Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros











Intervalo de año de publicación
1.
Int J Mol Sci ; 22(19)2021 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-34638607

RESUMEN

Asymmetric cell division (ACD) of neural stem cells and progenitors not only renews the stem cell population but also ensures the normal development of the nervous system, producing various types of neurons with different shapes and functions in the brain. One major mechanism to achieve ACD is the asymmetric localization and uneven segregation of intracellular proteins and organelles into sibling cells. Recent studies have demonstrated that liquid-liquid phase separation (LLPS) provides a potential mechanism for the formation of membrane-less biomolecular condensates that are asymmetrically distributed on limited membrane regions. Moreover, mechanical forces have emerged as pivotal regulators of asymmetric neural stem cell division by generating sibling cell size asymmetry. In this review, we will summarize recent discoveries of ACD mechanisms driven by LLPS and mechanical forces.


Asunto(s)
División Celular Asimétrica/fisiología , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Animales , Fenómenos Biomecánicos , División Celular/fisiología , Polaridad Celular/fisiología , Tamaño de la Célula , Proteínas de Drosophila/fisiología , Drosophila melanogaster/citología , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/fisiología , Modelos Neurológicos , Miosinas/fisiología , Neurogénesis/fisiología , Orgánulos/fisiología
2.
Development ; 148(18)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34370012

RESUMEN

Drosophila female germline stem cells (GSCs) are found inside the cellular niche at the tip of the ovary. They undergo asymmetric divisions to renew the stem cell lineage and to produce sibling cystoblasts that will in turn enter differentiation. GSCs and cystoblasts contain spectrosomes, membranous structures essential for orientation of the mitotic spindle and that, particularly in GSCs, change shape depending on the cell cycle phase. Using live imaging and a fusion protein of GFP and the spectrosome component Par-1, we follow the complete spectrosome cycle throughout GSC division and quantify the relative duration of the different spectrosome shapes. We also determine that the Par-1 kinase shuttles between the spectrosome and the cytoplasm during mitosis and observe the continuous addition of new material to the GSC and cystoblast spectrosomes. Next, we use the Fly-FUCCI tool to define, in live and fixed tissues, that GSCs have a shorter G1 compared with the G2 phase. The observation of centrosomes in dividing GSCs allowed us to determine that centrosomes separate very early in G1, before centriole duplication. Furthermore, we show that the anterior centrosome associates with the spectrosome only during mitosis and that, upon mitotic spindle assembly, it translocates to the cell cortex, where it remains anchored until centrosome separation. Finally, we demonstrate that the asymmetric division of GSCs is not an intrinsic property of these cells, as the spectrosome of GSC-like cells located outside of the niche can divide symmetrically. Thus, GSCs display unique properties during division, a behaviour influenced by the surrounding niche.


Asunto(s)
División Celular Asimétrica/fisiología , Centrosoma/fisiología , Drosophila/fisiología , Células Germinativas/fisiología , Ovario/fisiología , Células Madre/fisiología , Animales , Diferenciación Celular/fisiología , Centrosoma/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Femenino , Fase G1/fisiología , Fase G2/fisiología , Células Germinativas/metabolismo , Mitosis/fisiología , Ovario/metabolismo , Huso Acromático/fisiología , Células Madre/metabolismo
3.
Int J Mol Sci ; 21(21)2020 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-33153113

RESUMEN

Hematopoietic stem cells (HSCs) are responsible for life-long production of all mature blood cells. Under homeostasis, HSCs in their native bone marrow niches are believed to undergo asymmetric cell divisions (ACDs), with one daughter cell maintaining HSC identity and the other committing to differentiate into various mature blood cell types. Due to the lack of key niche signals, in vitro HSCs differentiate rapidly, making it challenging to capture and study ACD. To overcome this bottleneck, in this study, we used interferon alpha (IFNα) treatment to "pre-instruct" HSC fate directly in their native niche, and then systematically studied the fate of dividing HSCs in vitro at the single cell level via time-lapse analysis, as well as multigene and protein expression analysis. Triggering HSCs' exit from dormancy via IFNα was found to significantly increase the frequency of asynchronous divisions in paired daughter cells (PDCs). Using single-cell gene expression analyses, we identified 12 asymmetrically expressed genes in PDCs. Subsequent immunocytochemistry analysis showed that at least three of the candidates, i.e., Glut1, JAM3 and HK2, were asymmetrically distributed in PDCs. Functional validation of these observations by colony formation assays highlighted the implication of asymmetric distribution of these markers as hallmarks of HSCs, for example, to reliably discriminate committed and self-renewing daughter cells in dividing HSCs. Our data provided evidence for the importance of in vivo instructions in guiding HSC fate, especially ACD, and shed light on putative molecular players involved in this process. Understanding the mechanisms of cell fate decision making should enable the development of improved HSC expansion protocols for therapeutic applications.


Asunto(s)
División Celular Asimétrica/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/fisiología , Interferón-alfa/farmacología , Animales , División Celular Asimétrica/genética , División Celular Asimétrica/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Interferón-alfa/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis de la Célula Individual
4.
Ann N Y Acad Sci ; 1466(1): 73-82, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31814150

RESUMEN

The asymmetric inheritance of NUMB during mitosis determines future daughter cell fates in multiple model organisms. NUMB asymmetric inheritance has also been postulated for hematopoietic stem cell (HSC) divisions but remained controversial until recently. To reconcile conflicting reports, we revisited the evidence for asymmetric inheritance of NUMB during HSC divisions. We demonstrate that previously used strategies to identify dividing cells in fixed samples suffer from multiple systematic errors. Nonmitotic cells in close proximity are frequently mistaken as dividing cells, while mitotic cells are not detected. Furthermore, microtubule depolymerization by either nocodazole or low temperatures prevents the reliable detection of mitosis and introduces mitotic artifacts. Without artificial microtubule depolymerization and by the use of reliable mitotic markers, we find NUMB differences in daughter cells to be reduced and restricted to cells with low NUMB expression and thus low signal over background. This bias fits the expected random distribution of simulated noise data, suggesting that the putative asymmetric inheritance of NUMB in HSCs could be merely technical noise. We conclude that functionally relevant asymmetric inheritance of NUMB and other factors in mitotic HSCs and other cells cannot be conclusively demonstrated using snapshot data and requires alternative approaches, such as continuous quantitative single-cell analysis.


Asunto(s)
División Celular Asimétrica/fisiología , Diferenciación Celular , División Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , División Celular Asimétrica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , División Celular/efectos de los fármacos , Células Cultivadas , Células Madre Hematopoyéticas/efectos de los fármacos , Patrón de Herencia/efectos de los fármacos , Patrón de Herencia/fisiología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Mitosis/efectos de los fármacos , Mitosis/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Nocodazol/farmacología , Polimerizacion/efectos de los fármacos , Distribución Tisular , Moduladores de Tubulina/farmacología
5.
Cell Cycle ; 18(20): 2590-2597, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31475621

RESUMEN

The orientation of the mitotic spindle is a crucial process during development and adult tissue homeostasis and multiple mechanisms have been shown to intrinsically regulate this process. However, much less is known about the extrinsic cues involved in modulating spindle orientation. We have recently uncovered a novel function of Eph intercellular signaling in regulating spindle alignment by ultimately ensuring the correct cortical distribution of central components within the intrinsic spindle orientation machinery. Here, we comment on these results, novel questions that they open and potential additional research to address in the future.


Asunto(s)
Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Receptores de la Familia Eph/metabolismo , Transducción de Señal/fisiología , Huso Acromático/metabolismo , Animales , División Celular Asimétrica/fisiología , Carcinogénesis/metabolismo , Polaridad Celular/fisiología , Efrinas/metabolismo , Células Neuroepiteliales/metabolismo , Lóbulo Óptico de Animales no Mamíferos/citología
6.
Life Sci ; 231: 116520, 2019 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-31158379

RESUMEN

Cancer stem cells (CSCs) are self-renewal population localized within cancer niches and play critical roles in tumor initiation, recurrence and metastasis. Despite extensive research, challenges about identity of CSCs and combating them in cancer therapy still remain steady. Cellular plasticity is a cardinal feature of tumor microenvironment (TME) tremendously influencing tumor aggressive behavior. Plasticity and CSC a (symmetry) are interconnecting processes essential for shaping a cancer through nurturing a wide number of cells with tumor promoting capacities. The plastic nature of TME cellularity infers that destemming just CSCs is not sufficient in respect with therapy, especially for high-grade cancers-instead, deploying mechanisms to retard tumor type-dependent TME-CSC interplay is a suggested strategy for making a durable remission of cancer. This requires extending our understanding about CSC divisional profiling and plasticity in order to find critical drivers in cancer progression.


Asunto(s)
Plasticidad de la Célula/fisiología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , División Celular Asimétrica/fisiología , Carcinogénesis/metabolismo , Transformación Celular Neoplásica/patología , Transición Epitelial-Mesenquimal/fisiología , Humanos , Recurrencia Local de Neoplasia/patología , Transducción de Señal , Microambiente Tumoral/fisiología
7.
Open Biol ; 9(2): 180243, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30958098

RESUMEN

Precise specification of cell fate or identity within stem cell lineages is critical for ensuring correct stem cell lineage progression and tissue homeostasis. Failure to specify cell fate or identity in a timely and robust manner can result in developmental abnormalities and diseases such as cancer. However, the molecular basis of timely cell fate/identity specification is only beginning to be understood. In this review, we discuss key regulatory strategies employed in cell fate specification and highlight recent results revealing how timely and robust cell fate/identity commitment is achieved through transcriptional control.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Proliferación Celular/fisiología , Modelos Biológicos , Células Madre/citología , Animales , División Celular Asimétrica/genética , División Celular Asimétrica/fisiología , Diferenciación Celular/genética , Linaje de la Célula/genética , Proliferación Celular/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica , Receptores Notch/genética , Receptores Notch/metabolismo , Células Madre/metabolismo
8.
Biol Rev Camb Philos Soc ; 94(3): 810-827, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30467934

RESUMEN

Stem cells are a promising cell source for regenerative medicine due to their characteristics of self-renewal and differentiation. The intricate balance between these two cell fates is maintained by precisely controlled symmetric and asymmetric cell divisions. Asymmetric division has a fundamental importance in maintaining tissue homeostasis and in the development of multi-cellular organisms. For example, during development, asymmetric cell divisions are responsible for the formation of the body axis. Mechanistically, mitotic spindle dynamics determine the assembly and separation of chromosomes and regulate the orientation of cell division. Interestingly, symmetric and asymmetric cell division is not mutually exclusive and a range of factors are involved in such cell-fate decisions, the measurement of which can provide efficient and reliable information on the regenerative potential of a cell. The balance between self-renewal and differentiation in stem cells is controlled by various biophysical and biochemical cues. Although the role of biochemical factors in asymmetric stem cell division has been widely studied, the effect of biophysical cues in stem-cell self-renewal is not comprehensively understood. Herein, we review the biological relevance of stem-cell asymmetric division to regenerative medicine and discuss the influences of various intrinsic and extrinsic biophysical cues in stem-cell self-renewal. This review particularly aims to inform the clinical translation of efforts to control the self-renewal ability of stem cells through the tuning of various biophysical cues.


Asunto(s)
División Celular Asimétrica/fisiología , Células Madre/fisiología , Animales , Carcinogénesis , Desarrollo Embrionario , Humanos , Cicatrización de Heridas
9.
Front Biosci (Landmark Ed) ; 24(3): 451-462, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30468666

RESUMEN

Cell polarity, the asymmetric distribution of proteins, organelles, and cytoskeleton, plays an important role in development, homeostasis, and disease. Understanding the mechanisms that govern cell polarity is critical for creating strategies to treat developmental defects, accelerate tissue regeneration, and hinder cancer progression. This review focuses on the role of cell polarity in a number of physiological processes, including asymmetric division, cell migration, immune response mediated by T lymphocytes, and cancer progression and metastasis, and highlights microfabrication techniques to systematically parse the role of microenvironmental cues in the regulation of cell polarity.


Asunto(s)
Movimiento Celular/fisiología , Polaridad Celular/fisiología , Citoesqueleto/fisiología , Transducción de Señal/fisiología , Animales , División Celular Asimétrica/fisiología , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/fisiología , Neoplasias/fisiopatología
10.
J Cell Biol ; 218(3): 771-782, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30563876

RESUMEN

Asymmetric divisions are essential for the generation of cell fate and size diversity. They implicate cortical domains where minus end-directed motors, such as dynein, are activated to pull on microtubules to decenter asters attached to centrosomes, nuclei, or spindles. In asymmetrically dividing cells, aster decentration typically follows a centering phase, suggesting a time-dependent regulation in the competition between microtubule centering and decentering forces. Using symmetrically dividing sea urchin zygotes, we generated cortical domains of magnetic particles that spontaneously cluster endogenous dynein activity. These domains efficiently attract asters and nuclei, yielding marked asymmetric divisions. Remarkably, aster decentration only occurred after asters had first reached the cell center. Using intracellular force measurement and models, we demonstrate that this time-regulated imbalance results from a global reduction of centering forces rather than a local maturation of dynein activity at the domain. Those findings define a novel paradigm for the regulation of division asymmetry.


Asunto(s)
División Celular Asimétrica/fisiología , Centrosoma/metabolismo , Microtúbulos/metabolismo , Modelos Biológicos , Paracentrotus/metabolismo , Animales , Dineínas/metabolismo
11.
EMBO J ; 37(16)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29997178

RESUMEN

Stem cells establish cortical polarity and divide asymmetrically to simultaneously maintain themselves and generate differentiating offspring cells. Several chromatin modifiers have been identified as stemness factors in mammalian pluripotent stem cells, but whether these factors control stem cell polarity and asymmetric division has not been investigated so far. We addressed this question in Drosophila neural stem cells called neuroblasts. We identified the Tip60 chromatin remodeling complex and its interaction partner Myc as regulators of genes required for neuroblast maintenance. Knockdown of Tip60 complex members results in loss of cortical polarity, symmetric neuroblast division, and premature differentiation through nuclear entry of the transcription factor Prospero. We found that aPKC is the key target gene of Myc and the Tip60 complex subunit Domino in regulating neuroblast polarity. Our transcriptome analysis further showed that Domino regulates the expression of mitotic spindle genes previously identified as direct Myc targets. Our findings reveal an evolutionarily conserved functional link between Myc, the Tip60 complex, and the molecular network controlling cell polarity and asymmetric cell division.


Asunto(s)
División Celular Asimétrica/fisiología , Polaridad Celular/fisiología , Proteínas de Drosophila/metabolismo , Histona Acetiltransferasas/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Histona Acetiltransferasas/genética , Células-Madre Neurales/citología , Proteínas Proto-Oncogénicas c-myc/genética , Huso Acromático/genética , Huso Acromático/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Development ; 145(11)2018 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-29724757

RESUMEN

Asymmetric division is crucial for embryonic development and stem cell lineages. In the one-cell Caenorhabditis elegans embryo, a contractile cortical actomyosin network contributes to asymmetric division by segregating partitioning-defective (PAR) proteins to discrete cortical domains. In the current study, we found that the plasma membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP2) localizes to polarized dynamic structures in C. elegans zygotes, distributing in a PAR-dependent manner along the anterior-posterior (A-P) embryonic axis. PIP2 cortical structures overlap with F-actin, and coincide with the actin regulators RHO-1 and CDC-42, as well as ECT-2. Particle image velocimetry analysis revealed that PIP2 and F-actin cortical movements are coupled, with PIP2 structures moving slightly ahead of F-actin. Importantly, we established that PIP2 cortical structure formation and movement is actin dependent. Moreover, we found that decreasing or increasing the level of PIP2 resulted in severe F-actin disorganization, revealing interdependence between these components. Furthermore, we determined that PIP2 and F-actin regulate the sizing of PAR cortical domains, including during the maintenance phase of polarization. Overall, our work establishes that a lipid membrane component, PIP2, modulates actin organization and cell polarity in C. elegans embryos.


Asunto(s)
Actinas/metabolismo , División Celular Asimétrica/fisiología , Caenorhabditis elegans/embriología , Polaridad Celular/fisiología , Lípidos de la Membrana/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Animales , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/metabolismo , Embrión no Mamífero/metabolismo , Proteínas de Unión al GTP/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas de Unión al GTP rho/metabolismo
13.
Exp Hematol ; 64: 1-11, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29807063

RESUMEN

Hematopoietic stem cells maintain a quiescent state in the bone marrow to preserve their self-renewal capacity, but also undergo cell divisions as required. Organelles such as the mitochondria sustain cumulative damage during these cell divisions and this damage may eventually compromise the cells' self-renewal capacity. Hematopoietic stem cell divisions result in either self-renewal or differentiation, with the balance between the two affecting hematopoietic homeostasis directly; however, the heterogeneity of available hematopoietic stem cell-enriched fractions, together with the technical challenges of observing hematopoietic stem cell behavior, has long hindered the analysis of individual hematopoietic stem cells and prevented the elucidation of this process. Recent advances in genetic models, metabolomics analyses, and single-cell approaches have revealed the contributions made to hematopoietic stem cell self-renewal by metabolic cues, mitochondrial biogenesis, and autophagy/mitophagy, which have highlighted mitochondrial quality control as a key factor in the equilibrium of hematopoietic stem cells. A deeper understanding of precisely how specific modes of metabolism control hematopoietic stem cells fate at the single-cell level is therefore not only of great biological interest, but will also have clear clinical implications for the development of therapies for hematological diseases.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Animales , División Celular Asimétrica/fisiología , Biomarcadores , Diferenciación Celular/fisiología , Autorrenovación de las Células/fisiología , Células Cultivadas , Microambiente Celular , Metabolismo Energético , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Humanos , Mitocondrias/metabolismo , Mitofagia , Proyectos de Investigación
14.
Results Probl Cell Differ ; 61: 243-260, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28409308

RESUMEN

Asymmetric cell divisions produce two daughter cells with distinct fate. During embryogenesis, this mechanism is fundamental to build tissues and organs because it generates cell diversity. In adults, it remains crucial to maintain stem cells. The enthusiasm for asymmetric cell division is not only motivated by the beauty of the mechanism and the fundamental questions it raises, but has also very pragmatic reasons. Indeed, misregulation of asymmetric cell divisions is believed to have dramatic consequences potentially leading to pathogenesis such as cancers. In diverse model organisms, asymmetric cell divisions result in two daughter cells, which differ not only by their fate but also in size. This is the case for the early Xenopus laevis embryo, in which the two first embryonic divisions are perpendicular to each other and generate two pairs of blastomeres, which usually differ in size: one pair of blastomeres is smaller than the other. Small blastomeres will produce embryonic dorsal structures, whereas the larger pair will evolve into ventral structures. Here, we present a speculative model on the origin of the asymmetry of this cell division in the Xenopus embryo. We also discuss the apparently coincident asymmetric distribution of cell fate determinants and cell-size asymmetry of the 4-cell stage embryo. Finally, we discuss the asymmetric furrowing during epithelial cell cytokinesis occurring later during Xenopus laevis embryo development.


Asunto(s)
División Celular Asimétrica/fisiología , Blastómeros/citología , Modelos Biológicos , Xenopus laevis/embriología , Animales , Tamaño de la Célula
15.
Results Probl Cell Differ ; 61: 183-210, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28409305

RESUMEN

Asymmetric cell division (ACD) is a fundamental mechanism to generate cell diversity, giving rise to daughter cells with different developmental potentials. ACD is manifested in the asymmetric segregation of proteins or mRNAs, when the two daughter cells differ in size or are endowed with different potentials to differentiate into a particular cell type (Horvitz and Herskowitz, Cell 68:237-255, 1992). Drosophila neuroblasts, the neural stem cells of the developing fly brain, are an ideal system to study ACD since this system encompasses all of these characteristics. Neuroblasts are intrinsically polarized cells, utilizing polarity cues to orient the mitotic spindle, segregate cell fate determinants asymmetrically, and regulate spindle geometry and physical asymmetry. The neuroblast system has contributed significantly to the elucidation of the basic molecular mechanisms underlying ACD. Recent findings also highlight its usefulness to study basic aspects of stem cell biology and tumor formation. In this review, we will focus on what has been learned about the basic mechanisms underlying ACD in fly neuroblasts.


Asunto(s)
División Celular Asimétrica/fisiología , Drosophila melanogaster/fisiología , Células-Madre Neurales/citología , Animales , Polaridad Celular/fisiología , Proteínas de Drosophila/metabolismo , Modelos Animales
16.
Results Probl Cell Differ ; 61: 351-373, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28409313

RESUMEN

Stem cells use mode of cell division, symmetric (SCD) versus asymmetric (ACD), to balance expansion with self-renewal and the generation of daughter cells with different cell fates. Studies in model organisms have identified intrinsic mechanisms that govern this process, which involves partitioning molecular components between daughter cells, frequently through the regulation of the mitotic spindle. Research performed in vertebrate tissues is revealing both conservation of these intrinsic mechanisms and crucial roles for extrinsic cues in regulating the frequency of these divisions. Morphogens and positional cues, including planar cell polarity proteins and guidance molecules, regulate key signaling pathways required to organize cell/ECM contacts and spindle pole dynamics. Noncanonical WNT7A/VANGL2 signaling governs asymmetric cell division and the acquisition of cell fates through spindle pole orientation in satellite stem cells of regenerating muscle fibers. During cortical neurogenesis, the same pathway regulates glial cell fate determination by regulating spindle size, independent of its orientation. Sonic hedgehog (SHH) stimulates the symmetric expansion of cortical stem and cerebellar progenitor cells and contributes to cell fate acquisition in collaboration with Notch and Wnt signaling pathways. SLIT2 also contributes to stem cell homeostasis by restricting ACD frequency through the regulation of spindle orientation. The capacity to influence stem cells makes these secreted factors excellent targets for therapeutic strategies designed to enhance cell populations in degenerative disease or restrict cell proliferation in different types of cancers.


Asunto(s)
División Celular Asimétrica/fisiología , Transducción de Señal/fisiología , Huso Acromático/fisiología , Células Madre/citología , Animales , Humanos , Neurogénesis/fisiología
17.
Results Probl Cell Differ ; 61: 401-421, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28409315

RESUMEN

Asymmetric division of stem cells is a highly conserved and tightly regulated process by which a single stem cell produces two unequal daughter cells. One retains its stem cell identity while the other becomes specialized through a differentiation program and loses stem cell properties. Coordinating these events requires control over numerous intra- and extracellular biological processes and signaling networks. In the initial stages, critical events include the compartmentalization of fate determining proteins within the mother cell and their subsequent passage to the appropriate daughter cell in order to direct their destiny. Disturbance of these events results in an altered dynamic of self-renewing and differentiation within the cell population, which is highly relevant to the growth and progression of cancer. Other critical events include proper asymmetric spindle assembly, extrinsic regulation through micro-environmental cues, and non-canonical signaling networks that impact cell division and fate determination. In this review, we discuss mechanisms that maintain the delicate balance of asymmetric cell division in normal tissues and describe the current understanding how some of these mechanisms are deregulated in cancer.


Asunto(s)
División Celular Asimétrica/fisiología , Células Madre Neoplásicas/citología , Animales , Humanos , Transducción de Señal/fisiología , Células Madre/citología
18.
Results Probl Cell Differ ; 61: 375-399, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28409314

RESUMEN

Stem and progenitor cells are characterized by their abilities to self-renew and produce differentiated progeny. The balance between self-renewal and differentiation is achieved through control of cell division mode, which can be either asymmetric or symmetric. Failure to properly control cell division mode may result in premature depletion of the stem/progenitor cell pool or abnormal growth and impaired differentiation. In many tissues, including the brain, stem cells and progenitor cells undergo asymmetric cell division through the establishment of cell polarity. Cell polarity proteins are therefore potentially critical regulators of asymmetric cell division. Decrease or loss of asymmetric cell division can be associated with reduced differentiation common during aging or impaired remyelination as seen in demyelinating diseases. Progenitor-like glioma precursor cells show decreased asymmetric cell division rates and increased symmetric divisions, which suggests that asymmetric cell division suppresses brain tumor formation. Cancer stem cells, on the other hand, still undergo low rates of asymmetric cell division, which may provide them with a survival advantage during therapy. These findings led to the hypotheses that asymmetric cell divisions are not always tumor suppressive but can also be utilized to maintain a cancer stem cell population. Proper control of cell division mode is therefore not only deemed necessary to generate cellular diversity during development and to maintain adult tissue homeostasis but may also prevent disease and determine disease progression. Since brain cancer is most common in the adult and aging population, we review here the current knowledge on molecular mechanisms that regulate asymmetric cell divisions in the neural and oligodendroglial lineage during development and in the adult brain.


Asunto(s)
División Celular Asimétrica/fisiología , Células Madre Neoplásicas/citología , Células-Madre Neurales/citología , Animales , Humanos
19.
Proc Natl Acad Sci U S A ; 112(46): E6321-30, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26578792

RESUMEN

Centrosomes are microtubule-organizing centers that facilitate bipolar mitotic spindle assembly and chromosome segregation. Recognizing that centrosome amplification is a common feature of aneuploid cancer cells, we tested whether supernumerary centrosomes are sufficient to drive tumor development. To do this, we constructed and analyzed mice in which centrosome amplification can be induced by a Cre-recombinase-mediated increase in expression of Polo-like kinase 4 (Plk4). Elevated Plk4 in mouse fibroblasts produced supernumerary centrosomes and enhanced the expected mitotic errors, but proliferation continued only after inactivation of the p53 tumor suppressor. Increasing Plk4 levels in mice with functional p53 produced centrosome amplification in liver and skin, but this did not promote spontaneous tumor development in these tissues or enhance the growth of chemically induced skin tumors. In the absence of p53, Plk4 overexpression generated widespread centrosome amplification, but did not drive additional tumors or affect development of the fatal thymic lymphomas that arise in animals lacking p53. We conclude that, independent of p53 status, supernumerary centrosomes are not sufficient to drive tumor formation.


Asunto(s)
División Celular Asimétrica/fisiología , Centrosoma/metabolismo , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Regulación de la Expresión Génica , Linfoma/genética , Linfoma/metabolismo , Linfoma/patología , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Neoplasias del Timo/genética , Neoplasias del Timo/metabolismo , Neoplasias del Timo/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
20.
Proc Natl Acad Sci U S A ; 112(46): E6311-20, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26578791

RESUMEN

As nucleators of the mitotic spindle and primary cilium, centrosomes play crucial roles in equal segregation of DNA content to daughter cells, coordination of growth and differentiation, and transduction of homeostatic cues. Whereas the majority of mammalian cells carry no more than two centrosomes per cell, exceptions to this rule apply in certain specialized tissues and in select disease states, including cancer. Centrosome amplification, or the condition of having more than two centrosomes per cell, has been suggested to contribute to instability of chromosomes, imbalance in asymmetric divisions, and reorganization of tissue architecture; however, the degree to which these conditions are a direct cause of or simply a consequence of human disease is poorly understood. Here we addressed this issue by generating a mouse model inducing centrosome amplification in a naturally proliferative epithelial tissue by elevating Polo-like kinase 4 (Plk4) expression in the skin epidermis. By altering centrosome numbers, we observed multiciliated cells, spindle orientation errors, and chromosome segregation defects within developing epidermis. None of these defects was sufficient to impart a proliferative advantage within the tissue, however. Rather, impaired mitoses led to p53-mediated cell death and contributed to defective growth and stratification. Despite these abnormalities, mice remained viable and healthy, although epidermal cells with centrosome amplification were still appreciable. Moreover, these abnormalities were insufficient to disrupt homeostasis and initiate or enhance tumorigenesis, underscoring the powerful surveillance mechanisms in the skin.


Asunto(s)
División Celular Asimétrica/fisiología , Centrosoma/metabolismo , Epidermis/metabolismo , Homeostasis/fisiología , Animales , Muerte Celular/fisiología , Células Epidérmicas , Regulación Enzimológica de la Expresión Génica/fisiología , Humanos , Ratones , Ratones Noqueados , Mitosis/fisiología , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA