Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Genes Dev ; 36(11-12): 647-649, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35835509

RESUMEN

Polo-like kinase 4 (Plk4) is the master regulator of centriole assembly. Several evolutionarily conserved mechanisms strictly regulate Plk4 abundance and activity to ensure cells maintain a proper number of centrioles. In this issue of Genes & Development, Phan et al. (pp. 718-736) add to this growing list by describing a new mechanism of control that restricts Plk4 translation through competitive ribosome binding at upstream open reading frames (uORFs) in the mature Plk4 mRNA. Fascinatingly, this mechanism is especially critical in the development of primordial germ cells in mice that are transcriptionally hyperactive and thus exquisitely sensitive to Plk4 mRNA regulation.


Asunto(s)
Proteínas de Ciclo Celular , Centriolos , Animales , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centriolos/metabolismo , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo
2.
Cell Mol Life Sci ; 80(3): 73, 2023 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-36842139

RESUMEN

Barrier-to-autointegration factor (BAF/BANF) is a nuclear lamina protein essential for nuclear integrity, chromatin structure, and genome stability. Whereas complete loss of BAF causes lethality in multiple organisms, the A12T missense mutation of the BANF1 gene in humans causes a premature aging syndrome, called Néstor-Guillermo Progeria Syndrome (NGPS). Here, we report the first in vivo animal investigation of progeroid BAF, using CRISPR editing to introduce the NGPS mutation into the endogenous Drosophila baf gene. Progeroid BAF adults are born at expected frequencies, demonstrating that this BAF variant retains some function. However, tissue homeostasis is affected, supported by studies of the ovary, a tissue that depends upon BAF for stem cell survival and continuous oocyte production. We find that progeroid BAF causes defects in germline stem cell mitosis that delay anaphase progression and compromise chromosome segregation. We link these defects to decreased recruitment of centromeric proteins of the kinetochore, indicating dysfunction of cenBAF, a localized pool of dephosphorylated BAF produced by Protein Phosphatase PP4. We show that DNA damage increases in progenitor germ cells, which causes germ cell death due to activation of the DNA damage transducer kinase Chk2. Mitotic defects appear widespread, as aberrant chromosome segregation and increased apoptosis occur in another tissue. Together, these data highlight the importance of BAF in establishing centromeric structures critical for mitosis. Further, these studies link defects in cenBAF function to activation of a checkpoint that depletes progenitor reserves critical for tissue homeostasis, aligning with phenotypes of NGPS patients.


Asunto(s)
Drosophila , Progeria , Animales , Femenino , Humanos , Drosophila/metabolismo , Progeria/genética , Progeria/metabolismo , Proteínas de Unión al ADN/genética , Proteínas Nucleares/metabolismo , Centrómero/metabolismo , Homeostasis/genética
3.
Cell Mol Life Sci ; 78(21-22): 6775-6795, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34476544

RESUMEN

The centrosome is a tiny cytoplasmic organelle that organizes and constructs massive molecular machines to coordinate diverse cellular processes. Due to its many roles during both interphase and mitosis, maintaining centrosome homeostasis is essential to normal health and development. Centrosome instability, divergence from normal centrosome number and structure, is a common pathognomonic cellular state tightly associated with cancers and other genetic diseases. As novel connections are investigated linking the centrosome to disease, it is critical to understand the breadth of centrosome functions to inspire discovery. In this review, we provide an introduction to normal centrosome function and highlight recent discoveries that link centrosome instability to specific disease states.


Asunto(s)
Centrosoma/fisiología , Inestabilidad Cromosómica/genética , Animales , Enfermedades Genéticas Congénitas/genética , Humanos , Interfase/genética , Mitosis/genética , Neoplasias/genética , Orgánulos/genética
4.
J Cell Sci ; 130(1): 104-110, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27609833

RESUMEN

Basal cells in a simple secretory epithelium adhere to the extracellular matrix (ECM), providing contextual cues for ordered repopulation of the luminal cell layer. Early high-grade prostatic intraepithelial neoplasia (HG-PIN) tissue has enlarged nuclei and nucleoli, luminal layer expansion and genomic instability. Additional HG-PIN markers include loss of α6ß4 integrin or its ligand laminin-332, and budding of tumor clusters into laminin-511-rich stroma. We modeled the invasive budding phenotype by reducing expression of α6ß4 integrin in spheroids formed from two normal human stable isogenic prostate epithelial cell lines (RWPE-1 and PrEC 11220). These normal cells continuously spun in culture, forming multicellular spheroids containing an outer laminin-332 layer, basal cells (expressing α6ß4 integrin, high-molecular-weight cytokeratin and p63, also known as TP63) and luminal cells that secrete PSA (also known as KLK3). Basal cells were optimally positioned relative to the laminin-332 layer as determined by spindle orientation. ß4-integrin-defective spheroids contained a discontinuous laminin-332 layer corresponding to regions of abnormal budding. This 3D model can be readily used to study mechanisms that disrupt laminin-332 continuity, for example, defects in the essential adhesion receptor (ß4 integrin), laminin-332 or abnormal luminal expansion during HG-PIN progression.


Asunto(s)
Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/patología , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Humanos , Integrina alfa6beta4/metabolismo , Masculino , Modelos Biológicos , Morfogénesis , Clasificación del Tumor , Invasividad Neoplásica , Fenotipo , Próstata/metabolismo , Próstata/patología , Neoplasia Intraepitelial Prostática/metabolismo , Neoplasias de la Próstata/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Kalinina
5.
Proc Natl Acad Sci U S A ; 112(7): E657-66, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25646492

RESUMEN

Polo-like kinase 4 (Plk4) is a master regulator of centriole duplication, and its hyperactivity induces centriole amplification. Homodimeric Plk4 has been shown to be ubiquitinated as a result of autophosphorylation, thus promoting its own degradation and preventing centriole amplification. Unlike other Plks, Plk4 contains three rather than two Polo box domains, and the function of its third Polo box (PB3) is unclear. Here, we performed a functional analysis of Plk4's structural domains. Like other Plks, Plk4 possesses a previously unidentified autoinhibitory mechanism mediated by a linker (L1) near the kinase domain. Thus, autoinhibition is a conserved feature of Plks. In the case of Plk4, autoinhibition is relieved after homodimerization and is accomplished by PB3 and by autophosphorylation of L1. In contrast, autophosphorylation of the second linker promotes separation of the Plk4 homodimer. Therefore, autoinhibition delays the multiple consequences of activation until Plk4 dimerizes. These findings reveal a complex mechanism of Plk4 regulation and activation which govern the process of centriole duplication.


Asunto(s)
Proteínas de Drosophila/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Dimerización , Drosophila , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Microscopía Fluorescente , Datos de Secuencia Molecular , Electroforesis en Gel de Poliacrilamida Nativa , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Homología de Secuencia de Aminoácido , Espectrometría de Masas en Tándem , Ubiquitinación
6.
PLoS Genet ; 11(2): e1005014, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25723539

RESUMEN

The spatial organization of chromosomes within interphase nuclei is important for gene expression and epigenetic inheritance. Although the extent of physical interaction between chromosomes and their degree of compaction varies during development and between different cell-types, it is unclear how regulation of chromosome interactions and compaction relate to spatial organization of genomes. Drosophila is an excellent model system for studying chromosomal interactions including homolog pairing. Recent work has shown that condensin II governs both interphase chromosome compaction and homolog pairing and condensin II activity is controlled by the turnover of its regulatory subunit Cap-H2. Specifically, Cap-H2 is a target of the SCFSlimb E3 ubiquitin-ligase which down-regulates Cap-H2 in order to maintain homologous chromosome pairing, chromosome length and proper nuclear organization. Here, we identify Casein Kinase I alpha (CK1α) as an additional negative-regulator of Cap-H2. CK1α-depletion stabilizes Cap-H2 protein and results in an accumulation of Cap-H2 on chromosomes. Similar to Slimb mutation, CK1α depletion in cultured cells, larval salivary gland, and nurse cells results in several condensin II-dependent phenotypes including dispersal of centromeres, interphase chromosome compaction, and chromosome unpairing. Moreover, CK1α loss-of-function mutations dominantly suppress condensin II mutant phenotypes in vivo. Thus, CK1α facilitates Cap-H2 destruction and modulates nuclear organization by attenuating chromatin localized Cap-H2 protein.


Asunto(s)
Caseína Quinasa Ialfa/genética , Proteínas Cromosómicas no Histona/genética , Emparejamiento Cromosómico/genética , Proteínas de Drosophila/genética , Mitosis/genética , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Animales , Caseína Quinasa Ialfa/metabolismo , Centrómero/genética , Cromatina/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Drosophila , Proteínas de Drosophila/metabolismo , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Glándulas Salivales/metabolismo
7.
Development ; 141(15): 2978-83, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25053431

RESUMEN

The Par-3/Par-6/aPKC complex is the primary determinant of apical polarity in epithelia across animal species, but how the activity of this complex is restricted to allow polarization of the basolateral domain is less well understood. In Drosophila, several multiprotein modules antagonize the Par complex through a variety of means. Here we identify a new mechanism involving regulated protein degradation. Strong mutations in supernumerary limbs (slmb), which encodes the substrate adaptor of an SCF-class E3 ubiquitin ligase, cause dramatic loss of polarity in imaginal discs accompanied by tumorous proliferation defects. Slmb function is required to restrain apical aPKC activity in a manner that is independent of endolysosomal trafficking and parallel to the Scribble module of junctional scaffolding proteins. The involvement of the Slmb E3 ligase in epithelial polarity, specifically limiting Par complex activity to distinguish the basolateral domain, points to parallels with polarization of the C. elegans zygote.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas de Drosophila/fisiología , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteína Quinasa C/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Alelos , Animales , Proteínas de Ciclo Celular/genética , Proliferación Celular , Transformación Celular Neoplásica , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Endosomas/metabolismo , Proteínas F-Box/fisiología , Femenino , Lisosomas/metabolismo , Mutación , Fenotipo , Transporte de Proteínas , Ubiquitina-Proteína Ligasas/genética
8.
Cell Mol Life Sci ; 70(6): 1021-34, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22892665

RESUMEN

Centrosomes are organelles involved in generating and organizing the interphase microtubule cytoskeleton, mitotic spindles and cilia. At the centrosome core are a pair of centrioles, structures that act as the duplicating elements of this organelle. Centrioles function to recruit and organize pericentriolar material which nucleates microtubules. While centrioles are relatively simple in construction, the mechanics of centriole biogenesis remain an important yet poorly understood process. More mysterious still are the regulatory mechanisms that oversee centriole assembly. The fidelity of centriole duplication is critical as defects in either the assembly or number of centrioles promote aneuploidy, primary microcephaly, birth defects, ciliopathies and tumorigenesis. In addition, some pathogens employ mechanisms to promote centriole overduplication to the detriment of the host cell. This review summarizes our current understanding of this important topic, highlighting the need for further study if new therapeutics are to be developed to treat diseases arising from defects of centrosome duplication.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiología , Centriolos/fisiología , Mitosis/fisiología , Modelos Biológicos , Animales , Drosophila melanogaster , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo
9.
Mol Biol Cell ; 34(8): ar80, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37163316

RESUMEN

Polo-like kinase 4 (Plk4) is the master-regulator of centriole assembly, and cell cycle-dependent regulation of its activity maintains proper centrosome number. During most of the cell cycle, Plk4 levels are nearly undetectable due to its ability to autophosphorylate and trigger its own ubiquitin-mediated degradation. However, during mitotic exit, Plk4 forms a single aggregate on the centriole surface to stimulate centriole duplication. Whereas most Polo-like kinase family members are monomeric, Plk4 is unique because it forms homodimers. Notably, Plk4 trans-autophosphorylates a degron near its kinase domain, a critical step in autodestruction. While it is thought that the purpose of homodimerization is to promote trans-autophosphorylation, this has not been tested. Here, we generated separation-of-function Plk4 mutants that fail to dimerize and show that homodimerization creates a binding site for the Plk4 activator, Asterless. Surprisingly, however, Plk4 dimer mutants are catalytically active in cells, promote centriole assembly, and can trans-autophosphorylate through concentration-dependent condensate formation. Moreover, we mapped and then deleted the weak-interacting regions within Plk4 that mediate condensation and conclude that dimerization and condensation are not required for centriole assembly. Our findings suggest that Plk4 dimerization and condensation function simply to down-regulate Plk4 and suppress centriole overduplication.


Asunto(s)
Proteínas de Ciclo Celular , Centriolos , Centriolos/metabolismo , Dimerización , Línea Celular , Proteínas de Ciclo Celular/metabolismo , Centrosoma/metabolismo , Fosforilación
10.
Curr Biol ; 33(19): 4202-4216.e9, 2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37729913

RESUMEN

Proper centrosome number and function relies on the accurate assembly of centrioles, barrel-shaped structures that form the core duplicating elements of the organelle. The growth of centrioles is regulated in a cell cycle-dependent manner; while new daughter centrioles elongate during the S/G2/M phase, mature mother centrioles maintain their length throughout the cell cycle. Centriole length is controlled by the synchronized growth of the microtubules that ensheathe the centriole barrel. Although proteins exist that target the growing distal tips of centrioles, such as CP110 and Cep97, these proteins are generally thought to suppress centriolar microtubule growth, suggesting that distal tips may also contain unidentified counteracting factors that facilitate microtubule polymerization. Currently, a mechanistic understanding of how distal tip proteins balance microtubule growth and shrinkage to either promote daughter centriole elongation or maintain centriole length is lacking. Using a proximity-labeling screen in Drosophila cells, we identified Cep104 as a novel component of a group of evolutionarily conserved proteins that we collectively refer to as the distal tip complex (DTC). We found that Cep104 regulates centriole growth and promotes centriole elongation through its microtubule-binding TOG domain. Furthermore, analysis of Cep104 null flies revealed that Cep104 and Cep97 cooperate during spermiogenesis to align spermatids and coordinate individualization. Lastly, we mapped the complete DTC interactome and showed that Cep97 is the central scaffolding unit required to recruit DTC components to the distal tip of centrioles.


Asunto(s)
Centriolos , Proteínas Asociadas a Microtúbulos , Masculino , Animales , Centriolos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Drosophila/metabolismo , Centrosoma/metabolismo , Espermatogénesis , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
11.
Blood ; 116(16): 3108-17, 2010 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-20664058

RESUMEN

Regulated vascular endothelial growth factor (VEGF) signaling is required for proper angiogenesis, and excess VEGF signaling results in aberrantly formed vessels that do not function properly. Tumor endothelial cells have excess centrosomes and are aneuploid, properties that probably contribute to the morphologic and functional abnormalities of tumor vessels. We hypothesized that endothelial cell centrosome number is regulated by signaling via angiogenic factors, such as VEGF. We found that endothelial cells in developing vessels exposed to elevated VEGF signaling display centrosome overduplication. Signaling from VEGF, through either MEK/ERK or AKT to cyclin E/Cdk2, is amplified in association with centrosome overduplication, and blockade of relevant pathway components rescued the centrosome overduplication defect. Endothelial cells exposed to elevated FGF also had excess centrosomes, suggesting that multiple angiogenic factors regulate centrosome number. Endothelial cells with excess centrosomes survived and formed aberrant spindles at mitosis. Developing vessels exposed to elevated VEGF signaling also exhibited increased aneuploidy of endothelial cells, which is associated with cellular dysfunction. These results provide the first link between VEGF signaling and regulation of the centrosome duplication cycle, and suggest that endothelial cell centrosome overduplication contributes to aberrant angiogenesis in developing vessel networks exposed to excess angiogenic factors.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Vasos Sanguíneos/crecimiento & desarrollo , Centrosoma/metabolismo , Células Endoteliales/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Aneuploidia , Animales , Vasos Sanguíneos/metabolismo , Línea Celular , Proliferación Celular , Células Cultivadas , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Células Endoteliales/citología , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Saco Vitelino/citología
12.
Nat Cell Biol ; 7(3): 235-45, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15723056

RESUMEN

Regulation of microtubule polymerization and depolymerization is required for proper cell development. Here, we report that two proteins of the Drosophila melanogaster kinesin-13 family, KLP10A and KLP59C, cooperate to drive microtubule depolymerization in interphase cells. Analyses of microtubule dynamics in S2 cells depleted of these proteins indicate that both proteins stimulate depolymerization, but alter distinct parameters of dynamic instability; KLP10A stimulates catastrophe (a switch from growth to shrinkage) whereas KLP59C suppresses rescue (a switch from shrinkage to growth). Moreover, immunofluorescence and live analyses of cells expressing tagged kinesins reveal that KLP10A and KLP59C target to polymerizing and depolymerizing microtubule plus ends, respectively. Our data also suggest that KLP10A is deposited on microtubules by the plus-end tracking protein, EB1. Our findings support a model in which these two members of the kinesin-13 family divide the labour of microtubule depolymerization.


Asunto(s)
Interfase , Cinesinas/fisiología , Microtúbulos/ultraestructura , Animales , Western Blotting , Línea Celular , Drosophila , Drosophila melanogaster , Glutatión Transferasa/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Cinesinas/química , Cinesinas/metabolismo , Microscopía Fluorescente , Modelos Biológicos , Polímeros/química , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Bicatenario/química , Factores de Tiempo
13.
J Cell Biol ; 177(2): 231-42, 2007 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-17452528

RESUMEN

Chromosomes move toward mitotic spindle poles by a Pacman-flux mechanism linked to microtubule depolymerization: chromosomes actively depolymerize attached microtubule plus ends (Pacman) while being reeled in to spindle poles by the continual poleward flow of tubulin subunits driven by minus-end depolymerization (flux). We report that Pacman-flux in Drosophila melanogaster incorporates the activities of three different microtubule severing enzymes, Spastin, Fidgetin, and Katanin. Spastin and Fidgetin are utilized to stimulate microtubule minus-end depolymerization and flux. Both proteins concentrate at centrosomes, where they catalyze the turnover of gamma-tubulin, consistent with the hypothesis that they exert their influence by releasing stabilizing gamma-tubulin ring complexes from minus ends. In contrast, Katanin appears to function primarily on anaphase chromosomes, where it stimulates microtubule plus-end depolymerization and Pacman-based chromatid motility. Collectively, these findings reveal novel and significant roles for microtubule severing within the spindle and broaden our understanding of the molecular machinery used to move chromosomes.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Cromosomas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Microtúbulos/metabolismo , Huso Acromático/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Anafase , Animales , Línea Celular , Centrosoma/metabolismo , Segregación Cromosómica , Drosophila melanogaster/citología , Katanina , Metafase , Proteínas Asociadas a Microtúbulos , Proteínas Nucleares/metabolismo , Tubulina (Proteína)/metabolismo
14.
J Cell Biol ; 176(5): 641-51, 2007 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-17325206

RESUMEN

Dynactin links cytoplasmic dynein and other motors to cargo and is involved in organizing radial microtubule arrays. The largest subunit of dynactin, p150(glued), binds the dynein intermediate chain and has an N-terminal microtubule-binding domain. To examine the role of microtubule binding by p150(glued), we replaced the wild-type p150(glued) in Drosophila melanogaster S2 cells with mutant DeltaN-p150 lacking residues 1-200, which is unable to bind microtubules. Cells treated with cytochalasin D were used for analysis of cargo movement along microtubules. Strikingly, although the movement of both membranous organelles and messenger ribonucleoprotein complexes by dynein and kinesin-1 requires dynactin, the substitution of full-length p150(glued) with DeltaN-p150(glued) has no effect on the rate, processivity, or step size of transport. However, truncation of the microtubule-binding domain of p150(glued) has a dramatic effect on cell division, resulting in the generation of multipolar spindles and free microtubule-organizing centers. Thus, dynactin binding to microtubules is required for organizing spindle microtubule arrays but not cargo motility in vivo.


Asunto(s)
Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Secuencia de Aminoácidos , Animales , Sitios de Unión , Transporte Biológico/fisiología , Línea Celular , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/ultraestructura , Complejo Dinactina , Dineínas/metabolismo , Dineínas/fisiología , Cinesinas/fisiología , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/química , Centro Organizador de los Microtúbulos/metabolismo , Centro Organizador de los Microtúbulos/ultraestructura , Peroxisomas/fisiología , Estructura Terciaria de Proteína , Interferencia de ARN , Eliminación de Secuencia , Huso Acromático/metabolismo , Huso Acromático/ultraestructura
15.
Diagnostics (Basel) ; 12(3)2022 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-35328229

RESUMEN

The microenvironment of solid tumors is dynamic and frequently contains pockets of low oxygen levels (hypoxia) surrounded by oxygenated tissue. Indeed, a compromised vasculature is a hallmark of the tumor microenvironment, creating both spatial gradients and temporal variability in oxygen availability. Notably, hypoxia associates with increased metastasis and poor survival in patients. Therefore, to aid therapeutic decisions and better understand hypoxia's role in cancer progression, it is critical to identify endogenous biomarkers of hypoxia to spatially phenotype oncogenic lesions in human tissue, whether precancerous, benign, or malignant. Here, we characterize the glucose transporter GLUT3/SLC2A3 as a biomarker of hypoxic prostate epithelial cells and prostate tumors. Transcriptomic analyses of non-tumorigenic, immortalized prostate epithelial cells revealed a highly significant increase in GLUT3 expression under hypoxia. Additionally, GLUT3 protein increased 2.4-fold in cultured hypoxic prostate cell lines and was upregulated within hypoxic regions of xenograft tumors, including two patient-derived xenografts (PDX). Finally, GLUT3 out-performs other established hypoxia markers; GLUT3 staining in PDX specimens detects 2.6-8.3 times more tumor area compared to a mixture of GLUT1 and CA9 antibodies. Therefore, given the heterogeneous nature of tumors, we propose adding GLUT3 to immunostaining panels when trying to detect hypoxic regions in prostate samples.

16.
Traffic ; 10(5): 472-81, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19192251

RESUMEN

Tight regulation of centrosome duplication is critical to ensure that centrosome number doubles once and only once per cell cycle. Superimposed onto this centrosome duplication cycle is a functional centrosome cycle in which they alternate between phases of quiescence and robust microtubule (MT) nucleation and MT-anchoring activities. In vertebrate cycling cells, interphase centrioles accumulate less pericentriolar material (PCM), reducing their MT nucleation capacity. In mitosis, centrosomes mature, accumulating more PCM to increase their nucleation and anchoring capacities to form robust MT asters. Interestingly, functional cycles of centrosomes can be altered to suit the cell's needs. Some interphase centrosomes function as a microtubule-organizing center by increasing their ability to anchor MTs to form centrosomal radial arrays. Other interphase centrosomes maintain their MT nucleation capacity but reduce/eliminate their MT-anchoring capacity. Recent work demonstrates that Drosophila cells take this to the extreme, whereby centrioles lose all detectable PCM during interphase, offering an explanation as to how centrosome-deficient flies develop to adulthood. Drosophila stem cells further modify the functional cycle by differentially regulating their two centrioles - a situation that seems important for stem cell asymmetric divisions, as misregulation of centrosome duplication in stem/progenitor cells can promote tumor formation. Here, we review recent findings that describe variations in the functional cycle of centrosomes.


Asunto(s)
Centrosoma/metabolismo , Secuencia de Aminoácidos , Animales , Ciclo Celular , División Celular , Centriolos/metabolismo , Centriolos/fisiología , Centrosoma/fisiología , Drosophila/metabolismo , Interfase/fisiología , Centro Organizador de los Microtúbulos/metabolismo , Microtúbulos/metabolismo , Mitosis , Células Madre/metabolismo
17.
Nature ; 427(6972): 364-70, 2004 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-14681690

RESUMEN

During anaphase identical sister chromatids separate and move towards opposite poles of the mitotic spindle. In the spindle, kinetochore microtubules have their plus ends embedded in the kinetochore and their minus ends at the spindle pole. Two models have been proposed to account for the movement of chromatids during anaphase. In the 'Pac-Man' model, kinetochores induce the depolymerization of kinetochore microtubules at their plus ends, which allows chromatids to move towards the pole by 'chewing up' microtubule tracks. In the 'poleward flux' model, kinetochores anchor kinetochore microtubules and chromatids are pulled towards the poles through the depolymerization of kinetochore microtubules at the minus ends. Here, we show that two functionally distinct microtubule-destabilizing KinI kinesin enzymes (so named because they possess a kinesin-like ATPase domain positioned internally within the polypeptide) are responsible for normal chromatid-to-pole motion in Drosophila. One of them, KLP59C, is required to depolymerize kinetochore microtubules at their kinetochore-associated plus ends, thereby contributing to chromatid motility through a Pac-Man-based mechanism. The other, KLP10A, is required to depolymerize microtubules at their pole-associated minus ends, thereby moving chromatids by means of poleward flux.


Asunto(s)
Anafase , Cromátides/fisiología , Segregación Cromosómica , Proteínas de Drosophila/metabolismo , Cinesinas/metabolismo , Mitosis , Anafase/efectos de los fármacos , Animales , Cromátides/efectos de los fármacos , Emparejamiento Cromosómico/efectos de los fármacos , Segregación Cromosómica/efectos de los fármacos , Cromosomas/efectos de los fármacos , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Cinesinas/antagonistas & inhibidores , Cinesinas/genética , Mitosis/efectos de los fármacos , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo
18.
Trends Cancer ; 6(8): 627-630, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32291237

RESUMEN

Anticancer agents often cause drug-induced tetraploidy (DIT) in cancer cells. DIT is not only a mechanism of inherited drug resistance, but proliferating DIT cells can produce progeny with increased ploidy or aneuploid genomes that drive aggressive disease. Here, we explore combinatorial therapeutic strategies for either preventing or eliminating DIT cells.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Segregación Cromosómica/efectos de los fármacos , Linfoma no Hodgkin/tratamiento farmacológico , Tetraploidía , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Inestabilidad Cromosómica/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Humanos , Linfoma no Hodgkin/genética
19.
Oncogene ; 39(2): 399-413, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31477840

RESUMEN

Localized, nonindolent prostate cancer (PCa) is characterized by large-scale genomic rearrangements, aneuploidy, chromothripsis, and other forms of chromosomal instability (CIN), yet how this occurs remains unclear. A well-established mechanism of CIN is the overproduction of centrosomes, which promotes tumorigenesis in various mouse models. Therefore, we developed a single-cell assay for quantifying centrosomes in human prostate tissue. Surprisingly, centrosome loss-which has not been described in human cancer-was associated with PCa progression. By chemically or genetically inducing centrosome loss in nontumorigenic prostate epithelial cells, mitotic errors ensued, producing aneuploid, and multinucleated cells. Strikingly, transient or chronic centrosome loss transformed prostate epithelial cells, which produced highly proliferative and poorly differentiated malignant tumors in mice. Our findings suggest that centrosome loss could create a cellular crisis with oncogenic potential in prostate epithelial cells.


Asunto(s)
Centrosoma/metabolismo , Inestabilidad Genómica , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Aneuploidia , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Fragmentación del ADN , Células Epiteliales/patología , Humanos , Masculino , Ratones , Mitosis/genética , Próstata/patología
20.
J Cell Biol ; 219(2)2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31841145

RESUMEN

During centriole duplication, a preprocentriole forms at a single site on the mother centriole through a process that includes the hierarchical recruitment of a conserved set of proteins, including the Polo-like kinase 4 (Plk4), Ana2/STIL, and the cartwheel protein Sas6. Ana2/STIL is critical for procentriole assembly, and its recruitment is controlled by the kinase activity of Plk4, but how this works remains poorly understood. A structural motif called the G-box in the centriole outer wall protein Sas4 interacts with a short region in the N terminus of Ana2/STIL. Here, we show that binding of Ana2 to the Sas4 G-box enables hyperphosphorylation of the Ana2 N terminus by Plk4. Hyperphosphorylation increases the affinity of the Ana2-G-box interaction, and, consequently, promotes the accumulation of Ana2 at the procentriole to induce daughter centriole formation.


Asunto(s)
Proteínas de Ciclo Celular/genética , Centriolos/genética , Proteínas de Drosophila/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Ciclo Celular/genética , Línea Celular , Drosophila melanogaster/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Asociadas a Microtúbulos/genética , Fosforilación/genética , Unión Proteica/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA