Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 245
Filtrar
1.
Elife ; 122023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37737843

RESUMO

The primary cilium plays important roles in regulating cell differentiation, signal transduction, and tissue organization. Dysfunction of the primary cilium can lead to ciliopathies and cancer. The formation and organization of the primary cilium are highly associated with cell polarity proteins, such as the apical polarity protein CRB3. However, the molecular mechanisms by which CRB3 regulates ciliogenesis and the location of CRB3 remain unknown. Here, we show that CRB3, as a navigator, regulates vesicle trafficking in γ-tubulin ring complex (γTuRC) assembly during ciliogenesis and cilium-related Hh and Wnt signaling pathways in tumorigenesis. Crb3 knockout mice display severe defects of the primary cilium in the mammary ductal lumen and renal tubule, while mammary epithelial-specific Crb3 knockout mice exhibit the promotion of ductal epithelial hyperplasia and tumorigenesis. CRB3 is essential for lumen formation and ciliary assembly in the mammary epithelium. We demonstrate that CRB3 localizes to the basal body and that CRB3 trafficking is mediated by Rab11-positive endosomes. Significantly, CRB3 interacts with Rab11 to navigate GCP6/Rab11 trafficking vesicles to CEP290, resulting in intact γTuRC assembly. In addition, CRB3-depleted cells are unresponsive to the activation of the Hh signaling pathway, while CRB3 regulates the Wnt signaling pathway. Therefore, our studies reveal the molecular mechanisms by which CRB3 recognizes Rab11-positive endosomes to facilitate ciliogenesis and regulates cilium-related signaling pathways in tumorigenesis.


Assuntos
Carcinogênese , Centro Organizador dos Microtúbulos , Animais , Camundongos , Corpos Basais , Diferenciação Celular , Transformação Celular Neoplásica , Hiperplasia
2.
Methods Cell Biol ; 178: 1-12, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37516519

RESUMO

The immunological synapse (IS) between NK cells and cancer cells is instrumental for the initiation of tumor-specific cytotoxicity. Improper function of processes at the IS can lead to NK cell unresponsiveness, contributing to tumor immune escape. Critical steps at the IS include target cell recognition, conjugation of NK cell and cancer cell, cytotoxic granule convergence to the microtubule-organizing center (MTOC), granule polarization to the IS, and degranulation. Here, we describe confocal live-cell imaging methods for the analysis of these processes at the immunological synapse, with a focus on mechanisms of cancer cell resistance facilitating escape from NK cell cytotoxicity.


Assuntos
Sinapses Imunológicas , Células Matadoras Naturais , Grânulos Citoplasmáticos , Centro Organizador dos Microtúbulos
3.
J Vis Exp ; (189)2022 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-36440837

RESUMO

The fidelity of oocyte meiosis is critical for generating developmentally competent euploid eggs. In mammals, the oocyte undergoes a lengthy arrest at prophase I of the first meiotic division. After puberty and upon meiotic resumption, the nuclear membrane disassembles (nuclear envelope breakdown), and the spindle is assembled mainly at the oocyte center. Initial central spindle positioning is essential to protect against abnormal kinetochore-microtubule (MT) attachments and aneuploidy. The centrally positioned spindle migrates in a time-sensitive manner toward the cortex, and this is a necessary process to extrude a tiny polar body. In mitotic cells, spindle positioning relies on the interaction between centrosome-mediated astral MTs and the cell cortex. On the contrary, mouse oocytes lack classic centrosomes and, instead, contain numerous acentriolar MT organizing centers (MTOCs). At the metaphase I stage, mouse oocytes have two different sets of MTOCs: (1) MTOCs that are clustered and sorted to assemble spindle poles (polar MTOCs), and (2) metaphase cytoplasmic MTOCs (mcMTOCs) that remain in the cytoplasm and do not contribute directly to spindle formation but play a crucial role in regulating spindle positioning and timely spindle migration. Here, a multi-photon laser ablation method is described to selectively deplete endogenously labeled mcMTOCs in oocytes collected from Cep192-eGfp reporter mice. This method contributes to the understanding of the molecular mechanisms underlying spindle positioning and migration in mammalian oocytes.


Assuntos
Terapia a Laser , Centro Organizador dos Microtúbulos , Camundongos , Animais , Centro Organizador dos Microtúbulos/metabolismo , Fuso Acromático/metabolismo , Maturidade Sexual , Oócitos , Segregação de Cromossomos , Mamíferos
4.
J Cell Biol ; 221(12)2022 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-36214847

RESUMO

Centrosomes play a crucial role during immune cell interactions and initiation of the immune response. In proliferating cells, centrosome numbers are tightly controlled and generally limited to one in G1 and two prior to mitosis. Defects in regulating centrosome numbers have been associated with cell transformation and tumorigenesis. Here, we report the emergence of extra centrosomes in leukocytes during immune activation. Upon antigen encounter, dendritic cells pass through incomplete mitosis and arrest in the subsequent G1 phase leading to tetraploid cells with accumulated centrosomes. In addition, cell stimulation increases expression of polo-like kinase 2, resulting in diploid cells with two centrosomes in G1-arrested cells. During cell migration, centrosomes tightly cluster and act as functional microtubule-organizing centers allowing for increased persistent locomotion along gradients of chemotactic cues. Moreover, dendritic cells with extra centrosomes display enhanced secretion of inflammatory cytokines and optimized T cell responses. Together, these results demonstrate a previously unappreciated role of extra centrosomes for regular cell and tissue homeostasis.


Assuntos
Centrossomo , Células Dendríticas , Pontos de Checagem do Ciclo Celular , Movimento Celular , Centrossomo/metabolismo , Quimiotaxia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Humanos , Centro Organizador dos Microtúbulos , Mitose , Proteínas Serina-Treonina Quinases/metabolismo , Linfócitos T/metabolismo
5.
J Cell Sci ; 135(21)2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36217793

RESUMO

The gene mutated in colorectal cancer (MCC) encodes a coiled-coil protein implicated, as its name suggests, in the pathogenesis of hereditary human colon cancer. To date, however, the contributions of MCC to intestinal homeostasis and disease remain unclear. Here, we examine the subcellular localization of MCC, both at the mRNA and protein levels, in the adult intestinal epithelium. Our findings reveal that Mcc transcripts are restricted to proliferating crypt cells, including Lgr5+ stem cells, where the Mcc protein is distinctly associated with the centrosome. Upon intestinal cellular differentiation, Mcc is redeployed to the apical domain of polarized villus cells where non-centrosomal microtubule organizing centers (ncMTOCs) are positioned. Using intestinal organoids, we show that the shuttling of the Mcc protein depends on phosphorylation by casein kinases 1δ and ε, which are critical modulators of WNT signaling. Together, our findings support a role for MCC in establishing and maintaining the cellular architecture of the intestinal epithelium as a component of both the centrosome and ncMTOC.


Assuntos
Centrossomo , Centro Organizador dos Microtúbulos , Humanos , Centro Organizador dos Microtúbulos/metabolismo , Centrossomo/metabolismo , Intestinos , Diferenciação Celular , Proteínas/metabolismo , Mucosa Intestinal/metabolismo
6.
Elife ; 112022 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-35758262

RESUMO

Centrosomes act as the main microtubule organizing center (MTOC) in metazoans. Centrosome number is tightly regulated by limiting centriole duplication to a single round per cell cycle. This control is achieved by multiple mechanisms, including the regulation of the protein kinase PLK4, the most upstream facilitator of centriole duplication. Altered centrosome numbers in mouse and human cells cause p53-dependent growth arrest through poorly defined mechanisms. Recent work has shown that the E3 ligase TRIM37 is required for cell cycle arrest in acentrosomal cells. To gain additional insights into this process, we undertook a series of genome-wide CRISPR/Cas9 screens to identify factors important for growth arrest triggered by treatment with centrinone B, a selective PLK4 inhibitor. We found that TRIM37 is a key mediator of growth arrest after partial or full PLK4 inhibition. Interestingly, PLK4 cellular mobility decreased in a dose-dependent manner after centrinone B treatment. In contrast to recent work, we found that growth arrest after PLK4 inhibition correlated better with PLK4 activity than with mitotic length or centrosome number. These data provide insights into the global response to changes in centrosome number and PLK4 activity and extend the role for TRIM37 in regulating the abundance, localization, and function of centrosome proteins.


Assuntos
Centríolos , Centrossomo , Proteínas Serina-Treonina Quinases , Pirimidinas , Sulfonas , Animais , Proteínas de Ciclo Celular/metabolismo , Centríolos/efeitos dos fármacos , Centríolos/metabolismo , Centrossomo/metabolismo , Camundongos , Centro Organizador dos Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Pirimidinas/farmacologia , Fuso Acromático/metabolismo , Sulfonas/farmacologia , Ubiquitina-Proteína Ligases/metabolismo
7.
Elife ; 112022 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-35758650

RESUMO

Cells encountering stressful situations activate the integrated stress response (ISR) pathway to limit protein synthesis and redirect translation to better cope. The ISR has also been implicated in cancers, but redundancies in the stress-sensing kinases that trigger the ISR have posed hurdles to dissecting physiological relevance. To overcome this challenge, we targeted the regulatory node of these kinases, namely, the S51 phosphorylation site of eukaryotic translation initiation factor eIF2α and genetically replaced eIF2α with eIF2α-S51A in mouse squamous cell carcinoma (SCC) stem cells of skin. While inconsequential under normal growth conditions, the vulnerability of this ISR-null state was unveiled when SCC stem cells experienced proteotoxic stress. Seeking mechanistic insights into the protective roles of the ISR, we combined ribosome profiling and functional approaches to identify and probe the functional importance of translational differences between ISR-competent and ISR-null SCC stem cells when exposed to proteotoxic stress. In doing so, we learned that the ISR redirects translation to centrosomal proteins that orchestrate the microtubule dynamics needed to efficiently concentrate unfolded proteins at the microtubule-organizing center so that they can be cleared by the perinuclear degradation machinery. Thus, rather than merely maintaining survival during proteotoxic stress, the ISR also functions in promoting cellular recovery once the stress has subsided. Remarkably, this molecular program is unique to transformed skin stem cells, hence exposing a vulnerability in cancer that could be exploited therapeutically.


Assuntos
Centro Organizador dos Microtúbulos , Estresse Fisiológico , Animais , Fator de Iniciação 2 em Eucariotos/metabolismo , Camundongos , Centro Organizador dos Microtúbulos/metabolismo , Fosforilação , Proteínas/metabolismo
8.
Front Immunol ; 13: 871106, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35558071

RESUMO

NK cell-mediated cytotoxicity is a critical element of our immune system required for protection from microbial infections and cancer. NK cells bind to and eliminate infected or cancerous cells via direct secretion of cytotoxic molecules toward the bound target cells. In this review, we summarize the current understanding of the molecular regulations of NK cell cytotoxicity, focusing on lytic granule development and degranulation processes. NK cells synthesize apoptosis-inducing proteins and package them into specialized organelles known as lytic granules (LGs). Upon activation of NK cells, LGs converge with the microtubule organizing center through dynein-dependent movement along microtubules, ultimately polarizing to the cytotoxic synapse where they subsequently fuse with the NK plasma membrane. From LGs biogenesis to degranulation, NK cells utilize several strategies to protect themselves from their own cytotoxic molecules. Additionally, molecular pathways that enable NK cells to perform serial killing are beginning to be elucidated. These advances in the understanding of the molecular pathways behind NK cell cytotoxicity will be important to not only improve current NK cell-based anti-cancer therapies but also to support the discovery of additional therapeutic opportunities.


Assuntos
Citotoxicidade Imunológica , Células Matadoras Naturais , Grânulos Citoplasmáticos , Imunoterapia Adotiva , Centro Organizador dos Microtúbulos
9.
J Cell Biol ; 221(7)2022 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-35604367

RESUMO

Branching microtubule (MT) nucleation is mediated by the augmin complex and γ-tubulin ring complex (γ-TuRC). However, how these two complexes work together to promote this process remains elusive. Here, using purified components from native and recombinant sources, we demonstrate that human augmin and γ-TuRC are sufficient to reconstitute the minimal MT branching machinery, in which NEDD1 bridges between augmin holo complex and GCP3/MZT1 subcomplex of γ-TuRC. The single-molecule experiment suggests that oligomerization of augmin may activate the branching machinery. We provide direct biochemical evidence that CDK1- and PLK1-dependent phosphorylation are crucial for NEDD1 binding to augmin, for their synergistic MT-binding activities, and hence for branching MT nucleation. In addition, we unveil that NEDD1 possesses an unanticipated intrinsic affinity for MTs via its WD40 domain, which also plays a pivotal role in the branching process. In summary, our study provides a comprehensive understanding of the underlying mechanisms of branching MT nucleation in human cells.


Assuntos
Proteínas Associadas aos Microtúbulos , Centro Organizador dos Microtúbulos , Microtúbulos , Tubulina (Proteína) , Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Tubulina (Proteína)/metabolismo , Repetições WD40 , Quinase 1 Polo-Like
10.
J Biol Chem ; 298(4): 101778, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35231444

RESUMO

Cytoskeletal microtubules (MTs) are nucleated from γ-tubulin ring complexes (γTuRCs) located at MT organizing centers (MTOCs), such as the centrosome. However, the exact regulatory mechanism of γTuRC assembly is not fully understood. Here, we showed that the nonreceptor tyrosine kinase c-Abl was associated with and phosphorylated γ-tubulin, the essential component of the γTuRC, mainly on the Y443 residue by in vivo (immunofluorescence and immunoprecipitation) or in vitro (surface plasmon resonance) detection. We further demonstrated that phosphorylation deficiency significantly impaired γTuRC assembly, centrosome construction, and MT nucleation. c-Abl/Arg deletion and γ-tubulin Y443F mutation resulted in an abnormal morphology and compromised spindle function during mitosis, eventually causing uneven chromosome segregation. Our findings reveal that γTuRC assembly and nucleation function are regulated by Abl kinase-mediated γ-tubulin phosphorylation, revealing a fundamental mechanism that contributes to the maintenance of MT function.


Assuntos
Centro Organizador dos Microtúbulos , Microtúbulos , Proteínas Proto-Oncogênicas c-abl , Tubulina (Proteína) , Centrossomo/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Proto-Oncogênicas c-abl/metabolismo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
11.
Science ; 375(6581): eabj3944, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35143306

RESUMO

Human oocytes are prone to assembling meiotic spindles with unstable poles, which can favor aneuploidy in human eggs. The underlying causes of spindle instability are unknown. We found that NUMA (nuclear mitotic apparatus protein)-mediated clustering of microtubule minus ends focused the spindle poles in human, bovine, and porcine oocytes and in mouse oocytes depleted of acentriolar microtubule-organizing centers (aMTOCs). However, unlike human oocytes, bovine, porcine, and aMTOC-free mouse oocytes have stable spindles. We identified the molecular motor KIFC1 (kinesin superfamily protein C1) as a spindle-stabilizing protein that is deficient in human oocytes. Depletion of KIFC1 recapitulated spindle instability in bovine and aMTOC-free mouse oocytes, and the introduction of exogenous KIFC1 rescued spindle instability in human oocytes. Thus, the deficiency of KIFC1 contributes to spindle instability in human oocytes.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cinesinas/deficiência , Oócitos/fisiologia , Oócitos/ultraestrutura , Fuso Acromático/fisiologia , Polos do Fuso/fisiologia , 1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Animais , Bovinos , Complexo Dinactina/metabolismo , Dineínas/metabolismo , Feminino , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Centro Organizador dos Microtúbulos/fisiologia , Centro Organizador dos Microtúbulos/ultraestrutura , Microtúbulos/metabolismo , Proteínas Recombinantes/metabolismo , Fuso Acromático/ultraestrutura , Polos do Fuso/ultraestrutura , Suínos
12.
Biophys J ; 121(7): 1246-1265, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35196513

RESUMO

Cytotoxic T lymphocytes (T cells) and natural killer cells form a tight contact, the immunological synapse (IS), with target cells, where they release their lytic granules containing perforin/granzyme and cytokine-containing vesicles. During this process the cell repolarizes and moves the microtubule organizing center (MTOC) toward the IS. In the first part of our work we developed a computational model for the molecular-motor-driven motion of the microtubule cytoskeleton during T cell polarization and analyzed the effects of cortical-sliding and capture-shrinkage mechanisms. Here we use this model to analyze the dynamics of the MTOC repositioning in situations in which 1) the IS is in an arbitrary position with respect to the initial position of the MTOC and 2) the T cell has two IS at two arbitrary positions. In the case of one IS, we found that the initial position determines which mechanism is dominant and that the time of repositioning does not rise monotonously with the MTOC-IS distance. In the case of two IS, we observe several scenarios that have also been reported experimentally: the MTOC alternates stochastically (but with a well-defined average transition time) between the two IS; it wiggles in between the two IS without transiting to one of the two; or it is at some point pulled to one of the two IS and stays there. Our model allows one to predict which scenario emerges in dependency of the mechanisms in action and the number of dyneins present. We report that the presence of capture-shrinkage mechanism in at least one IS is necessary to assure the transitions in every cell configuration. Moreover, the frequency of transitions does not decrease with the distance between the two IS and is the highest when both mechanisms are present in both IS.


Assuntos
Sinapses Imunológicas , Centro Organizador dos Microtúbulos , Dineínas/metabolismo , Sinapses Imunológicas/metabolismo , Células Matadoras Naturais , Ativação Linfocitária , Centro Organizador dos Microtúbulos/metabolismo
14.
J Cell Biol ; 220(12)2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34779859

RESUMO

The γ-tubulin complex acts as the predominant microtubule (MT) nucleator that initiates MT formation and is therefore an essential factor for cell proliferation. Nonetheless, cellular MTs are formed after experimental depletion of the γ-tubulin complex, suggesting that cells possess other factors that drive MT nucleation. Here, by combining gene knockout, auxin-inducible degron, RNA interference, MT depolymerization/regrowth assay, and live microscopy, we identified four microtubule-associated proteins (MAPs), ch-TOG, CLASP1, CAMSAPs, and TPX2, which are involved in γ-tubulin-independent MT generation in human colon cancer cells. In the mitotic MT regrowth assay, nucleated MTs organized noncentriolar MT organizing centers (ncMTOCs) in the absence of γ-tubulin. Depletion of CLASP1 or TPX2 substantially delayed ncMTOC formation, suggesting that these proteins might promote MT nucleation in the absence of γ-tubulin. In contrast, depletion of ch-TOG or CAMSAPs did not affect the timing of ncMTOC appearance. CLASP1 also accelerates γ-tubulin-independent MT regrowth during interphase. Thus, MT generation can be promoted by MAPs without the γ-tubulin template.


Assuntos
Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Aurora Quinases/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Humanos , Interfase , Metáfase , Centro Organizador dos Microtúbulos/metabolismo , Mitose , Polimerização , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas , Fuso Acromático/metabolismo , Quinase 1 Polo-Like
15.
Science ; 374(6569): 874-879, 2021 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-34762476

RESUMO

In mammals and flies, only one cell in a multicellular female germline cyst becomes an oocyte, but how symmetry is broken to select the oocyte is unknown. Here, we show that the microtubule (MT) minus end-stabilizing protein Patronin/CAMSAP marks the future Drosophila oocyte and is required for oocyte specification. The spectraplakin Shot recruits Patronin to the fusome, a branched structure extending into all cyst cells. Patronin stabilizes more MTs in the cell with the most fusome material. Our data suggest that this weak asymmetry is amplified by Dynein-dependent transport of Patronin-stabilized MTs. This forms a polarized MT network, along which Dynein transports oocyte determinants into the presumptive oocyte. Thus, Patronin amplifies a weak fusome anisotropy to break symmetry and select one cell to become the oocyte.


Assuntos
Proteínas de Drosophila/metabolismo , Células Germinativas/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Oócitos/fisiologia , Animais , Anisotropia , Drosophila melanogaster , Dineínas/metabolismo , Feminino , Células Germinativas/ultraestrutura , Proteínas dos Microfilamentos/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Oócitos/ultraestrutura , Organelas/metabolismo , Organelas/ultraestrutura
16.
Cell Cycle ; 20(23): 2443-2451, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34672905

RESUMO

Loss of function mutations in the E3 ubiquitin ligase TRIM37 result in MULIBREY nanism, a disease characterized by impaired organ growth and a high propensity to develop different tumor types. Additionally, increased copy number of TRIM37 is a feature of some breast cancers and neuroblastomas. The molecular role played by TRIM37 in such loss and gain of function conditions has been a focus of research in the last decade, which led notably to the identification of critical roles of TRIM37 in centrosome biology. Specifically, deletion of TRIM37 results in the formation of aberrant centrosomal proteins assemblies, including Centrobin-PLK4 assemblies, which can act as extra MTOCs, thus resulting in defective chromosome segregation. Additionally, TRIM37 overexpression targets the centrosomal protein CEP192 for degradation, thereby preventing centrosome maturation and increasing the frequency of mitotic errors. Interestingly, increased TRIM37 protein levels sensitize cells to the PLK4 inhibitor centrinone. In this review, we cover the emerging roles of TRIM37 in centrosome biology and discuss how this knowledge may lead to new therapeutic strategies to target specific cancer cells.


Assuntos
Nanismo de Mulibrey , Ubiquitina-Proteína Ligases , Centrossomo/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Humanos , Centro Organizador dos Microtúbulos/metabolismo , Nanismo de Mulibrey/genética , Nanismo de Mulibrey/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
17.
Elife ; 102021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-34137371

RESUMO

Sustained polarity and adhesion of epithelial cells is essential for the protection of our organs and bodies, and this epithelial integrity emerges during organ development amidst numerous programmed morphogenetic assaults. Using the developing Caenorhabditis elegans intestine as an in vivo model, we investigated how epithelia maintain their integrity through cell division and elongation to build a functional tube. Live imaging revealed that apical PAR complex proteins PAR-6/Par6 and PKC-3/aPkc remained apical during mitosis while apical microtubules and microtubule-organizing center (MTOC) proteins were transiently removed. Intestine-specific depletion of PAR-6, PKC-3, and the aPkc regulator CDC-42/Cdc42 caused persistent gaps in the apical MTOC as well as in other apical and junctional proteins after cell division and in non-dividing cells that elongated. Upon hatching, gaps coincided with luminal constrictions that blocked food, and larvae arrested and died. Thus, the apical PAR complex maintains apical and junctional continuity to construct a functional intestinal tube.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Células Epiteliais , Mucosa Intestinal , Animais , Caenorhabditis elegans , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiologia , Intestinos/citologia , Intestinos/crescimento & desenvolvimento , Larva/crescimento & desenvolvimento , Centro Organizador dos Microtúbulos/metabolismo , Proteína Quinase C/metabolismo
18.
J Cell Sci ; 134(14)2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34152366

RESUMO

Oocyte-specific knockdown of pericentrin (PCNT) in transgenic (Tg) mice disrupts acentriolar microtubule-organizing center (aMTOC) formation, leading to spindle instability and error-prone meiotic division. Here, we show that PCNT-depleted oocytes lack phosphorylated Aurora A (pAURKA) at spindle poles, while overall levels are unaltered. To test aMTOC-associated AURKA function, metaphase II (MII) control (WT) and Tg oocytes were briefly exposed to a specific AURKA inhibitor (MLN8237). Similar defects were observed in Tg and MLN8237-treated WT oocytes, including altered spindle structure, increased chromosome misalignment and impaired microtubule regrowth. Yet, AURKA inhibition had a limited effect on Tg oocytes, revealing a critical role for aMTOC-associated AURKA in regulating spindle stability. Notably, spindle instability was associated with disrupted γ-tubulin and lack of the liquid-like meiotic spindle domain (LISD) in Tg oocytes. Analysis of this Tg model provides the first evidence that LISD assembly depends expressly on aMTOC-associated AURKA, and that Ran-mediated spindle formation ensues without the LISD. These data support that loss of aMTOC-associated AURKA and failure of LISD assembly contribute to error-prone meiotic division in PCNT-depleted oocytes, underscoring the essential role of aMTOCs for spindle stability.


Assuntos
Aurora Quinase A , Centro Organizador dos Microtúbulos , Fuso Acromático , Animais , Aurora Quinase A/genética , Meiose , Camundongos , Oócitos , Fuso Acromático/genética , Polos do Fuso/genética
19.
Curr Biol ; 31(10): R506-R511, 2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-34033781

RESUMO

Encircling and traversing the cell are architectural struts and dynamic intracellular highways made of cylindrical polymers called microtubules. Built from structurally asymmetric subunits of αß-tubulin heterodimers, microtubules have an inherent structural polarity with a slow-growing minus end and a comparatively dynamic plus end that grows and shrinks. Thus, a key feature of microtubules is that each polymer is polarized, allowing for the execution of cellular tasks that are directional in nature. For example, microtubules build polarized highways allowing directional intracellular transport, generate directional force such as in chromosome alignment and segregation, provide structural support for cell shape, and assemble into highly ordered polar structures like centrioles and cilia. The output of these microtubule-based functions is the performance of different tasks, including establishment and maintenance of cellular polarity, secretion and absorption, cell-cell communication, migration, mechanical resiliency, and mitosis. Different cells accomplish these functions by using distinct sites within the cell called microtubule-organizing centers (MTOCs) to build cell-specific microtubule arrangements. While the specific requirement for microtubules in many in vivo cell types is unknown, disrupting even a subset of microtubule-supported functions is often lethal and is associated with many diseases (e.g., cancer and neuropathies), suggesting that specific patterns of microtubule organization are likely important for cellular function in vivo. This Primer focuses on how differentiated animal and plant cells use distinct MTOCs to generate specific microtubule arrangements, how those arrangements support cellular functions, and how cells rearrange their microtubules to accommodate changing cellular tasks.


Assuntos
Centro Organizador dos Microtúbulos , Tubulina (Proteína) , Animais , Centríolos , Microtúbulos , Mitose
20.
PLoS Genet ; 17(4): e1009327, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33901174

RESUMO

The Aurora protein kinases are well-established regulators of spindle building and chromosome segregation in mitotic and meiotic cells. In mouse oocytes, there is significant Aurora kinase A (AURKA) compensatory abilities when the other Aurora kinase homologs are deleted. Whether the other homologs, AURKB or AURKC can compensate for loss of AURKA is not known. Using a conditional mouse oocyte knockout model, we demonstrate that this compensation is not reciprocal because female oocyte-specific knockout mice are sterile, and their oocytes fail to complete meiosis I. In determining AURKA-specific functions, we demonstrate that its first meiotic requirement is to activate Polo-like kinase 1 at acentriolar microtubule organizing centers (aMTOCs; meiotic spindle poles). This activation induces fragmentation of the aMTOCs, a step essential for building a bipolar spindle. We also show that AURKA is required for regulating localization of TACC3, another protein required for spindle building. We conclude that AURKA has multiple functions essential to completing MI that are distinct from AURKB and AURKC.


Assuntos
Aurora Quinase A/genética , Proteínas de Ciclo Celular/genética , Proteínas Fetais/genética , Meiose/genética , Proteínas Associadas aos Microtúbulos/genética , Oócitos/crescimento & desenvolvimento , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Animais , Aurora Quinase B/genética , Aurora Quinase C/genética , Divisão do Núcleo Celular/genética , Segregação de Cromossomos/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Camundongos , Centro Organizador dos Microtúbulos/metabolismo , Oócitos/metabolismo , Fuso Acromático/genética , Polos do Fuso/genética , Quinase 1 Polo-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA