 mAb demonstrated little to no immunoreactivity by Western blot analysis with protein extracts of spermatozoa from the baboon, marmoset, bonnet, cynomolgus, and pigtailed macaques. Immunohistochemical analysis identified CD52 in the bonnet monkey epididymis; however, the N-linked carbohydrate moiety recognized by the S19 mAb, and unique to SAGA-1, was absent. In contrast, the S19 carbohydrate epitope was identified in chimpanzee sperm extracts by Western blot analysis and in chimpanzee epididymal tissue sections by immunohistochemical analysis, indicating that it is conserved in this close relative of the human. Chimpanzee testis, seminal vesicle, and prostate do not express the S19 epitope. Although anti-CD52 immunoreactivity was identified in the spleen, the carbohydrate moiety recognized by the S19 mAb was absent, corroborating data in the human that demonstrated tissue-specific glycosylation of sperm CD52. Immunofluorescent analysis indicated that the chimpanzee homologue of sperm CD52 was present over the entire spermatozoon. In addition, the S19 mAb agglutinated chimpanzee spermatozoa in a manner similar to the effect observed on human spermatozoa. These data indicate that the distinctive carbohydrate moiety of human sperm CD52 is present in the chimpanzee, and they identify the chimpanzee as the most appropriate primate model to study the potential of this unique CD52 glycoform as a contraceptive immunogen.


Archive | 2003

CAPACITATION: SIGNALING PATHWAYS INVOLVED IN SPERM ACQUISITION OF FERTILIZING CAPACITY

V. Anne Westbrook; Alan B. Diekman; John C. Herr; Pablo E. Visconti

Mammalian testicular spermatozoa are morphologically differentiated but are neither progressively motile nor able to fertilize an egg. Although the ability to move forward is acquired during maturation in the epididymis, sperm require a finite period of residence in the female reproductive tract before they become fertilization competent. The molecular, biochemical, and physiological changes that occur to sperm while in the female tract are collectively referred to as capacitation. Capacitation is associated with the attainment of a distinct motility pattern, namely hyperactivation. Another endpoint of sperm capacitation is the ability of spermatozoa to undergo the acrosome reaction in response to physiological stimuli such as the zona pellucida (1) and progesterone (2). During capacitation, changes in membrane properties, enzyme activities, and motility render spermatozoa responsive to stimuli that induce the acrosome reaction and prepare spermatozoa for penetration of the egg investments prior to fertilization. These changes are facilitated by the activation of cell signaling cascades in the female reproductive tract in vivo or in defined media in vitro. The purposes of this chapter are to consider some recent contributions towards our understanding of capacitation, to summarize open questions in this field, and to discuss future avenues of research.


Developmental Biology | 2002

CABYR, a novel calcium binding tyrosine-phosphorylation regulated fibrous sheath protein involved in capacitation

Soren Naaby-Hansen; Arabinda Mandal; Michael J. Wolkowicz; Buer Sen; V. Anne Westbrook; Jagathpala Shetty; Scott A. Coonrod; Kenneth L. Klotz; Young-Howan Kim; Leigh Ann Bush; Charles J. Flickinger; John C. Herr


Molecular Human Reproduction | 2004

Expression analysis of the human testis‐specific serine/threonine kinase (TSSK) homologues. A TSSK member is present in the equatorial segment of human sperm

Zhonglin Hao; Kula N. Jha; Young-Hwan Kim; Soumya Vemuganti; V. Anne Westbrook; Olga Chertihin; Karin Markgraf; Charles J. Flickinger; Michael A. Coppola; John C. Herr; Pablo E. Visconti

Collaboration


Dive into the V. Anne Westbrook's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alan B. Diekman

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pablo E. Visconti

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge