Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Elife ; 122023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36656118

RESUMO

Trisomy 21, the genetic cause of Down syndrome, disrupts primary cilia formation and function, in part through elevated Pericentrin, a centrosome protein encoded on chromosome 21. Yet how trisomy 21 and elevated Pericentrin disrupt cilia-related molecules and pathways, and the in vivo phenotypic relevance remain unclear. Utilizing ciliogenesis time course experiments combined with light microscopy and electron tomography, we reveal that chromosome 21 polyploidy elevates Pericentrin and microtubules away from the centrosome that corral MyosinVA and EHD1, delaying ciliary membrane delivery and mother centriole uncapping essential for ciliogenesis. If given enough time, trisomy 21 cells eventually ciliate, but these ciliated cells demonstrate persistent trafficking defects that reduce transition zone protein localization and decrease sonic hedgehog signaling in direct anticorrelation with Pericentrin levels. Consistent with cultured trisomy 21 cells, a mouse model of Down syndrome with elevated Pericentrin has fewer primary cilia in cerebellar granule neuron progenitors and thinner external granular layers at P4. Our work reveals that elevated Pericentrin from trisomy 21 disrupts multiple early steps of ciliogenesis and creates persistent trafficking defects in ciliated cells. This pericentrosomal crowding mechanism results in signaling deficiencies consistent with the neurological phenotypes found in individuals with Down syndrome.


Human cells typically have 23 pairs of structures known as chromosomes. Each chromosome contains a unique set of genes which provide the instructions needed to make proteins and other essential molecules found in the body. Individuals with Down syndrome have an extra copy of chromosome 21. This genetic alteration is known as trisomy 21 and affects many different organs in the body, leading to various medical conditions including intellectual disability, heart defects, and immune deficiencies. A recent study showed that cells from individuals with Down syndrome had defects in forming primary cilia ­ structures on the surface of cells which work as signaling hubs to control how cells grow and develop. These cilia defects were in large part due to excess levels of a protein known as Pericentrin, which is encoded by a gene found on chromosome 21. But it is unclear how Pericentrin disrupts cilia assembly, and how this may contribute to the medical conditions observed in individuals with Down syndrome. To address these questions, Jewett et al. studied human cells that had been engineered to have trisomy 21. The experiments found that trisomy 21 led to higher levels of Pericentrin and altered the way molecules were organized at the sites where primary cilia form. This caused the components required to build and maintain the primary cilium to become trapped in the wrong locations. The trisomy 21 cells were eventually able to rearrange the molecules and build a primary cilium, but it took them twice as long as cells with 23 pairs of chromosomes and their primary cilium did not properly work. Further experiments were then conducted on mice that had been engineered to have an extra copy of a portion of genes on human chromosome 21, including the gene for Pericentrin. Jewett et al. found that these mice assembled cilia later and had defects in cilia signaling, similar to the human trisomy 21 cells. This resulted in mild abnormalities in brain development that were consistent with what occurs in individuals with Down syndrome. These findings suggest that the elevated levels of Pericentrin in trisomy 21 causes changes in cilia formation and function which, in turn, may alter how the mouse brain develops. Further studies will be required to find out whether defects in primary cilia may contribute to other medical conditions observed in individuals with Down syndrome.


Assuntos
Síndrome de Down , Camundongos , Animais , Proteínas Hedgehog/metabolismo , Centríolos/metabolismo , Centrossomo/metabolismo , Cílios/metabolismo
2.
Mol Biol Cell ; 33(8)2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35476505

RESUMO

Trisomy 21, the source of Down syndrome, causes a 0.5-fold protein increase of the chromosome 21-resident gene Pericentrin (PCNT) and reduces primary cilia formation and signaling. We investigate how PCNT imbalances disrupt cilia. Using isogenic RPE-1 cells with increased chromosome 21 dosage, we find PCNT accumulates around the centrosome as a cluster of enlarged cytoplasmic puncta that localize along microtubules (MTs) and at MT ends. Cytoplasmic PCNT puncta impact the density, stability, and localization of the MT trafficking network required for primary cilia. The PCNT puncta appear to sequester cargo peripheral to centrosomes in what we call pericentrosomal crowding. The centriolar satellite proteins PCM1, CEP131, and CEP290, important for ciliogenesis, accumulate at enlarged PCNT puncta in trisomy 21 cells. Reducing PCNT when chromosome 21 ploidy is elevated is sufficient to decrease PCNT puncta and pericentrosomal crowding, reestablish a normal density of MTs around the centrosome, and restore ciliogenesis to wild-type levels. A transient reduction in MTs also decreases pericentrosomal crowding and partially rescues ciliogenesis in trisomy 21 cells, indicating that increased PCNT leads to defects in the MT network deleterious to normal centriolar satellite distribution. We propose that chromosome 21 aneuploidy disrupts MT-dependent intracellular trafficking required for primary cilia.


Assuntos
Síndrome de Down , Antígenos de Neoplasias/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centríolos/metabolismo , Centrossomo/metabolismo , Cílios/metabolismo , Proteínas do Citoesqueleto/metabolismo , Síndrome de Down/metabolismo , Humanos , Microtúbulos/metabolismo
3.
Curr Biol ; 31(7): R337-R340, 2021 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-33848487

RESUMO

Extra centrosomes are linked to cancer-associated errors in cell division, metastasis and signaling. A new study reveals that centrosome amplification disrupts lysosome function, leading to the release of small extracellular vesicles and to invasive activity in pancreatic cells.


Assuntos
Vesículas Extracelulares , Neoplasias , Biologia , Divisão Celular , Centrossomo , Humanos
4.
Dev Cell ; 56(3): 325-340.e8, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33561422

RESUMO

Primary cilia are sensory organelles that utilize the compartmentalization of membrane and cytoplasm to communicate signaling events, and yet, how the formation of a cilium is coordinated with reorganization of the cortical membrane and cytoskeleton is unclear. Using polarized epithelia, we find that cortical actin clearing and apical membrane partitioning occur where the centrosome resides at the cell surface prior to ciliation. RAB19, a previously uncharacterized RAB, associates with the RAB-GAP TBC1D4 and the HOPS-tethering complex to coordinate cortical clearing and ciliary membrane growth, which is essential for ciliogenesis. This RAB19-directed pathway is not exclusive to polarized epithelia, as RAB19 loss in nonpolarized cell types blocks ciliogenesis with a docked ciliary vesicle. Remarkably, inhibiting actomyosin contractility can substitute for the function of the RAB19 complex and restore ciliogenesis in knockout cells. Together, this work provides a mechanistic understanding behind a cytoskeletal clearing and membrane partitioning step required for ciliogenesis.


Assuntos
Membrana Celular/metabolismo , Cílios/metabolismo , Organogênese , Proteínas rab de Ligação ao GTP/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular , Polaridade Celular , Centrossomo/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Proteínas Ativadoras de GTPase , Humanos , Espaço Intracelular/metabolismo , Complexos Multiproteicos/metabolismo , Ligação Proteica , Transporte Proteico
5.
J Cell Sci ; 133(14)2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32591487

RESUMO

Microtubules (MTs) promote important cellular functions including migration, intracellular trafficking, and chromosome segregation. The centrosome, comprised of two centrioles surrounded by the pericentriolar material (PCM), is the cell's central MT-organizing center. Centrosomes in cancer cells are commonly numerically amplified. However, the question of how the amplification of centrosomes alters MT organization capacity is not well studied. We developed a quantitative image-processing and machine learning-aided approach for the semi-automated analysis of MT organization. We designed a convolutional neural network-based approach for detecting centrosomes, and an automated pipeline for analyzing MT organization around centrosomes, encapsulated in a semi-automatic graphical tool. Using this tool, we find that breast cancer cells with supernumerary centrosomes not only have more PCM protein per centrosome, which gradually increases with increasing centriole numbers, but also exhibit expansion in PCM size. Furthermore, cells with amplified centrosomes have more growing MT ends, higher MT density and altered spatial distribution of MTs around amplified centrosomes. Thus, the semi-automated approach developed here enables rapid and quantitative analyses revealing important facets of centrosomal aberrations.


Assuntos
Centríolos , Centrossomo , Segregação de Cromossomos , Aprendizado de Máquina , Microtúbulos
6.
Mol Biol Cell ; 30(10): 1230-1244, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30811267

RESUMO

The centrosome, composed of two centrioles surrounded by pericentriolar material, is the cell's central microtubule-organizing center. Centrosome duplication is coupled with the cell cycle such that centrosomes duplicate once in S phase. Loss of such coupling produces supernumerary centrosomes, a condition called centrosome amplification (CA). CA promotes cell invasion and chromosome instability, two hallmarks of cancer. We examined the contribution of centriole overduplication to CA and the consequences for genomic stability in breast cancer cells. CEP135, a centriole assembly protein, is dysregulated in some breast cancers. We previously identified a short isoform of CEP135, CEP135mini, that represses centriole duplication. Here, we show that the relative level of full-length CEP135 (CEP135full) to CEP135mini (the CEP135full:mini ratio) is increased in breast cancer cell lines with high CA. Inducing expression of CEP135full in breast cancer cells increases the frequency of CA, multipolar spindles, anaphase-lagging chromosomes, and micronuclei. Conversely, inducing expression of CEP135mini reduces centrosome number. The differential expression of the CEP135 isoforms in vivo is generated by alternative polyadenylation. Directed genetic mutations near the CEP135mini alternative polyadenylation signal reduces the CEP135full:mini ratio and decreases CA. We conclude that dysregulation of CEP135 isoforms promotes centriole overduplication and contributes to chromosome segregation errors in breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteínas de Transporte/metabolismo , Centrossomo/metabolismo , Aneuploidia , Proteínas de Transporte/genética , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Centríolos/metabolismo , Instabilidade Cromossômica , Segregação de Cromossomos/genética , Segregação de Cromossomos/fisiologia , Feminino , Instabilidade Genômica , Humanos , Mitose/fisiologia , Isoformas de Proteínas , Fase S/fisiologia
7.
J Cell Biol ; 217(12): 4298-4313, 2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30217954

RESUMO

Cilia, essential motile and sensory organelles, have several compartments: the basal body, transition zone, and the middle and distal axoneme segments. The distal segment accommodates key functions, including cilium assembly and sensory activities. While the middle segment contains doublet microtubules (incomplete B-tubules fused to complete A-tubules), the distal segment contains only A-tubule extensions, and its existence requires coordination of microtubule length at the nanometer scale. We show that three conserved proteins, two of which are mutated in the ciliopathy Joubert syndrome, determine the geometry of the distal segment, by controlling the positions of specific microtubule ends. FAP256/CEP104 promotes A-tubule elongation. CHE-12/Crescerin and ARMC9 act as positive and negative regulators of B-tubule length, respectively. We show that defects in the distal segment dimensions are associated with motile and sensory deficiencies of cilia. Our observations suggest that abnormalities in distal segment organization cause a subset of Joubert syndrome cases.


Assuntos
Proteínas do Domínio Armadillo/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cílios/metabolismo , Microtúbulos/metabolismo , Proteínas de Protozoários/metabolismo , Tetrahymena thermophila/metabolismo , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Proteínas do Domínio Armadillo/genética , Proteínas de Ciclo Celular/genética , Cerebelo/anormalidades , Cerebelo/metabolismo , Cílios/genética , Anormalidades do Olho/genética , Anormalidades do Olho/metabolismo , Humanos , Doenças Renais Císticas/genética , Doenças Renais Císticas/metabolismo , Microtúbulos/genética , Proteínas de Protozoários/genética , Retina/anormalidades , Retina/metabolismo , Tetrahymena thermophila/genética
8.
Oncotarget ; 8(4): 6742-6762, 2017 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-28039486

RESUMO

Increased expression of CD147 in pancreatic cancer has been proposed to play a critical role in cancer progression via CD147 chaperone function for lactate monocarboxylate transporters (MCTs). Here, we show for the first time that CD147 interacts with membrane transporters beyond MCTs and exhibits a protective role for several of its interacting partners. CD147 prevents its interacting partner's proteasome-dependent degradation and incorrect plasma membrane localization through the CD147 transmembrane (TM) region. The interactions with transmembrane small molecule and ion transporters identified here indicate a central role of CD147 in pancreatic cancer metabolic reprogramming, particularly with respect to amino acid anabolism and calcium signaling. Importantly, CD147 genetic ablation prevents pancreatic cancer cell proliferation and tumor growth in vitro and in vivo in conjunction with metabolic rewiring towards amino acid anabolism, thus paving the way for future combined pharmacological treatments.


Assuntos
Basigina/metabolismo , Reprogramação Celular , Metabolismo Energético , Proteínas de Membrana Transportadoras/metabolismo , Neoplasias Pancreáticas/metabolismo , Aminoácidos/metabolismo , Animais , Basigina/genética , Sinalização do Cálcio , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Membrana Transportadoras/genética , Camundongos Nus , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Proteólise , Interferência de RNA , Fatores de Tempo , Transfecção , Carga Tumoral
9.
Genetics ; 203(2): 649-65, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27270699

RESUMO

Tetrahymena thermophila is a ciliate model organism whose study has led to important discoveries and insights into both conserved and divergent biological processes. In this review, we describe the tools for the use of Tetrahymena as a model eukaryote, including an overview of its life cycle, orientation to its evolutionary roots, and methodological approaches to forward and reverse genetics. Recent genomic tools have expanded Tetrahymena's utility as a genetic model system. With the unique advantages that Tetrahymena provide, we argue that it will continue to be a model organism of choice.


Assuntos
Genes de Protozoários , Tetrahymena/genética , Técnicas Genéticas , Tetrahymena/crescimento & desenvolvimento , Tetrahymena/fisiologia
10.
Curr Biol ; 25(19): 2591-6, 2015 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-26412126

RESUMO

Centriole duplication is coordinated such that a single round of duplication occurs during each cell cycle. Disruption of this synchrony causes defects including supernumerary centrosomes in cancer and perturbed ciliary signaling [1-5]. To preserve the normal number of centrioles, the level, localization, and post-translational modification of centriole proteins is regulated so that, when centriole protein expression and/or activity are increased, centrioles self-assemble. Assembly is initiated by the formation of the cartwheel structure that comprises the base of centrioles [6-11]. SAS-6 constitutes the cartwheel, and SAS-6 levels remain low until centriole assembly is initiated at S phase onset [3, 12, 13]. CEP135 physically links to SAS-6 near the site of microtubule nucleation and binds to CPAP for triplet microtubule formation [13, 14]. We identify two distinct protein isoforms of CEP135 that antagonize each other to modulate centriole duplication: full-length CEP135 (CEP135(full)) promotes new assembly, whereas a short isoform, CEP135(mini), represses it. CEP135(mini) represses centriole duplication by limiting the centriolar localization of CEP135(full) binding proteins (SAS-6 and CPAP) and the pericentriolar localization of γ-tubulin. The CEP135 isoforms exhibit distinct and complementary centrosomal localization during the cell cycle. CEP135(mini) protein decreases from centrosomes upon anaphase onset. We suggest that the decrease in CEP135(mini) from centrosomes promotes centriole assembly. The repression of centriole duplication by a splice isoform of a protein that normally promotes it serves as a novel mechanism to limit centriole duplication.


Assuntos
Proteínas de Transporte/metabolismo , Centríolos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/metabolismo , Centríolos/genética , Centrossomo/metabolismo , Células HeLa , Humanos , Microtúbulos/metabolismo , Ligação Proteica , Isoformas de Proteínas , Splicing de RNA , Fase S , Tubulina (Proteína)/metabolismo
11.
PLoS One ; 8(8): e71225, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23936495

RESUMO

Extracellular vesicles (EVs) are key contributors to cancer where they play an integral role in cell-cell communication and transfer pro-oncogenic molecules to recipient cells thereby conferring a cancerous phenotype. Here, we purified EVs using straightforward biochemical approaches from multiple cancer cell lines and subsequently characterized these EVs via multiple biochemical and biophysical methods. In addition, we used fluorescence microscopy to directly show internalization of EVs into the recipient cells within a few minutes upon addition of EVs to recipient cells. We confirmed that the transmembrane protein EMMPRIN, postulated to be a marker of EVs, was indeed secreted from all cell lines studied here. We evaluated the response to EV stimulation in several different types of recipient cells lines and measured the ability of these purified EVs to induce secretion of several factors highly upregulated in human cancers. Our data indicate that purified EVs preferentially stimulate secretion of several proteins implicated in driving cancer in monocytic cells but only harbor limited activity in epithelial cells. Specifically, we show that EVs are potent stimulators of MMP-9, IL-6, TGF-ß1 and induce the secretion of extracellular EMMPRIN, which all play a role in driving immune evasion, invasion and inflammation in the tumor microenvironment. Thus, by using a comprehensive approach that includes biochemical, biological, and spectroscopic methods, we have begun to elucidate the stimulatory roles.


Assuntos
Basigina/metabolismo , Espaço Extracelular/metabolismo , Interleucina-6/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Retroalimentação Fisiológica , Humanos , Transporte Proteico , Regulação para Cima
12.
F1000 Biol Rep ; 2: 58, 2010 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-21173875

RESUMO

Centrioles organize both centrosomes and cilia. Centriole duplication is tightly regulated and coordinated with the cell cycle to limit duplication to only once per cell cycle. Defects in centriole number and structure are commonly found in cancer. Plk4/SAK and the functionally related Caenorhabditis elegans ZYG-1 kinases initiate centriole duplication. Several recent studies have elucidated the regulated activity of these kinases and potential downstream targets for centriole assembly.

13.
Nat Genet ; 39(6): 727-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17468754

RESUMO

Jeune asphyxiating thoracic dystrophy, an autosomal recessive chondrodysplasia, often leads to death in infancy because of a severely constricted thoracic cage and respiratory insufficiency; retinal degeneration, cystic renal disease and polydactyly may be complicating features. We show that IFT80 mutations underlie a subset of Jeune asphyxiating thoracic dystrophy cases, establishing the first association of a defective intraflagellar transport (IFT) protein with human disease. Knockdown of ift80 in zebrafish resulted in cystic kidneys, and knockdown in Tetrahymena thermophila produced shortened or absent cilia.


Assuntos
Asfixia/genética , Doenças do Desenvolvimento Ósseo/genética , Proteínas de Transporte/genética , Doenças Renais Císticas/genética , Mutação/genética , Tetrahymena thermophila/genética , Doenças Torácicas/genética , Peixe-Zebra/genética , Animais , Feminino , Humanos , Recém-Nascido , Masculino , Linhagem , Polidactilia/genética , Tetrahymena thermophila/crescimento & desenvolvimento , Peixe-Zebra/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA