Centrosomes in fertilization, early embryonic development, stem cell division, and cancer

Centrosomes in fertilization, early embryonic development, stem cell division, and cancer

 

Sigrid Hoyer-Fender

Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology - Developmental Biology,
GZMB, Ernst-Caspari-Haus, Justus-von-Liebig-Weg 11,
Georg-August-University of Göttingen, Germany

shoyer@gwdg.de
phone: 0049-551-395434
fax: 0049-551-395416

 

October 2011

 

 

 

Abstract


The centrosome, located at the cell center, is the main microtubule organizing center (MTOC) of the cell. In interphase, it assembles microtubules that are then anchored with their minus ends at the centrosome. Since microtubules (MTs) build the cellular framework for the positioning of cell organelles, the centrosome is important for the whole cellular architecture. In mitotic cells, the centrosome plays a central role in organizing the mitotic spindle to correctly separate the chromosomes. Aberrant centrosomes are often associated with aneuploidy and tumorigenesis. In animal cells the centrosome consists of a pair of centrioles surrounded by the pericentriolar material (PCM). Intrinsic proteins of the centrosome are mostly characterized by their overall coiled-coil structure, but otherwise lack specific domains. Additionally, a lot of proteins show a cell cycle dependent association with the centrosome leading to the assumption, that the centrosome might orchestrate cell cycle progression. In non-cycling cells, the centrosome is transformed into the basal body to generate a primary cilium, a cellular organelle that has long been neglected. Nowadays, the importance of primary cilia for signal transduction and cell homeostasis is widely acknowledged as indicated by the constantly growing list of cilia dependent diseases. The centrosome thus is an essential organelle in most animal cells affecting cell shape, cell cycle progression, and the cellular response to environmental cues.

 

Introduction


The centrosome is a tiny organelle approximately ~1-2 µm in diameter and positioned centrally in animal cells. It consists of two orthogonally arranged centrioles embedded in an electron dense mesh called the pericentriolar matrix (PCM) (reviewed in: Kellogg et al., 1994; Stearns and Winey, 1997; Urbani and Stearns, 1999; Zimmerman et al., 1999; Bornens, 2002) (Fig. 1). Centrioles are microtubule based organelles mostly with a characteristic radial array of nine microtubule (MT) triplets. They are usually about 0,5 µm long and 0,2 µm in diameter. Both centrioles of a typical centrosome differ in age (see below) and structure and are associated by interconnecting fibers. The older centriole, also called the mature or mother centriole, is characterized by distal and subdistal appendages. Subdistal appendages function by anchoring microtubules, whereas distal appendages seem to be involved in anchoring the centriole/basal body to the cell membrane (reviewed in: Hoyer-Fender, 2010).

The PCM consists of a large number of proteins that share a characteristic coiled-coil structure. Although the pericentriolar material has long been described as an "amorphous cloud of electron dense material" centrosomal coiled-coil proteins may form a lattice which serves as a platform to which regulatory proteins may bind (reviewed in: Fry and Hames, 2004). An integral coiled-coil protein of the PCM is, e.g., pericentrin/kendrin that plays a role in centrosome and spindle organization and forms a complex with γ-tubulin (Dictenberg et al., 1998; Doxsey et al., 1994; Li et al., 2000; Flory et al., 2000; Flory and Davis, 2003). Some other coiled-coil proteins are e.g. ninein, Cep135, centriolin, CG-NAP/AKAP350/AKAP450, and ODF2/cenexin (Bouckson-Castaing et al., 1996; Lange and Gull, 1995; Takahashi et al., 1999; Takahashi et al., 2002; Schmidt et al., 1999; Ohta et al., 2002; Gromley et al., 2003; Donkor et al., 2004). Ninein may function in microtubule minus-end capping and anchoring and may promote microtubule nucleation by docking the γ-tubulin ring complex (γ-TuRC) at the centrosome (Mogensen et al., 2000; Delgehyr et al., 2005). Cep135 is a universal component of the centrosome in a wide range of organisms and important for MT organization (Ohta et al., 2002). Organization of centrosomal microtubules in addition seems to be dependent on transforming acidic coiled-coil (TACC) proteins, a family of proteins that concentrate to centrosomes through a conserved coiled coil carboxy-terminal domain called the TACC domain (Gergely et al., 2000a; Gergely et al., 2000b; Lee et al., 2001). Centriolin localizes to the maternal centriole and functions in both cytokinesis and cell cycle progression (Gromley et al., 2003). ODF2/cenexin is a marker of the mature centriole and essential for primary cilia formation (Nakagawa et al., 2001; Ishikawa et al., 2005).

In the interphase cell, the centrosome functions as the main microtubule organizing center (MTOC) to nucleate and anchor microtubules (MTs). Microtubules are assembled from soluble tubulin subunits and are important for the maintenance of cell morphology and the position of organelles. Whereas the centrioles may rather act as a scaffold on which to assemble the pericentriolar material, the MT nucleation and anchorage capacity of centrosomes relies on components of the PCM. MT are anchored at their minus ends by the γ-tubulin ring complex (γ-TuRC) located in the PCM (Moritz et al., 1995a; Moritz et al., 1995b; Wiese and Zheng, 2000). The γ-tubulin ring complex (γ-TuRC) consists of γ-tubulin and a couple of associated proteins (Moritz et al., 1998; Murphy et al., 1998; Tassin et al., 1998; Oegema et al., 1999; Fava et al., 1999; Murphy et al., 2001). γ-tubulin is essential for MT nucleation as has been shown by depletion experiments (Raynaud-Messina et al., 2004). γ-tubulin binds to the minus ends of MTs and by capping them prevents minus end growth (Li and Joshi, 1995; Leguy et al., 2000). Although in animal cells most MTs nucleate from centrosomes, the bulk of soluble γ-tubulin is found in the cytoplasm (about 80% in contrast to only 20% found at the centrosome) but is devoid of significant MT nucleation activity (Moudjou et al., 1996). At the onset of mitosis, concomitant with an increase in MT nucleation activity, additional γ-tubulin is recruited to the centrosome as well as numerous regulatory proteins (reviewed in: Schiebel, 2000). This structural reorganization of centrosomes in the preparation for mitosis is termed maturation and protein kinases as well as phosphatases have been implicated in its regulation (reviewed in: Meraldi and Nigg, 2002).

In addition to intrinsic centrosomal components the centrosome is a platform for the binding of a lot of different proteins and enzymes, i.e. protein kinases and phosphatases, which are brought into contact with their substrates and upstream regulators. Therefore, the centrosome plays important roles in orchestrating cell cycle progression (reviewed in: Fry and Hames, 2004).

 

 

Fig. 1 Schematic representation of a typical mammalian centrosome consisting of mother and daughter centrioles enclosed by the pericentriolar matrix (PCM). The mother centriole contains distal and subdistal appendages (not all are shown) and both centrioles are associated by interconnecting fibers. In the PCM, γ-TuRCs are located that anchor MT minus ends.

 

In mitosis, the two poles of the bipolar mitotic spindle are established by two centrosomes that are generated during the previous cell cycle. Since centrosomes establish the spindle poles, the presence of more than two centrosomes in a single cell may influence the formation of the bipolar spindle eventually resulting in multipolar spindles and unequal distribution of chromosomes. Centrosome duplication therefore has to be restricted to once and only once per cell cycle. The centrosome division cycle starts at G1-S phase and by the end of G2, each cell has two centrosomes, each one comprising two centrioles surrounded by its own PCM (reviewed in: Kochanski and Borisy, 1990; Delattre and Gönczy, 2004; Tsou and Stearns, 2006a; Nigg, 2007; Azimzadeh and Bornens, 2007; Bettencourt-Dias and Glover, 2009). Mitosis then separates the two centrosomes into the two daughter cells. Each daughter cell therefore receives one centrosome. Centrosome duplication starts with the separation of the two otherwise tightly opposed ("engaged") centrioles. Although both centrioles are still attached via fibers, disengagement might license subsequent duplication in the next cell cycle (Tsou and Stearns, 2006b). Both centrioles then nucleate a new daughter centriole in a perpendicular orientation at their proximal ends. Centriole duplication starts at the same time as initiation of DNA synthesis. Throughout S to G2 phases, the procentrioles elongate to full-length daughter centrioles. Eventually, each newly formed centriole pair assembles its own PCM. By the end of G2, two centrosomes are present in the cell that eventually separate to build up the spindle poles during mitosis. Mitosis then distributes each of the two centrosomes to one daughter cell. Therefore, one daughter cell receives the original mother centriole associated with a daughter centriole generated during the last cell cycle, whereas the other daughter cell received the original daughter centriole associated with a new centriole generated during the last cell cycle. The original daughter centriole then matures into a mother centriole by formation of appendages. Eventually, each new daughter cell contains one centrosome comprising one mother and one daughter centriole (Fig. 2).

Fig. 2 Canonical centrosome duplication cycle of somatic cells. Centriole disengagement in G1 phase licenses centrioles for duplication. Centriole duplication starts in S phase, at the same time as DNA replication, by the formation of procentrioles. During G2 phase procentrioles elongate to full-length daughter centrioles and each centriole pair assembles its own PCM. Centrosomes are then separated to build up the poles of the bipolar mitotic spindle in mitosis.

 

Centriole duplication begins at the G1/S transition with the appearance of one procentriole at the proximal end of each parental centriole. Procentrioles organise around a cartwheel, and microtubules are nucleated by γ-TuRC like structures. Centriole duplication therefore depends on the presence of γ-tubulin, and of NEDD1, which recruits γ-TuRCs to the centrosome (Haren et al., 2006; Guichard et al., 2010). During S to G2 phases of the cell cycle, γ-tubulin is targeted for degradation by BRCA1 mediated ubiquitinylation thus blocking centrosome reduplication (Kasi and Parvin, 2008; reviewed in: Parvin, 2009). The initial steps of procentriole assembly are additionally dependent on several other proteins. An intrinsic centriolar protein is centrin-2. Centrins are small Ca2+-binding proteins of the calmodulin superfamily. They are highly conserved in evolution and are required for centrosome duplication across the eukaryotic kingdom. In HeLa cells, RNAi-mediated inactivation of centrin-2 prevents daughter centriole formation (Salisbury et al., 2002). Sas-6, Cep135, Sas-4 (CPAP in humans), Cep152, CP110, and geminin are likewise essential for centriole duplication as well as an array of kinases, such as polo-like kinases Plk1, and Plk2, cyclin-dependent kinase 2 (Cdk2)/cyclin A/E, and Mps1, and the phosphatase Cdc14B (Lacey et al., 1999; Meraldi et al., 1999; Chen et al., 2002; Fisk et al., 2003; Warnke et al., 2004; Leidel et al., 2005; Tachibana et al., 2005; Dammermann et al., 2008; Wu et al., 2008; Tsou et al., 2009; Lu et al., 2009; reviewed in: Sillibourne and Bornens, 2010; Pike and Fisk, 2011). Plk2 is activated near the G1/S phase transition and regulates centriole duplication by phosphorylation of NPM/B23 (Nucleophosmin) and CPAP (Krause and Hoffmann, 2010; Chang et al., 2010). The master regulator of centriole duplication, however, is the protein kinase Plk4/SAK (Habedanck et al., 2005). Plk4 belongs to the polo-like kinase family and localizes to centrosomes. Depletion of Plk4 impairs centriole duplication, whereas overexpression of Plk4 leads to the formation of multiple daughter centrioles in a single cell. Additionally, the correct level of Plk4 is essential for cytokinesis. By regulating the localization of Sas-6, a conserved coiled-coil protein essential for centriole duplication, Plk4 may affect centriole biogenesis (reviewed in: Pearson and Winey, 2010). On the other hand, Plk4 itself is recruited to the centrosome by Cep152 along with CPAP (Cizmecioglu et al., 2010). In vitro, Cep152 can be phosphorylated by Plk4 (Hatch et al., 2010). Plk4 negatively regulates the F-box protein FBXW5 by phosphorylation to suppress ubiquitylation of Sas-6. Depletion of FBXW5 leads to centrosome overduplication (Puklowski et al., 2011). These results show, that phosphorylation and controlled protein degradation are important regulators of centriole duplication.

Even though centriole duplication is typically coupled with DNA replication, there are some important exceptions: Uncoupling of centrosome duplication from DNA replication seems to occur during meiotic prophase in male gametogenesis and after fertilization (see below). Primary spermatocytes as well as secondary spermatocytes have two centrioles in each centrosome. The same holds true for spermatids, the products of meiotic divisions. Each secondary spermatocyte (with one centriole pair) generates two spermatids (also with one centriole pair per cell) during meiosis II. Centrosome duplication therefore has to occur prior to meiosis I, most likely concurrently with the last DNA replication step, and additionally prior to meiosis II, despite the absence of DNA replication, to ensure correct centrosome segregation. At fertilization, the sperm of most mammals contributes one centriole to the acentriolar oocyte. The single paternally contributed centriole therefore must duplicate once, in spite of absence of DNA replication, prior to first mitosis in zygotes to give rise to all in all four centrioles in two centrosomes.

The centrosome is a dynamic structure that changes in size and protein composition synchronous with the cell cycle. As centrosomes are not encircled by a membrane, the definition of true centrosomal components is hampered. Centrosomal proteins could therefore roughly be classified into three groups: 1. centriolar proteins, that are intrinsic to centrioles, as e.g. α- and β-tubulin, or permanently associated with centrioles; 2. intrinsic proteins of the PCM, as e.g. γ-tubulin and; anpericentrind 3. proteins temporarily associated with the centrosome, as e.g. kinases. About 500 different proteins have been associated with the centrosome of somatic cells by proteomic studies (Andersen et al., 2003). However, not all of them may be true centrosomal proteins but co-isolated molecules that use the centrosome as a docking platform. Since most studies have been performed in somatic cells in culture, it is largely unknown whether centrosomes of somatic cells differ from those of stem cells, neither concerning their overall organisation nor their protein composition. These constrains are mainly attributed to the difficulties to identify and handle stem cells. Since the most important stem cell in development is constituted by the fusion of male and female gametes, the centrosome of gametes, zygotes, and early developmental stages have attracted major attention. This review focuses on the centrosome of germ cells, zygotes, and stem cells in mammals, and addresses centrosome aberrations in cancer.

 

1. Centrosomes in Germ cells


Duplication and segregation of centrosomes are based on cell-cycle dependent conservative replication of centrioles, and semi-conservative segregation during cell division to ensure equal centrosome distribution to daughter cells. Each mitotically cycling cell thus comprises one and only one centrosome. To ensure proper centrosome number in each cell of the body, even during successive generations, centrosomes have to be reduced during gametogenesis, in analogy to the reduction of DNA content. Thus, fusion of male and female gametes not only restores the diploid DNA content but also the centrosome.
It was Theodor Boveri in 1901 who introduced the theory of uniparental distribution of the centrosome by recognizing that the egg typically loses the centrosome during oogenesis whereas the sperm typically introduces this structure at fertilization (Boveri 1901). Centrosome reduction thus occurs differentially in males and females.
However, thenceforward, the picture has become much more complicated owing to the emergence of even more sophisticated methods and detection skills (comprehensively reviewed by Schatten, 1994; Manandhar et al., 2005; Schatten and Sun, 2010). Although most animals show the typical pattern of paternal contribution of the centrosome, there are remarkable exceptions. Investigation of microtubule patterns throughout fertilization in the mouse, and, later on, studies using centrosomal antibodies showed that the sperm do not contain a centrosome (Schatten et al., 1985; Schatten et al., 1986). In mice and other rodents, the centrosome, which is largely defined by its microtubule polymerizing activity, thus seems to be maternally inherited (reviewed in: Schatten et al., 1991).

The centrosome of mature spermatozoa
In early stages of post-meiotic male germ cells, the centrosome is transformed into a basal body to generate the sperm flagellum. Usually, the distal centriole which is oriented perpendicular to the membrane, functions as the basal body to give rise to the axoneme. The proximal centriole is orthogonally oriented but closely associated with the distal centriole. The proximal centriole organizes the anlage of the capitulum which will later articulate with the implantation fossa of the sperm nucleus. The distal end of the proximal centriole additionally polymerizes the centriolar adjunct, a temporary structure no longer found in mature spermatozoa. Both distal and proximal centrioles are engaged in assembly of the striated columns of the connecting piece (reviewed in: Fawcett, 1981). Later during spermiogenesis, the distal centriole, which formerly has initiated outgrowth of the sperm flagellum, disintegrates and is no longer present in the mature spermatozoon whereas the proximal centriole usually persists (Fig. 3).

 

 

Fig. 3 Centrioles in elongating spermatids and mature spermatozoa of most mammals including human. In elongating spermatids both proximal and distal centrioles are present. The distal centriole functions in assembling the sperm tail. In mature spermatozoa the distal centriole is degenerated and no longer discernible.

 

The disintegration of only the distal centriole in mature spermatozoa is typical for most mammals including humans and rhesus monkeys (Manandhar et al., 2000). Therefore, in these species the centriole of the prospective zygotic centrosome is paternally inherited at fertilization. Paternal inheritance of the centriolar component has been confirmed for human (Sathananthan et al., 1991; Simerly et al., 1995), sheep (Le Guen and Crozet, 1989; Crozet, 1990), pig (Szöllösi and Hunter, 1973) and cow (Sathananthan et al., 1997; Long et al., 1993; Navara et al., 1994). Albeit centrioles are present in mature spermatozoa, the composition and accessibility of centrosomal proteins seem to be altered. Centrin and γ-tubulin are both found in human as well as in bovine mature spermatozoa. However, γ-tubulin is largely inaccessible in the mature spermatozoa in vitro until after disulfide bond reduction. It is therefore assumed, that the sperm centrosome must be primed by the reducing environment within the mammalian oocyte's cytoplasm to be transformed into an active centrosome. Since γ-tubulin is not only present in the oocyte, but also introduced by the sperm at fertilization, it is biparentally inherited (Simerly et al., 1999). Remarkably, anti-centrin staining of human and bull mature spermatozoa revealed two adjacent immunoreactive spots at the base of the sperm head which is reminiscent of two adjacent centrioles despite the degeneration of the distal centriole. However, immuno-EM studies revealed decoration of only the remaining proximal centriole (Simerly et al., 1999). These results suggest that the absence of a distinct centriolar structure does not mean that all centrosomal components are missing as well. The mature sperm nevertheless might retain some centrosomal components that are transmitted to the oocyte at fertilization to enable reconstitution of the zygotic centrosome.

In contrast to most mammals (including human), both distal and proximal centrioles degenerate during spermiogenesis in mice and other rodents. Additionally, mouse sperm are devoid of key centrosomal components and do not have microtubule nucleation capacity anymore. The MT nucleation capacity can be monitored by the formation of a monastral aster adjacent to the incorporated sperm nucleus in fertilized oocytes (Schatten et al., 1986). Loss of key PCM proteins and degeneration of centrioles are consecutive events. Round spermatids as well as elongating spermatids display distal and proximal centrioles along with γ-tubulin and centrin containing foci. In elongating spermatids γ-tubulin is mainly found around the centriolar adjunct which nucleates microtubules. At the time of spermiation, γ-tubulin is lost from the neck region and shed along with the residual body. The distal centriole disintegrates during testicular stage, and the proximal centriole degenerates in the epididymis. Mature mouse sperm neither contain centrioles nor γ-tubulin or centrin (Manandhar et al., 1998; Manandhar et al., 1999). Partial versus complete degeneration of centrioles and associated centrosomal proteins in human and mouse spermatozoa, respectively, is also supported by the species-specific distribution of centrosomal proteins (e.g. TSKS, substrate of testis-specific serine kinases 1 and 2; Xu et al., 2008).

The proper function of the centrosome is vitally important for the unification of male and female pronuclei after fertilization and ongoing development. In most animals, the centrosome of the spermatozoon organizes the sperm aster, which brings the parental genomes together during fertilization. Improper centrosomal inheritance or dysfunction of the sperm centriole might be associated with cleavage irregularities of the fertilized oocyte and abnormal embryonic development thus linking centrosomes to infertility (Simerly et al., 1995; Palermo et al., 1997; Nagy, 2000; Chatzimeletiou et al., 2008; reviewed in: Schatten and Sun, 2009a). It has been shown in bull that naturally occurring variations in their centrosomes correlate with reproductive success (Navara et al., 1996). Likewise, centrosome dysfunction might be responsible for low efficiency rate of cloning mammals by somatic cell nuclear transfer (SCNT), and might even affect the success of intracytoplasmic sperm injection (ICSI) in assisted reproduction techniques (Chemes et al., 1999; Zhong et al., 2005; Dai et al., 2006; Zhong et al., 2007; Terada, 2007).

The centrosome during oogenesis and in oocytes
Oogonia and oocytes up to the pachytene stage of hamsters, mice, rats, gerbils, and man contain centrioles, but in subsequent meiotic stages, centrioles are absent. At the time of germinal vesicle breakdown microtubule polymerization focuses on several electron-dense aggregates inside the ooplasm (Szöllösi et al., 1972; Sathananthan et al., 2006). Mature oocytes of rabbits, cows, and sheep are likewise devoid of centrioles (Sathananthan et al., 1997; Crozet et al., 2000). However, as has been revealed by immunolocalization studies mostly during mouse oogenesis, PCM proteins, including γ-tubulin, pericentrin, and NuMA are not eliminated during meiotic stages but show a dynamic redistribution. NuMA is a nuclear matrix protein which plays an important role in establishment and maintenance of the bipolar spindle apparatus (Maro et al., 1985; Gueth-Halonet et al., 1993; Calarco, 2000; Carabatsos et al., 2000; Lee et al., 2000; Can et al., 2003; Meng et al., 2004; Tang et al., 2004; Sedo et al., 2011). 4-D imaging in live maturing mouse oocytes revealed, that numerous acentrosomal MTOCs, spread in the ooplasm, then assemble into the functional acentrosomal meiotic spindle (Schuh and Ellenberg, 2007). Thus, centrosomal material is stored in the oocyte albeit in a somehow unfocussed manner, that is then attracted by the sperm after fertilization to generate a functional centrosome. Studies of aster formation in the cow suggested that the centrosome generated after fertilization is a blending of paternal and maternal components (Navara et al., 1994).

 

2. The centrosome during early development


In humans, the sperm contributes the proximal centriole and the mid-piece of the sperm tail to the acentriolar oocyte at fertilization. Proximal centriole and sperm mid-piece are closely associated with the decondensing sperm nucleus that gradually develops into a male pronucleus. Shortly after sperm incorporation into the oocyte, a sperm aster is formed from the proximal centriole. The aster is build up of microtubules required for the apposition of the male and female pronuclei - an essential prerequisite for merging of paternal and maternal genomes and successful fertilization. In prometaphase, when male and female pronuclei breakdown, the centrosome is separated to form the two poles of the mitotic spindle. Transmission electron microscopy revealed single or double centrioles within centrosomes at one pole of the first cleavage spindle (Sathananthan et al., 1991). Most likely due to the difficulties to gather both centrioles of one centrosome or even both centrosomes at the same section plane in the voluminous ooplasm, double centrioles are not found at each pole of the first cleavage spindle. But afterwards, centrioles are then found at every stage of early embryo development (Sathananthan et al., 1996; Sathananthan, 1997).
In bovine, centrioles are likewise paternally inherited, and organize the sperm aster after fertilization (Sathananthan et al., 1997). Zygotes from humans, sheep and cows thus display centrioles at the spindle poles at first mitosis, although extensive electron microscopic studies have failed to find duplex centrioles in all spindle poles during the first cleavage of human zygotes (Sathananthan et al., 1991; Sathananthan et al., 1996; Santhananthan et al., 1997; Le Guen and Crozet, 1989). In sheep zygotes, the first cleavage spindle possesses two centrioles at one pole and only one centriole at the other pole, probably owing to difficulties to gather all centrioles at the same section plane (Crozet et al., 2000). However, in late-stage embryonic cells centriolar duplexes are invariably found in all cells (Sathananthan, 1997).
The paternal centrosomal constituents of human and bovine mature sperm are subject to phosphorylation in vitro after exposure to X. laevis cell-free egg extracts as well as in vivo after oocyte penetration as visualized by the phosphoprotein specific antibody MPM-2 (Simerly et al., 1999). This suggested that paternal centrosomal proteins might be intensively modified post-translationally after exposure to ooplasm at fertilization. After fertilization, the sperm centrosome recruits γ-tubulin from the ooplasm resulting in an accumulation of mostly maternal γ-tubulin at the zygotic centrosome (Simerly et al., 1999; Shin and Kim, 2003).

Mature oocytes are devoid of centrioles but contain PCM proteins. Moreover, in mice and other rodents, the sperm does not contribute a centriole to the oocyte at fertilization. The microtubules are therefore organized after fertilization by components stored in the oocyte but not by the incorporated sperm nucleus. During early mouse development important changes in composition and structure of MTOCs take place, reflected by the varying number of PCM foci as well as by their protein composition (Gueth-Hallonet et al., 1993). Though, first and second cleavage stage mouse embryos are devoid of centrioles, centrioles arise during the blastocyst stage by de novo synthesis (Szöllösi et al., 1972). De novo formation of centrioles has also been observed in rabbit oocytes parthenogenetically activated (Szöllösi and Ozil, 1991). Centriole precursors are similar to deuterosomes, intermediate structures of de novo formation of basal bodies in multi-ciliated cells, or procentrioles. Procentriole-like bodies may first form solitary centrioles that replicate in an orthogonal manner afterwards like in parthenogenetically activated sea urchin eggs (Kallenbach and Mazia, 1982). Centrioles with the usual morphology could first be found in the 16-cell stage by electron microscopy (Szöllösi et al., 1972; Gueth-Hallonet et al., 1993). These observations suggest once more that centrioles are not essential for formation of bipolar spindles in animals, and that centrioles are generated de novo in the early mouse embryo.

 

Summary: From germ cells to zygotes
In mammalian species (including human), a gradual degeneration of the centrosome during spermiogenesis is observed. Loss of key PCM proteins follows loss of microtubule nucleation capacity and eventually results in partial or complete degeneration of centrioles. In most mammals (including human), the sperm retains the proximal centriole but degenerates the distal one. In contrast, oogonia originally contain a pair of well-defined centrioles that are lost during oogenesis, resulting in the mature oocyte being devoid of centrioles. However, PCM is retained albeit dispersed throughout the egg cytoplasm. As in most mammals the human oocyte has no granular centrosomal material at meiotic spindle poles which is a remarkable difference to mouse oocytes that have a dominant maternal centrosome. Upon fertilization, a functional centrosome is restored in the zygote. In mouse and other rodents, in which the sperm does not contribute a centriole, the zygotic centrosome is constituted from maternal material, whereas in most other mammals (including human) the zygotic centrosome is restored by the proximal centriole submitted by the sperm and maternal PCM components that are attracted by the sperm (Schatten and Sun, 2009b). Restoration of the centrosome after fertilization thus occurs in most mammals (with the exception of rodents) by the sperm supplying the centriole and the PCM supplied by the egg cytoplasm.

 

3. The centrosome in stem cells


The formation of a bipolar mitotic spindle is a crucial event for proper cell division ensuring equal distribution of chromosomes as well as equal amounts of cytoplasmic components to daughter cells. Concurrently, centrosomes likewise may be equally distributed to daughter cells. However, this might not always be the case. Unequal centrosome distribution to daughter cells may set up the pattern for differentiation. A key feature in stem cell biology is asymmetric cell division. Asymmetric cell division results in two unequal daughter cells that receive different amounts of cytoplasmic material and organelles and possibly also centrosomal components. One of the daughter cells maintains stem cell characteristics by remaining at the stem cell niche and receiving signals from the hub (reviewed in: Morrison and Spradling, 2008). The sibling cell is no longer in contact with the hub and undergoes differentiation. This kind of cell division which results in two sibling cells undergoing different cell fates requires a specific arrangement of the mitotic spindle that has to be perpendicular to the hub. Disturbances of spindle position therefore affect the fate of the daughter cells. If both sibling cells will remain in contact with the hub, two stem cells instead of one stem cell and one differentiating cell will be the result. Disturbances in spindle position therefore may derange the balance between stem cells and differentiated cells and result in uncontrolled proliferation and cancer (reviewed in: Pease and Tirnauer, 2011). However, mammals are not a good model system to study the mode of cell division in stem cell maintenance and the contributions the centrosome provides to cell differentiation and development.

Our knowledge on asymmetric stem cell division and stem cell centrosomes is mainly based on studies in the fruit fly Drosophila, and the nematode Caenorhabditis elegans (reviewed in: Yamashita and Fuller, 2008; Gönczy, 2008; Neumüller and Knoblich, 2009). In Drosophila males, germline stem cells (GSC) preferentially inherit and maintain the mature centrosome (comprising the mother centriole) as long as they remain in the niche. The daughter centrosome is inherited by the daughter cell fated to differentiate. Asymmetric distribution of centrosomes is determined by the differential microtubule nucleation capacity of mother and daughter centrosomes affecting spindle orientation (Yamashita et al., 2003; Yamashita et al., 2007). Misorientation of centrosomes during ageing affects stem cell division (Cheng et al., 2008). Unequal centrosome distribution has also been reported in Drosophila neurogenesis. Larval neural stem cells called neuroblasts divide asymmetrically to produce a larger self-renewing neuroblast and a smaller larval basal ganglion mother cell that pursues neural differentiation. One neuroblast centrosome remains associated with the neuroblast cortex while the other centriole pair moves away (Rebello et al., 2007; Rusan and Pfeifer, 2007). However, in this case the mother centrosome is inherited by the differentiating daughter cell (Januschke et al., 2011). In the female Drosophila germline centrosomes segregate randomly (Stevens et al., 2007). Thus, asymmetric centrosome inheritance is not a general feature of stem cell lineages.

In mammals, the best studied example of asymmetric cell division is provided by the developing mouse forebrain. Early in development, progenitor cells in the ventricular zone divide symmetrically to increase the progenitor cell pool. From day 10 of embryonic development, progenitor cells, now called radial glia progenitors, divide asymmetrically allowing more differentiated cells to be produced (Götz and Huttner, 2005). Asymmetric division of radial glia progenitors produce a self-renewing radial glia and a differentiating cell that leaves the ventricular zone to become a neuron. Radial glia progenitors preferentially inherit the older centrosome, whereas the cell that leaves the ventricular zone to become a neuron preferentially inherits the daughter centrosome. By removal of ninein that is associated with the mother centriole, centrosome asymmetry may be impaired leading to disruption of asymmetric centrosome inheritance and depletion of progenitor cells from the ventricular zone (Wang et al., 2009). Asymmetric centrosome inheritance thus maintains neural progenitor cells in the neocortex of the mouse. The importance of the mature centrosome to maintain the progenitor pool in the ventricular zone is further substantiated by silencing of genes encoding centrosomal proteins. Likewise, the human disease microcephaly, characterised by a severely decreased brain size, is caused by loss of function of centrosome-related proteins (reviewed in: Lesage et al., 2010).

Symmetric cell division, as apparently seen in cells in culture, does by no means demonstrate equal distribution of cellular components to daughter cells (Fuentealba et al., 2008). As shown by Fuentealba et al. in mammalian cultured cell lines, including human ES cells, and in vivo in Drosophila embryos, both centrosomes of a mitotic cell can differ in composition. Daughter cells therefore receive a differential protein pool.

 

4. The centrosome in cancer


Although centrosome duplication is tightly linked to the progression of the cell cycle under normal conditions, surprisingly faulty centrosome duplication did not cause cell cycle arrest (Uetake and Sluder, 2004; Wong and Stearns, 2005). In almost all types of solid tumors as well as in hematological malignancies like lymphoma and leukemia abnormal centrosome amplification is common. Centrosome amplification together with aneuploidy are hallmarks of many cancers (reviewed in: Salisbury et al., 1999; Nigg, 2002; Wang et al., 2004; Sagona and Stenmark, 2010). Although typical centrosomes with two centrioles have been identified by electron microscopy in undifferentiated human ES cells, a very high percentage of cultured human embryonic stem cells display numerical centrosome abnormalities that may be responsible for the observed chromosome instabilities in these cells (Sathananthan et al., 2002; Holubcova et al., 2011). The question whether centrosome amplification causes aneuploidy, or vice versa, is still a matter of debate. It has been demonstrated that cells with supernumerary centrosomes and tetraploid genomes can continue to cycle eventually resulting in aneuploid cells. Moreover, tetraploid cells are more likely to be tumorigenic than their diploid counterparts (reviewed in: Sankaran and Parvin, 2006). A first direct causative link between centrosome amplification and tumorigenesis has been provided by studies in the fruit fly Drosophila melanogaster. Larval brain from animals possessing supernumerary centrosomes due to Plk4 overexpression was able to initiate tumorigenesis in wild type host flies when transplanted (Basto et al., 2008; Castellanos et al., 2008). Surprisingly, in mouse fibroblasts Plk4 heterozygosity impairs cytokinesis leading to tetraploid daughter cells with amplified centrosomes. Moreover, aberrant Plk4 activity seems to be involved in tumor formation in mice and in humans (Macmillan et al., 2001; Ko et al., 2005; Rosario et al., 2010). Tight control of Plk4 level and activity is therefore mandatory. Plk4 stability is regulated by autophosphorylation and subsequent ubiquitin-dependent degradation (Holland et al., 2010).

Besides polo-like kinase Plk4, various other proteins are dysregulated in a lot of tumors which concurrently exhibit centrosome aberrations. In breast and colorectal tumors the Aurora-A gene is frequently amplified. Aurora-A belongs to the family of aurora protein kinases which consists of three members in mammals. Aurora-A is a mitotic kinase which localizes to centrosomes. Apart from gene amplification, Aurora-A overexpression is found in many tumors. In cultured cells, overexpression of Aurora-A leads to centrosome amplification (Sen et al., 1997; Bischoff et al., 1998; Zhou et al., 1998; Meraldi et al., 2002; reviewed in: Giet et al., 2005). Aurora-A phosphorylates the E3-ubiquitin ligase BRCA1 to diminish its activity. Decreased ubiquitin-ligase activity inhibits γ-tubulin degradation leading to centrosome reduplication. Breast and ovary tumors are frequently associated with loss of the E3-ubiquitin ligase BRCA1 (Sankaran and Parvin, 2006). An important tumor suppressor is p53, and point mutations affecting p53 are common in many forms of human cancer (Hainaut et al., 1997). That p53 affects centrosome duplication has been shown in embryonic fibroblasts generated from p53-/- -mice. P53 deficient cells display multiple copies of functional centrosomes (Fukusawa et al., 1996), most likely due to tetraploidization caused by Aurora-A overexpression (Meraldi et al., 2002).

Centrosome amplification and chromosome instability are common hallmarks of cancer. Up to now, most results favour the view that overduplication of centrosomes is the primary cause of aneuploidy. Apart from centrosome amplification, structural and functional abnormalities of the centrosome have been observed in tumor cells as well. Moreover, centrosome dysfunction has been implicated in a wide variety of human diseases (reviewed in: Badano et al., 2005).

 

5. Conclusion


The centrosomes of stem cells are a relatively unexplored field. There are some hints, that centrosomes may be important for stem cell maintenance, and that mature and daughter centrosome differ from each other. But it is not known whether centrosomes from stem cells differ in composition from somatic centrosomes. Likewise, their functional contributions to stem cell maintenance or cell differentiation remain largely in the dark. Numerical, structural, and functional aberrations of the centrosome are associated with a wide variety of human diseases including cancer. Centrosome amplification in tumor cells may pave the way for chromosome instabilities leading to cancer progression.

Bibliography

Zellenstudien: Uber die Natur der Centrosomen. IV.
Boveri T.
Fischer, Jena, Germany. 1901.
 
Absence of centrioles in the first and second meiotic spindles of mouse oocytes.
Szollosi D, Calarco P, Donahue RP.
J Cell Sci. 1972 Sep;11(2):521-41.
PMID 5076360
 
The Cell.
Fawcett DW .
2nd edition. W. B. Saunders Company. 1981.
 
Origin and maturation of centrioles in association with the nuclear envelope in hypertonic-stressed sea urchin eggs.
Kallenbach RJ, Mazia D.
Eur J Cell Biol. 1982 Aug;28(1):68-76.
PMID 7128618
 
Non-spindle microtubule organizing centers in metaphase II-arrested mouse oocytes.
Maro B, Howlett SK, Webb M.
J Cell Biol. 1985 Nov;101(5 Pt 1):1665-72.
PMID 2865266
 
Microtubule configurations during fertilization, mitosis, and early development in the mouse and the requirement for egg microtubule-mediated motility during mammalian fertilization.
Schatten G, Simerly C, Schatten H.
Proc Natl Acad Sci U S A. 1985 Jun;82(12):4152-6.
PMID 3889922
 
Behavior of centrosomes during fertilization and cell division in mouse oocytes and in sea urchin eggs.
Schatten H, Schatten G, Mazia D, Balczon R, Simerly C.
Proc Natl Acad Sci U S A. 1986 Jan;83(1):105-9.
PMID 2417231
 
Microtubule and centrosome distribution during sheep fertilization.
Le Guen P, Crozet N.
Eur J Cell Biol. 1989 Apr;48(2):239-49.
PMID 2568260
 
Behavior of the sperm centriole during sheep oocyte fertilization.
Crozet N.
Eur J Cell Biol. 1990 Dec;53(2):326-32.
PMID 2081547
 
Mode of centriole duplication and distribution.
Kochanski RS, Borisy GG.
J Cell Biol. 1990 May;110(5):1599-605.
PMID 2335566
 
Centrioles in the beginning of human development.
Sathananthan AH, Kola I, Osborne J, Trounson A, Ng SC, Bongso A, Ratnam SS.
Proc Natl Acad Sci U S A. 1991 Jun 1;88(11):4806-10.
PMID 2052559
 
Maternal inheritance of centrosomes in mammals? Studies on parthenogenesis and polyspermy in mice.
Schatten G, Simerly C, Schatten H.
Proc Natl Acad Sci U S A. 1991 Aug 1;88(15):6785-9.
PMID 1862101
 
De novo formation of centrioles in parthenogenetically activated, diploidized rabbit embryos.
Szollosi D, Ozil JP.
Biol Cell. 1991;72(1-2):61-6.
PMID 1756312
 
gamma-Tubulin is present in acentriolar MTOCs during early mouse development.
Gueth-Hallonet C, Antony C, Aghion J, Santa-Maria A, Lajoie-Mazenc I, Wright M, Maro B.
J Cell Sci. 1993 May;105 ( Pt 1):157-66.
PMID 8360270
 
Chromatin and microtubule morphology during the first cell cycle in bovine zygotes.
Long CR, Pinto-Correia C, Duby RT, Ponce de Leon FA, Boland MP, Roche JF, Robl JM.
Mol Reprod Dev. 1993 Sep;36(1):23-32.
PMID 8398127
 
Pericentrin, a highly conserved centrosome protein involved in microtubule organization.
Doxsey SJ, Stein P, Evans L, Calarco PD, Kirschner M.
Cell. 1994 Feb 25;76(4):639-50.
PMID 8124707
 
The centrosome and cellular organization.
Kellogg DR, Moritz M, Alberts BM.
Annu Rev Biochem. 1994;63:639-74. (REVIEW)
PMID 7979251
 
Microtubule organization in the cow during fertilization, polyspermy, parthenogenesis, and nuclear transfer: the role of the sperm aster.
Navara CS, First NL, Schatten G.
Dev Biol. 1994 Mar;162(1):29-40.
PMID 8125194
 
The centrosome and its mode of inheritance: the reduction of the centrosome during gametogenesis and its restoration during fertilization.
Schatten G.
Dev Biol. 1994 Oct;165(2):299-335. (REVIEW)
PMID 7958403
 
A molecular marker for centriole maturation in the mammalian cell cycle.
Lange BM, Gull K.
J Cell Biol. 1995 Aug;130(4):919-27.
PMID 7642707
 
gamma-tubulin is a minus end-specific microtubule binding protein.
Li Q, Joshi HC.
J Cell Biol. 1995 Oct;131(1):207-14.
PMID 7559777
 
Three-dimensional structural characterization of centrosomes from early Drosophila embryos.
Moritz M, Braunfeld MB, Fung JC, Sedat JW, Alberts BM, Agard DA.
J Cell Biol. 1995a Sep;130(5):1149-59.
PMID 7657699
 
Microtubule nucleation by gamma-tubulin-containing rings in the centrosome.
Moritz M, Braunfeld MB, Sedat JW, Alberts B, Agard DA.
Nature. 1995b Dec 7;378(6557):638-40.
PMID 8524401
 
The paternal inheritance of the centrosome, the cell's microtubule-organizing center, in humans, and the implications for infertility.
Simerly C, Wu GJ, Zoran S, Ord T, Rawlins R, Jones J, Navara C, Gerrity M, Rinehart J, Binor Z, Asch R, Schatten G.
Nat Med. 1995 Jan;1(1):47-52.
PMID 7584952
 
Molecular characterisation of ninein, a new coiled-coil protein of the centrosome.
Bouckson-Castaing V, Moudjou M, Ferguson DJ, Mucklow S, Belkaid Y, Milon G, Crocker PR.
J Cell Sci. 1996 Jan;109 ( Pt 1):179-90.
PMID 8834802
 
Abnormal centrosome amplification in the absence of p53.
Fukasawa K, Choi T, Kuriyama R, Rulong S, Vande Woude GF.
Science. 1996 Mar 22;271(5256):1744-7.
PMID 8596939
 
gamma-Tubulin in mammalian cells: the centrosomal and the cytosolic forms.
Moudjou M, Bordes N, Paintrand M, Bornens M.
J Cell Sci. 1996 Apr;109 ( Pt 4):875-87.
PMID 8718679
 
Phenotypic variations among paternal centrosomes expressed within the zygote as disparate microtubule lengths and sperm aster organization: correlations between centrosome activity and developmental success.
Navara CS, First NL, Schatten G.
Proc Natl Acad Sci U S A. 1996 May 28;93(11):5384-8.
PMID 8643584
 
The sperm centriole: its inheritance, replication and perpetuation in early human embryos.
Sathananthan AH, Ratnam SS, Ng SC, Tarin JJ, Gianaroli L, Trounson A.
Hum Reprod. 1996 Feb;11(2):345-56.
PMID 8671223
 
Database of p53 gene somatic mutations in human tumors and cell lines: updated compilation and future prospects.
Hainaut P, Soussi T, Shomer B, Hollstein M, Greenblatt M, Hovig E, Harris CC, Montesano R.
Nucleic Acids Res. 1997 Jan 1;25(1):151-7.
PMID 9016527
 
The human sperm centrosome is responsible for normal syngamy and early embryonic development.
Palermo GD, Colombero LT, Rosenwaks Z.
Rev Reprod. 1997 Jan;2(1):19-27. (REVIEW)
PMID 9414462
 
Mitosis in the human embryo: the vital role of the sperm centrosome (centriole).
Sathananthan AH.
Histol Histopathol. 1997 Jul;12(3):827-56. (REVIEW)
PMID 9225167
 
inheritance of sperm centrioles and centrosomes in bovine embryos.
Sathananthan AH, Tatham B, Dharmawardena V, Grills B, Lewis I, Trounson A.
Arch Androl. 1997 Jan-Feb;38(1):37-48.
PMID 9017121
 
A putative serine/threonine kinase encoding gene BTAK on chromosome 20q13 is amplified and overexpressed in human breast cancer cell lines.
Sen S, Zhou H, White RA.
Oncogene. 1997 May 8;14(18):2195-200.
PMID 9174055
 
The cell center at 100.
Stearns T, Winey M.
Cell. 1997 Oct 31;91(3):303-9.
PMID 9363939
 
A homologue of Drosophila aurora kinase is oncogenic and amplified in human colorectal cancers.
Bischoff JR, Anderson L, Zhu Y, Mossie K, Ng L, Souza B, Schryver B, Flanagan P, Clairvoyant F, Ginther C, Chan CS, Novotny M, Slamon DJ, Plowman GD.
EMBO J. 1998 Jun 1;17(11):3052-65.
PMID 9606188
 
Pericentrin and gamma-tubulin form a protein complex and are organized into a novel lattice at the centrosome.
Dictenberg JB, Zimmerman W, Sparks CA, Young A, Vidair C, Zheng Y, Carrington W, Fay FS, Doxsey SJ.
J Cell Biol. 1998 Apr 6;141(1):163-74.
PMID 9531556
 
Centrosome reduction during mouse spermiogenesis.
Manandhar G, Sutovsky P, Joshi HC, Stearns T, Schatten G.
Dev Biol. 1998 Nov 15;203(2):424-34.
PMID 9808791
 
Recruitment of the gamma-tubulin ring complex to Drosophila salt-stripped centrosome scaffolds.
Moritz M, Zheng Y, Alberts BM, Oegema K.
J Cell Biol. 1998 Aug 10;142(3):775-86.
PMID 9700165
 
The mammalian gamma-tubulin complex contains homologues of the yeast spindle pole body components spc97p and spc98p.
Murphy SM, Urbani L, Stearns T.
J Cell Biol. 1998 May 4;141(3):663-74.
PMID 9566967
 
Characterization of the human homologue of the yeast spc98p and its association with gamma-tubulin.
Tassin AM, Celati C, Moudjou M, Bornens M.
J Cell Biol. 1998 May 4;141(3):689-701.
PMID 9566969
 
Tumour amplified kinase STK15/BTAK induces centrosome amplification, aneuploidy and transformation.
Zhou H, Kuang J, Zhong L, Kuo WL, Gray JW, Sahin A, Brinkley BR, Sen S.
Nat Genet. 1998 Oct;20(2):189-93.
PMID 9771714
 
Acephalic spermatozoa and abnormal development of the head-neck attachment: a human syndrome of genetic origin.
Chemes HE, Puigdomenech ET, Carizza C, Olmedo SB, Zanchetti F, Hermes R.
Hum Reprod. 1999 Jul;14(7):1811-8.
PMID 10402395
 
Human 76p: A new member of the gamma-tubulin-associated protein family.
Fava F, Raynaud-Messina B, Leung-Tack J, Mazzolini L, Li M, Guillemot JC, Cachot D, Tollon Y, Ferrara P, Wright M.
J Cell Biol. 1999 Nov 15;147(4):857-68.
PMID 10562286
 
Cyclin-dependent kinase control of centrosome duplication.
Lacey KR, Jackson PK, Stearns T.
Proc Natl Acad Sci U S A. 1999 Mar 16;96(6):2817-22.
PMID 10077594
 
Centriole and centrin degeneration during mouse spermiogenesis.
Manandhar G, Simerly C, Salisbury JL, Schatten G.
Cell Motil Cytoskeleton. 1999;43(2):137-44.
PMID 10379838
 
Centrosome duplication in mammalian somatic cells requires E2F and Cdk2-cyclin A.
Meraldi P, Lukas J, Fry AM, Bartek J, Nigg EA.
Nat Cell Biol. 1999 Jun;1(2):88-93.
PMID 10559879
 
Characterization of two related Drosophila gamma-tubulin complexes that differ in their ability to nucleate microtubules.
Oegema K, Wiese C, Martin OC, Milligan RA, Iwamatsu A, Mitchison TJ, Zheng Y.
J Cell Biol. 1999 Feb 22;144(4):721-33.
PMID 10037793
 
Centrosomes and cancer.
Salisbury JL, Whitehead CM, Lingle WL, Barrett SL.
Biol Cell. 1999 Jul;91(6):451-60. (REVIEW)
PMID 10519005
 
AKAP350, a multiply spliced protein kinase A-anchoring protein associated with centrosomes.
Schmidt PH, Dransfield DT, Claudio JO, Hawley RG, Trotter KW, Milgram SL, Goldenring JR.
J Biol Chem. 1999 Jan 29;274(5):3055-66.
PMID 9915845
 
Biparental inheritance of gamma-tubulin during human fertilization: molecular reconstitution of functional zygotic centrosomes in inseminated human oocytes and in cell-free extracts nucleated by human sperm.
Simerly C, Zoran SS, Payne C, Dominko T, Sutovsky P, Navara CS, Salisbury JL, Schatten G.
Mol Biol Cell. 1999 Sep;10(9):2955-69.
PMID 10473639
 
Characterization of a novel giant scaffolding protein, CG-NAP, that anchors multiple signaling enzymes to centrosome and the golgi apparatus.
Takahashi M, Shibata H, Shimakawa M, Miyamoto M, Mukai H, Ono Y.
J Biol Chem. 1999 Jun 11;274(24):17267-74.
PMID 10358086
 
The centrosome.
Urbani L, Stearns T.
Curr Biol. 1999 May 6;9(9):R315-7. (REVIEW)
PMID 10322119
 
Amorphous no longer: the centrosome comes into focus.
Zimmerman W, Sparks CA, Doxsey SJ.
Curr Opin Cell Biol. 1999 Feb;11(1):122-8. (REVIEW)
PMID 10047524
 
Centrosome precursors in the acentriolar mouse oocyte.
Calarco PG.
Microsc Res Tech. 2000 Jun 1;49(5):428-34.
PMID 10842369
 
Sorting and reorganization of centrosomes during oocyte maturation in the mouse.
Carabatsos MJ, Combelles CM, Messinger SM, Albertini DF.
Microsc Res Tech. 2000 Jun 1;49(5):435-44.
PMID 10842370
 
Centrosome inheritance in sheep zygotes: centrioles are contributed by the sperm.
Crozet N, Dahirel M, Chesne P.
Microsc Res Tech. 2000 Jun 1;49(5):445-50.
PMID 10842371
 
Identification of a human centrosomal calmodulin-binding protein that shares homology with pericentrin.
Flory MR, Moser MJ, Monnat RJ Jr, Davis TN.
Proc Natl Acad Sci U S A. 2000 May 23;97(11):5919-23.
PMID 10823944
 
The TACC domain identifies a family of centrosomal proteins that can interact with microtubules.
Gergely F, Karlsson C, Still I, Cowell J, Kilmartin J, Raff JW.
Proc Natl Acad Sci U S A. 2000a Dec 19;97(26):14352-7.
PMID 11121038
 
D-TACC: a novel centrosomal protein required for normal spindle function in the early Drosophila embryo.
Gergely F, Kidd D, Jeffers K, Wakefield JG, Raff JW.
EMBO J. 2000b Jan 17;19(2):241-52.
PMID 10637228
 
Spindle formation and dynamics of gamma-tubulin and nuclear mitotic apparatus protein distribution during meiosis in pig and mouse oocytes.
Lee J, Miyano T, Moor RM.
Biol Reprod. 2000 May;62(5):1184-92.
PMID 10775165
 
Monomeric gamma -tubulin nucleates microtubules.
Leguy R, Melki R, Pantaloni D, Carlier MF.
J Biol Chem. 2000 Jul 21;275(29):21975-80.
PMID 10764751
 
Highly degenerated distal centrioles in rhesus and human spermatozoa.
Manandhar G, Simerly C, Schatten G.
Hum Reprod. 2000 Feb;15(2):256-63.
PMID 10655294
 
Microtubule minus-end anchorage at centrosomal and non-centrosomal sites: the role of ninein.
Mogensen MM, Malik A, Piel M, Bouckson-Castaing V, Bornens M.
J Cell Sci. 2000 Sep;113 ( Pt 17):3013-23.
PMID 10934040
 
Sperm centriole disfunction and sperm immotility.
Nagy ZP.
Mol Cell Endocrinol. 2000 Aug 15;166(1):59-62. (REVIEW)
PMID 10989209
 
gamma-tubulin complexes: binding to the centrosome, regulation and microtubule nucleation.
Schiebel E.
Curr Opin Cell Biol. 2000 Feb;12(1):113-8. (REVIEW)
PMID 10679351
 
A new function for the gamma-tubulin ring complex as a microtubule minus-end cap.
Wiese C, Zheng Y.
Nat Cell Biol. 2000 Jun;2(6):358-64.
PMID 10854327
 
Msps/XMAP215 interacts with the centrosomal protein D-TACC to regulate microtubule behaviour.
Lee MJ, Gergely F, Jeffers K, Peak-Chew SY, Raff JW.
Nat Cell Biol. 2001 Jul;3(7):643-9.
PMID 11433296
 
Kendrin/pericentrin-B, a centrosome protein with homology to pericentrin that complexes with PCM-1.
Li Q, Hansen D, Killilea A, Joshi HC, Palazzo RE, Balczon R.
J Cell Sci. 2001 Feb;114(Pt 4):797-809.
PMID 11171385
 
Comparative expression of the mitotic regulators SAK and PLK in colorectal cancer.
Macmillan JC, Hudson JW, Bull S, Dennis JW, Swallow CJ.
Ann Surg Oncol. 2001 Oct;8(9):729-40.
PMID 11597015
 
GCP5 and GCP6: two new members of the human gamma-tubulin complex.
Murphy SM, Preble AM, Patel UK, O'Connell KL, Dias DP, Moritz M, Agard D, Stults JT, Stearns T.
Mol Biol Cell. 2001 Nov;12(11):3340-52.
PMID 11694571
 
Outer dense fiber 2 is a widespread centrosome scaffold component preferentially associated with mother centrioles: its identification from isolated centrosomes.
Nakagawa Y, Yamane Y, Okanoue T, Tsukita S, Tsukita S.
Mol Biol Cell. 2001 Jun;12(6):1687-97.
PMID 11408577
 
Centrosome composition and microtubule anchoring mechanisms.
Bornens M.
Curr Opin Cell Biol. 2002 Feb;14(1):25-34. (REVIEW)
PMID 11792541
 
CP110, a cell cycle-dependent CDK substrate, regulates centrosome duplication in human cells.
Chen Z, Indjeian VB, McManus M, Wang L, Dynlacht BD.
Dev Cell. 2002 Sep;3(3):339-50.
PMID 12361598
 
Aurora-A overexpression reveals tetraploidization as a major route to centrosome amplification in p53-/- cells.
Meraldi P, Honda R, Nigg EA.
EMBO J. 2002 Feb 15;21(4):483-92.
PMID 11847097
 
The centrosome cycle.
Meraldi P, Nigg EA.
FEBS Lett. 2002 Jun 19;521(1-3):9-13. (REVIEW)
PMID 12067716
 
Centrosome aberrations: cause or consequence of cancer progression?
Nigg EA.
Nat Rev Cancer. 2002 Nov;2(11):815-25. (REVIEW)
PMID 12415252
 
Characterization of Cep135, a novel coiled-coil centrosomal protein involved in microtubule organization in mammalian cells.
Ohta T, Essner R, Ryu JH, Palazzo RE, Uetake Y, Kuriyama R.
J Cell Biol. 2002 Jan 7;156(1):87-99. Epub 2002 Jan 7.
PMID 11781336
 
Centrin-2 is required for centriole duplication in mammalian cells.
Salisbury JL, Suino KM, Busby R, Springett M.
Curr Biol. 2002 Aug 6;12(15):1287-92.
PMID 12176356
 
The fine structure of human embryonic stem cells.
Sathananthan H, Pera M, Trounson A.
Reprod Biomed Online. 2002 Jan-Feb;4(1):56-61.
PMID 12470354
 
Centrosomal proteins CG-NAP and kendrin provide microtubule nucleation sites by anchoring gamma-tubulin ring complex.
Takahashi M, Yamagiwa A, Nishimura T, Mukai H, Ono Y.
Mol Biol Cell. 2002 Sep;13(9):3235-45.
PMID 12221128
 
Proteomic characterization of the human centrosome by protein correlation profiling.
Andersen JS, Wilkinson CJ, Mayor T, Mortensen P, Nigg EA, Mann M.
Nature. 2003 Dec 4;426(6966):570-4.
PMID 14654843
 
Centrosome and microtubule dynamics during early stages of meiosis in mouse oocytes.
Can A, Semiz O, Cinar O.
Mol Hum Reprod. 2003 Dec;9(12):749-56.
PMID 14614036
 
Human Mps1 protein kinase is required for centrosome duplication and normal mitotic progression.
Fisk HA, Mattison CP, Winey M.
Proc Natl Acad Sci U S A. 2003 Dec 9;100(25):14875-80. Epub 2003 Dec 1.
PMID 14657364
 
The centrosomal proteins pericentrin and kendrin are encoded by alternatively spliced products of one gene.
Flory MR, Davis TN.
Genomics. 2003 Sep;82(3):401-5.
PMID 12906865
 
A novel human protein of the maternal centriole is required for the final stages of cytokinesis and entry into S phase.
Gromley A, Jurczyk A, Sillibourne J, Halilovic E, Mogensen M, Groisman I, Blomberg M, Doxsey S.
J Cell Biol. 2003 May 12;161(3):535-45. Epub 2003 May 5.
PMID 12732615
 
Maternal gamma (gamma)-tubulin is involved in microtubule reorganization during bovine fertilization and parthenogenesis.
Shin MR, Kim NH.
Mol Reprod Dev. 2003 Apr;64(4):438-45.
PMID 12589656
 
Orientation of asymmetric stem cell division by the APC tumor suppressor and centrosome.
Yamashita YM, Jones DL, Fuller MT.
Science. 2003 Sep 12;301(5639):1547-50.
PMID 12970569
 
The arithmetic of centrosome biogenesis.
Delattre M, Gonczy P.
J Cell Sci. 2004 Apr 1;117(Pt 9):1619-30. (REVIEW)
PMID 15075224
 
Outer dense fibre protein 2 (ODF2) is a self-interacting centrosomal protein with affinity for microtubules.
Donkor FF, Monnich M, Czirr E, Hollemann T, Hoyer-Fender S.
J Cell Sci. 2004 Sep 15;117(Pt 20):4643-51. Epub 2004 Aug 31.
PMID 15340007
 
The role of the centrosome in cell cycle progression.
Fry AM, Hames RS.
In: E. A. Nigg: Centrosomes in Development and Disease. Wiley-VCH. 2004; pp. 143-166.
 
Localization of gamma-tubulin in mouse eggs during meiotic maturation, fertilization, and early embryonic development.
Meng XQ, Fan HY, Zhong ZS, Zhang G, Li YL, Chen DY, Sun QY.
J Reprod Dev. 2004 Feb;50(1):97-105.
PMID 15007207
 
Elongation of centriolar microtubule triplets contributes to the formation of the mitotic spindle in gamma-tubulin-depleted cells.
Raynaud-Messina B, Mazzolini L, Moisand A, Cirinesi AM, Wright M.
J Cell Sci. 2004 Nov 1;117(Pt 23):5497-507. Epub 2004 Oct 12.
PMID 15479719
 
NuMA expression and function in mouse oocytes and early embryos.
Tang CJ, Hu HM, Tang TK.
J Biomed Sci. 2004 May-Jun;11(3):370-6.
PMID 15067221
 
Cell cycle progression after cleavage failure: mammalian somatic cells do not possess a "tetraploidy checkpoint".
Uetake Y, Sluder G.
J Cell Biol. 2004 Jun 7;165(5):609-15.
PMID 15184397
 
The centrosome in normal and transformed cells.
Wang Q, Hirohashi Y, Furuuchi K, Zhao H, Liu Q, Zhang H, Murali R, Berezov A, Du X, Li B, Greene MI.
DNA Cell Biol. 2004 Aug;23(8):475-89. (REVIEW)
PMID 15307950
 
Polo-like kinase-2 is required for centriole duplication in mammalian cells.
Warnke S, Kemmler S, Hames RS, Tsai HL, Hoffmann-Rohrer U, Fry AM, Hoffmann I.
Curr Biol. 2004 Jul 13;14(13):1200-7.
PMID 15242618
 
The centrosome in human genetic disease.
Badano JL, Teslovich TM, Katsanis N.
Nat Rev Genet. 2005 Mar;6(3):194-205. (REVIEW)
PMID 15738963
 
Microtubule nucleation and anchoring at the centrosome are independent processes linked by ninein function.
Delgehyr N, Sillibourne J, Bornens M.
J Cell Sci. 2005 Apr 15;118(Pt 8):1565-75. Epub 2005 Mar 22.
PMID 15784680
 
Aurora kinases, aneuploidy and cancer, a coincidence or a real link?
Giet R, Petretti C, Prigent C.
Trends Cell Biol. 2005 May;15(5):241-50. (REVIEW)
PMID 15866028
 
The cell biology of neurogenesis.
Gotz M, Huttner WB.
Nat Rev Mol Cell Biol. 2005 Oct;6(10):777-88. (REVIEW)
PMID 16314867
 
The Polo kinase Plk4 functions in centriole duplication.
Habedanck R, Stierhof YD, Wilkinson CJ, Nigg EA.
Nat Cell Biol. 2005 Nov;7(11):1140-6.
PMID 16244668
 
Odf2-deficient mother centrioles lack distal/subdistal appendages and the ability to generate primary cilia.
Ishikawa H, Kubo A, Tsukita S, Tsukita S.
Nat Cell Biol. 2005 May;7(5):517-24. Epub 2005 Apr 24.
PMID 15852003
 
Plk4 haploinsufficiency causes mitotic infidelity and carcinogenesis.
Ko MA, Rosario CO, Hudson JW, Kulkarni S, Pollett A, Dennis JW, Swallow CJ.
Nat Genet. 2005 Aug;37(8):883-8. Epub 2005 Jul 17.
PMID 16025114
 
SAS-6 defines a protein family required for centrosome duplication in C. elegans and in human cells.
Leidel S, Delattre M, Cerutti L, Baumer K, Gonczy P.
Nat Cell Biol. 2005 Feb;7(2):115-25.
PMID 15665853
 
Centrosome reduction during gametogenesis and its significance.
Manandhar G, Schatten H, Sutovsky P.
Biol Reprod. 2005 Jan;72(1):2-13. Epub 2004 Sep 22. (REVIEW)
PMID 15385423
 
Depletion of licensing inhibitor geminin causes centrosome overduplication and mitotic defects.
Tachibana KE, Gonzalez MA, Guarguaglini G, Nigg EA, Laskey RA.
EMBO Rep. 2005 Nov;6(11):1052-7. Epub 2005 Sep 23
PMID 16179947
 
Mammalian cells lack checkpoints for tetraploidy, aberrant centrosome number, and cytokinesis failure.
Wong C, Stearns T.
BMC Cell Biol. 2005 Feb 15;6(1):6.
PMID 15713235
 
Function of donor cell centrosome in intraspecies and interspecies nuclear transfer embryos.
Zhong ZS, Zhang G, Meng XQ, Zhang YL, Chen DY, Schatten H, Sun QY.
Exp Cell Res. 2005 May 15;306(1):35-46. Epub 2005 Mar 17.
PMID 15878330
 
Fate of centrosomes following somatic cell nuclear transfer (SCNT) in bovine oocytes.
Dai Y, Wang L, Wang H, Liu Y, Li N, Lyu Q, Keefe DL, Albertini DF, Liu L.
Reproduction. 2006 Jun;131(6):1051-61.
PMID 16735544
 
NEDD1-dependent recruitment of the gamma-tubulin ring complex to the centrosome is necessary for centriole duplication and spindle assembly.
Haren L, Remy MH, Bazin I, Callebaut I, Wright M, Merdes A.
J Cell Biol. 2006 Feb 13;172(4):505-15. Epub 2006 Feb 6.
PMID 16461362
 
Centrosome function in normal and tumor cells.
Sankaran S, Parvin JD.
J Cell Biochem. 2006 Dec 1;99(5):1240-50. (REVIEW)
PMID 16817224
 
From oogonia to mature oocytes: inactivation of the maternal centrosome in humans.
Sathananthan AH, Selvaraj K, Girijashankar ML, Ganesh V, Selvaraj P, Trounson AO.
Microsc Res Tech. 2006 Jun;69(6):396-407. (REVIEW)
PMID 16718650
 
Mechanism limiting centrosome duplication to once per cell cycle.
Tsou MF, Stearns T.
Nature. 2006a Aug 24;442(7105):947-51. Epub 2006 Jul 19.
PMID 16862117
 
Controlling centrosome number: licenses and blocks.
Tsou MF, Stearns T.
Curr Opin Cell Biol. 2006b Feb;18(1):74-8. Epub 2005 Dec 19. (REVIEW)
PMID 16361091
 
Structure and duplication of the centrosome.
Azimzadeh J, Bornens M.
J Cell Sci. 2007 Jul 1;120(Pt 13):2139-42. (REVIEW)
PMID 17591686
 
Centrosome duplication: of rules and licenses.
Nigg EA.
Trends Cell Biol. 2007 May;17(5):215-21. Epub 2007 Mar 26. (REVIEW)
PMID 17383880
 
Functionally unequal centrosomes drive spindle orientation in asymmetrically dividing Drosophila neural stem cells.
Rebollo E, Sampaio P, Januschke J, Llamazares S, Varmark H, Gonzalez C.
Dev Cell. 2007 Mar;12(3):467-74.
PMID 17336911
 
A role for a novel centrosome cycle in asymmetric cell division.
Rusan NM, Peifer M.
J Cell Biol. 2007 Apr 9;177(1):13-20. Epub 2007 Apr 2.
PMID 17403931
 
Self-organization of MTOCs replaces centrosome function during acentrosomal spindle assembly in live mouse oocytes.
Schuh M, Ellenberg J.
Cell. 2007 Aug 10;130(3):484-98.
PMID 17693257
 
From stem cell to embryo without centrioles.
Stevens NR, Raposo AA, Basto R, St Johnston D, Raff JW.
Curr Biol. 2007 Sep 4;17(17):1498-503. Epub 2007 Aug 23.
PMID 17716897
 
Functional analyses of the sperm centrosome in human reproduction: implications for assisted reproductive technique.
Terada Y.
Soc Reprod Fertil Suppl. 2007;63:507-13. (REVIEW)
PMID 17566295
 
Asymmetric inheritance of mother versus daughter centrosome in stem cell division.
Yamashita YM, Mahowald AP, Perlin JR, Fuller MT.
Science. 2007 Jan 26;315(5811):518-21.
PMID 17255513
 
Remodeling of centrosomes in intraspecies and interspecies nuclear transfer porcine embryos.
Zhong Z, Spate L, Hao Y, Li R, Lai L, Katayama M, Sun QY, Prather RS, Schatten H.
Cell Cycle. 2007 Jun 15;6(12):1510-20. Epub 2007 Apr 16.
PMID 17581281
 
Centrosome amplification can initiate tumorigenesis in flies.
Basto R, Brunk K, Vinadogrova T, Peel N, Franz A, Khodjakov A, Raff JW.
Cell. 2008 Jun 13;133(6):1032-42
PMID 18555779
 
Centrosome dysfunction in Drosophila neural stem cells causes tumors that are not due to genome instability.
Castellanos E, Dominguez P, Gonzalez C.
Curr Biol. 2008 Aug 26;18(16):1209-14. Epub 2008 Jul 24.
PMID 18656356
 
The centrosome and early embryogenesis: clinical insights.
Chatzimeletiou K, Morrison EE, Prapas N, Prapas Y, Handyside AH.
Reprod Biomed Online. 2008 Apr;16(4):485-91. (REVIEW)
PMID 18413056
 
Centrosome misorientation reduces stem cell division during ageing.
Cheng J, Turkel N, Hemati N, Fuller MT, Hunt AJ, Yamashita YM.
Nature. 2008 Dec 4;456(7222):599-604. Epub 2008 Oct 15.
PMID 18923395
 
Asymmetric mitosis: Unequal segregation of proteins destined for degradation.
Fuentealba LC, Eivers E, Geissert D, Taelman V, De Robertis EM.
Proc Natl Acad Sci U S A. 2008 Jun 3;105(22):7732-7. Epub 2008 May 29.
PMID 18511557
 
Mechanisms of asymmetric cell division: flies and worms pave the way.
Gonczy P.
Nat Rev Mol Cell Biol. 2008 May;9(5):355-66. (REVIEW)
PMID 18431399
 
Regulation of centrosomes by the BRCA1-dependent ubiquitin ligase.
Kais Z, Parvin JD.
Cancer Biol Ther. 2008 Oct;7(10):1540-3. Epub 2008 Oct 19. (REVIEW)
PMID 18927495
 
Stem cells and niches: mechanisms that promote stem cell maintenance throughout life.
Morrison SJ, Spradling AC.
Cell. 2008 Feb 22;132(4):598-611. (REVIEW)
PMID 18295578
 
Cdc14B depletion leads to centriole amplification, and its overexpression prevents unscheduled centriole duplication.
Wu J, Cho HP, Rhee DB, Johnson DK, Dunlap J, Liu Y, Wang Y.
J Cell Biol. 2008 May 5;181(3):475-83.
PMID 18458157
 
TSKS concentrates in spermatid centrioles during flagellogenesis.
Xu B, Hao Z, Jha KN, Zhang Z, Urekar C, Digilio L, Pulido S, Strauss JF 3rd, Flickinger CJ, Herr JC.
Dev Biol. 2008 Jul 15;319(2):201-10. Epub 2008 Apr 11.
PMID 18495105
 
Asymmetric centrosome behavior and the mechanisms of stem cell division.
Yamashita YM, Fuller MT.
J Cell Biol. 2008 Jan 28;180(2):261-6. Epub 2008 Jan 21. (REVIEW)
PMID 18209101
 
SnapShot: centriole biogenesis.
Bettencourt-Dias M, Glover DM.
Cell. 2009 Jan 9;136(1):188-188.e1.
PMID 19135899
 
Geminin is partially localized to the centrosome and plays a role in proper centrosome duplication.
Lu F, Lan R, Zhang H, Jiang Q, Zhang C.
Biol Cell. 2009 May;101(5):273-85.
PMID 18798731
 
Dividing cellular asymmetry: asymmetric cell division and its implications for stem cells and cancer.
Neumuller RA, Knoblich JA.
Genes Dev. 2009 Dec 1;23(23):2675-99. (REVIEW)
PMID 19952104
 
The BRCA1-dependent ubiquitin ligase, gamma-tubulin, and centrosomes.
Parvin JD.
Environ Mol Mutagen. 2009 Oct;50(8):649-53. (REVIEW)
PMID 19274767
 
The role of centrosomes in mammalian fertilization and its significance for ICSI.
Schatten H, Sun QY.
Mol Hum Reprod. 2009a Sep;15(9):531-8. Epub 2009 Jun 23. (REVIEW)
PMID 19549764
 
The functional significance of centrosomes in mammalian meiosis, fertilization, development, nuclear transfer, and stem cell differentiation.
Schatten H, Sun QY.
Environ Mol Mutagen. 2009b Oct;50(8):620-36. (REVIEW)
PMID 19402157
 
Polo kinase and separase regulate the mitotic licensing of centriole duplication in human cells.
Tsou MF, Wang WJ, George KA, Uryu K, Stearns T, Jallepalli PV.
Dev Cell. 2009 Sep;17(3):344-54.
PMID 19758559
 
Asymmetric centrosome inheritance maintains neural progenitors in the neocortex.
Wang X, Tsai JW, Imai JH, Lian WN, Vallee RB, Shi SH.
Nature. 2009 Oct 15;461(7266):947-55.
PMID 19829375
 
PLK2 phosphorylation is critical for CPAP function in procentriole formation during the centrosome cycle.
Chang J, Cizmecioglu O, Hoffmann I, Rhee K.
EMBO J. 2010 Jul 21;29(14):2395-406. Epub 2010 Jun 8.
PMID 20531387
 
Cep152 acts as a scaffold for recruitment of Plk4 and CPAP to the centrosome.
Cizmecioglu O, Arnold M, Bahtz R, Settele F, Ehret L, Haselmann-Weiss U, Antony C, Hoffmann I.
J Cell Biol. 2010 Nov 15;191(4):731-9. Epub 2010 Nov 8.
PMID 21059844
 
Procentriole assembly revealed by cryo-electron tomography.
Guichard P, Chretien D, Marco S, Tassin AM.
EMBO J. 2010 May 5;29(9):1565-72. Epub 2010 Mar 25.
PMID 20339347
 
Cep152 interacts with Plk4 and is required for centriole duplication.
Hatch EM, Kulukian A, Holland AJ, Cleveland DW, Stearns T.
J Cell Biol. 2010 Nov 15;191(4):721-9. Epub 2010 Nov 8.
PMID 21059850
 
Centriole duplication: A lesson in self-control.
Holland AJ, Lan W, Cleveland DW.
Cell Cycle. 2010 Jul 15;9(14):2731-6. Epub 2010 Jul 27.
PMID 20647763
 
Centriole maturation and transformation to basal body.
Hoyer-Fender S.
Semin Cell Dev Biol. 2010 Apr;21(2):142-7. Epub 2009 Jul 10. (REVIEW)
PMID 19595783
 
Polo-like kinase 2-dependent phosphorylation of NPM/B23 on serine 4 triggers centriole duplication.
Krause A, Hoffmann I.
PLoS One. 2010 Mar 24;5(3):e9849.
PMID 20352051
 
Neural stem cells: the need for a proper orientation.
Lesage B, Gutierrez I, Marti E, Gonzalez C.
Curr Opin Genet Dev. 2010 Aug;20(4):438-42. Epub 2010 May 25. (REVIEW)
PMID 20537889
 
Plk4/SAK/ZYG-1 in the regulation of centriole duplication.
Pearson CG, Winey M.
F1000 Biol Rep. 2010 Aug 9;2:58.
PMID 21173875
 
Plk4 is required for cytokinesis and maintenance of chromosomal stability.
Rosario CO, Ko MA, Haffani YZ, Gladdy RA, Paderova J, Pollett A, Squire JA, Dennis JW, Swallow CJ.
Proc Natl Acad Sci U S A. 2010 Apr 13;107(15):6888-93. Epub 2010 Mar 26.
PMID 20348415
 
Cytokinesis and cancer.
Sagona AP, Stenmark H.
FEBS Lett. 2010 Jun 18;584(12):2652-61. Epub 2010 Apr 3. (REVIEW)
PMID 20371245
 
The role of centrosomes in fertilization, cell division and establishment of asymmetry during embryo development.
Schatten H, Sun QY.
Semin Cell Dev Biol. 2010 Apr;21(2):174-84. Epub 2010 Jan 18. (REVIEW)
PMID 20083214
 
Polo-like kinase 4: the odd one out of the family.
Sillibourne JE, Bornens M.
Cell Div. 2010 Sep 29;5:25.
PMID 20920249
 
The nuclear mitotic apparatus (NuMA) protein: localization and dynamics in human oocytes, fertilization and early embryos.
Alvarez Sedo C, Schatten H, Combelles CM, Rawe VY.
Mol Hum Reprod. 2011 Jun;17(6):392-8. doi: 10.1093/molehr/gar009. Epub 2011 Feb 5.
PMID 21297155
 
Human embryonic stem cells suffer from centrosomal amplification.
Holubcova Z, Matula P, Sedlackova M, Vinarsky V, Dolezalova D, Barta T, Dvorak P, Hampl A.
Stem Cells. 2011 Jan;29(1):46-56. doi: 10.1002/stem.549.
PMID 20960514
 
Drosophila neuroblasts retain the daughter centrosome.
Januschke J, Llamazares S, Reina J, Gonzalez C.
Nat Commun. 2011;2:243.
PMID 21407209
 
Mitotic spindle misorientation in cancer--out of alignment and into the fire.
Pease JC, Tirnauer JS.
J Cell Sci. 2011 Apr 1;124(Pt 7):1007-16.
PMID 21402874
 
Centriole assembly and the role of Mps1: defensible or dispensable?
Pike AN, Fisk HA.
Cell Div. 2011 Apr 14;6:9.
PMID 21492451
 
The SCF-FBXW5 E3-ubiquitin ligase is regulated by PLK4 and targets HsSAS-6 to control centrosome duplication.
Puklowski A, Homsi Y, Keller D, May M, Chauhan S, Kossatz U, Grunwald V, Kubicka S, Pich A, Manns MP, Hoffmann I, Gonczy P, Malek NP.
Nat Cell Biol. 2011 Jul 3;13(8):1004-9. doi: 10.1038/ncb2282.
PMID 21725316
 
Written2011-10Sigrid Hoyer-Fender
of Zoology, Anthropology - Developmental Biology, GZMB, Ernst-Caspari-Haus, Justus-von-Liebig-Weg 11, Georg-August-University of Gottingen, Germany

Citation

This paper should be referenced as such :
Hoyer-Fender, S
Centrosomes in fertilization, early embryonic development, stem cell division,, cancer
Atlas Genet Cytogenet Oncol Haematol. 2012;16(4):306-319.
Free journal version : [ pdf ]   [ DOI ]
On line version : http://AtlasGeneticsOncology.org/Deep/CentrosomeStemCellID20105.htm