Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34161290

RESUMEN

Defects or deficiencies in nuclear lamins cause pathology in many cell types, and recent studies have implicated nuclear membrane (NM) ruptures as a cause of cell toxicity. We previously observed NM ruptures and progressive cell death in the developing brain of lamin B1-deficient mouse embryos. We also observed frequent NM ruptures and DNA damage in nuclear lamin-deficient fibroblasts. Factors modulating susceptibility to NM ruptures remain unclear, but we noted low levels of LAP2ß, a chromatin-binding inner NM protein, in fibroblasts with NM ruptures. Here, we explored the apparent link between LAP2ß and NM ruptures in nuclear lamin-deficient neurons and fibroblasts, and we tested whether manipulating LAP2ß expression levels would alter NM rupture frequency. In cortical plate neurons of lamin B1-deficient embryos, we observed a strong correlation between low LAP2ß levels and NM ruptures. We also found low LAP2ß levels and frequent NM ruptures in neurons of cultured Lmnb1-/- neurospheres. Reducing LAP2ß expression in Lmnb1-/- neurons with an siRNA markedly increased the NM rupture frequency (without affecting NM rupture duration), whereas increased LAP2ß expression eliminated NM ruptures and reduced DNA damage. Consistent findings were observed in nuclear lamin-deficient fibroblasts. Reduced LAP2ß expression increased NM ruptures, whereas increased LAP2ß expression virtually abolished NM ruptures. Increased LAP2ß expression nearly abolished NM ruptures in cells subjected to mechanical stress (an intervention that increases NM ruptures). Our studies showed that increasing LAP2ß expression bolsters NM integrity in nuclear lamin-deficient cells and markedly reduces NM rupture frequency.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Lamina Tipo B/deficiencia , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Membrana Nuclear/metabolismo , Animales , Muerte Celular , Diferenciación Celular , Corteza Cerebral/patología , Daño del ADN , Embrión de Mamíferos/metabolismo , Lamina Tipo A/deficiencia , Lamina Tipo A/metabolismo , Lamina Tipo B/metabolismo , Ratones Noqueados , Especificidad de Órganos
2.
FEBS J ; 288(1): 56-80, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32961620

RESUMEN

Cellular senescence is a physiological mechanism whereby a proliferating cell undergoes a stable cell cycle arrest upon damage or stress and elicits a secretory phenotype. This highly dynamic and regulated cellular state plays beneficial roles in physiology, such as during embryonic development and wound healing, but it can also result in antagonistic effects in age-related pathologies, degenerative disorders, ageing and cancer. In an effort to better identify this complex state, and given that a universal marker has yet to be identified, a general set of hallmarks describing senescence has been established. However, as the senescent programme becomes more defined, further complexities, including phenotype heterogeneity, have emerged. This significantly complicates the recognition and evaluation of cellular senescence, especially within complex tissues and living organisms. To address these challenges, substantial efforts are currently being made towards the discovery of novel and more specific biomarkers, optimized combinatorial strategies and the development of emerging detection techniques. Here, we compile such advances and present a multifactorial guide to identify and assess cellular senescence in cell cultures, tissues and living organisms. The reliable assessment and identification of senescence is not only crucial for better understanding its underlying biology, but also imperative for the development of diagnostic and therapeutic strategies aimed at targeting senescence in the clinic.


Asunto(s)
Envejecimiento/genética , Puntos de Control del Ciclo Celular/genética , Senescencia Celular/genética , Heterocromatina/metabolismo , Mitocondrias/genética , Envejecimiento/metabolismo , Animales , Animales Modificados Genéticamente , Biomarcadores/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Forma de la Célula/genética , Células Cultivadas , Citocinas/genética , Citocinas/inmunología , Daño del ADN , Sitios Genéticos , Heterocromatina/química , Humanos , Lamina Tipo B/deficiencia , Lamina Tipo B/genética , Lisosomas/metabolismo , Mitocondrias/metabolismo , Mitocondrias/patología
3.
Nucleus ; 11(1): 237-249, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32910721

RESUMEN

The nuclear membranes function as a barrier to separate the cell nucleus from the cytoplasm, but this barrier can be compromised by nuclear membrane ruptures, leading to intermixing of nuclear and cytoplasmic contents. Spontaneous nuclear membrane ruptures (i.e., ruptures occurring in the absence of mechanical stress) have been observed in cultured cells, but they are more frequent in the setting of defects or deficiencies in nuclear lamins and when cells are subjected to mechanical stress. Nuclear membrane ruptures in cultured cells have been linked to DNA damage, but the relevance of ruptures to developmental or physiologic processes in vivo has received little attention. Recently, we addressed that issue by examining neuronal migration in the cerebral cortex, a developmental process that subjects the cell nucleus to mechanical stress. In the setting of lamin B1 deficiency, we observed frequent nuclear membrane ruptures in migrating neurons in the developing cerebral cortex and showed that those ruptures are likely the cause of observed DNA damage, neuronal cell death, and profound neuropathology. In this review, we discuss the physiologic relevance of nuclear membrane ruptures, with a focus on migrating neurons in cell culture and in the cerebral cortex of genetically modified mice.


Asunto(s)
Corteza Cerebral/metabolismo , Daño del ADN , Lamina Tipo B/deficiencia , Neuronas/metabolismo , Lámina Nuclear/metabolismo , Polineuropatías/metabolismo , Animales , Movimiento Celular/genética , Corteza Cerebral/patología , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Ratones , Ratones Transgénicos , Neuronas/patología , Lámina Nuclear/genética , Lámina Nuclear/patología , Polineuropatías/genética , Polineuropatías/patología
4.
Nucleus ; 6(3): 183-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25875575

RESUMEN

Gradual loss of tissue function (or homeostasis) is a natural process of aging and is believed to cause many age-associated diseases. In human epidemiology studies, the low-grade and chronic systemic inflammation in elderly has been correlated with the development of aging related pathologies. Although it is suspected that tissue decline is related to systemic inflammation, the cause and consequence of these aging phenomena are poorly understood. By studying the Drosophila fat body and gut, we have uncovered a mechanism by which lamin-B loss in the fat body upon aging induces age-associated systemic inflammation. This chronic inflammation results in the repression of gut local immune response, which in turn leads to the over-proliferation and mis-differentiation of the intestinal stem cells, thereby resulting in gut hyperplasia. Here we discuss the implications and remaining questions in light of our published findings and new observations.


Asunto(s)
Envejecimiento/metabolismo , Drosophila melanogaster/metabolismo , Cuerpo Adiposo/metabolismo , Lamina Tipo B/genética , Túbulos de Malpighi/metabolismo , Envejecimiento/genética , Envejecimiento/patología , Animales , Células Cultivadas , Senescencia Celular , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Cuerpo Adiposo/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , Homeostasis , Humanos , Inflamación , Lamina Tipo B/deficiencia , Túbulos de Malpighi/patología , Ratones , Lámina Nuclear/química , Lámina Nuclear/metabolismo , Lámina Nuclear/patología , Especificidad de Órganos , Transducción de Señal , Telómero/química , Telómero/metabolismo
5.
Nucleus ; 6(1): 66-76, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25738644

RESUMEN

More than 20 mutations in the gene encoding A-type lamins (LMNA) cause progeria, a rare premature aging disorder. The major pathognomonic hallmarks of progeria cells are seen as nuclear deformations or blebs that are related to the redistribution of A- and B-type lamins within the nuclear lamina. However, the functional significance of these progeria-associated blebs remains unknown. We have carried out an analysis of the structural and functional consequences of progeria-associated nuclear blebs in dermal fibroblasts from a progeria patient carrying a rare point mutation p.S143F (C428T) in lamin A/C. These blebs form microdomains that are devoid of major structural components of the nuclear envelope (NE)/lamina including B-type lamins and nuclear pore complexes (NPCs) and are enriched in A-type lamins. Using laser capture microdissection and comparative genomic hybridization (CGH) analyses, we show that, while these domains are devoid of centromeric heterochromatin and gene-poor regions of chromosomes, they are enriched in gene-rich chromosomal regions. The active form of RNA polymerase II is also greatly enriched in blebs as well as nascent RNA but the nuclear co-activator SKIP is significantly reduced in blebs compared to other transcription factors. Our results suggest that the p.S143F progeria mutation has a severe impact not only on the structure of the lamina but also on the organization of interphase chromatin domains and transcription. These structural defects are likely to contribute to gene expression changes reported in progeria and other types of laminopathies.


Asunto(s)
Núcleo Celular/genética , Cromosomas Humanos/genética , Lamina Tipo B/deficiencia , Progeria/genética , Progeria/patología , Núcleo Celular/metabolismo , Niño , Femenino , Humanos , Lamina Tipo B/genética , Mutación , Transcripción Genética
6.
FASEB J ; 28(8): 3423-34, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24732130

RESUMEN

Nuclear lamin B1 (LMNB1) constitutes one of the major structural proteins in the lamina mesh. We silenced the expression of LMNB1 by RNA interference in the colon cancer cell line DLD-1 and showed a dramatic redistribution of H3K27me3 from the periphery to a more homogeneous nuclear dispersion. In addition, we observed telomere attrition and an increased frequency of micronuclei and nuclear blebs. By 3D-FISH analyses, we demonstrated that the volume and surface of chromosome territories were significantly larger in LMNB1-depleted cells, suggesting that LMNB1 is required to maintain chromatin condensation in interphase nuclei. These changes led to a prolonged S phase due to activation of Chk1. Finally, silencing of LMNB1 resulted in extensive changes in alternative splicing of multiple genes and in a higher number of enlarged nuclear speckles. Taken together, our results suggest a mechanistic role of the nuclear lamina in the organization of chromosome territories, maintenance of genome integrity and proper gene splicing.


Asunto(s)
Lamina Tipo B/fisiología , Fase S/fisiología , Adenocarcinoma/patología , Línea Celular Tumoral , Núcleo Celular/ultraestructura , Cromosomas Humanos Par 18/ultraestructura , Cromosomas Humanos Par 19/ultraestructura , Neoplasias del Colon/patología , Heterocromatina/fisiología , Histonas/metabolismo , Humanos , Hibridación Fluorescente in Situ , Interfase , Lamina Tipo B/deficiencia , Metilación , Procesamiento Proteico-Postraduccional , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Fase S/efectos de los fármacos , Acortamiento del Telómero/efectos de los fármacos
7.
Mol Biol Cell ; 23(11): 2066-75, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22496421

RESUMEN

Cellular senescence is a potent tumor-suppressive mechanism that arrests cell proliferation and has been linked to aging. However, studies of senescence have been impeded by the lack of simple, exclusive biomarkers of the senescent state. Senescent cells develop characteristic morphological changes, which include enlarged and often irregular nuclei and chromatin reorganization. Because alterations to the nuclear lamina can affect both nuclear morphology and gene expression, we examined the nuclear lamina of senescent cells. We show here than lamin B1 is lost from primary human and murine cell strains when they are induced to senesce by DNA damage, replicative exhaustion, or oncogene expression. Lamin B1 loss did not depend on the p38 mitogen-activated protein kinase, nuclear factor-κB, ataxia telangiectasia-mutated kinase, or reactive oxygen species signaling pathways, which are positive regulators of senescent phenotypes. However, activation of either the p53 or pRB tumor suppressor pathway was sufficient to induce lamin B1 loss. Lamin B1 declined at the mRNA level via a decrease in mRNA stability rather than by the caspase-mediated degradation seen during apoptosis. Last, lamin B1 protein and mRNA declined in mouse tissue after senescence was induced by irradiation. Our findings suggest that lamin B1 loss can serve as biomarker of senescence both in culture and in vivo.


Asunto(s)
Senescencia Celular , Lamina Tipo B/deficiencia , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Biomarcadores/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Senescencia Celular/efectos de la radiación , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta en la Radiación , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Lamina Tipo B/metabolismo , Ratones , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Radiación Ionizante , Especies Reactivas de Oxígeno/metabolismo , Proteína de Retinoblastoma/metabolismo , Transducción de Señal/efectos de la radiación , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Med Clin (Barc) ; 138(5): 208-14, 2012 Mar 03.
Artículo en Español | MEDLINE | ID: mdl-21632068

RESUMEN

Laminopathies are a group of diseases that share wrong codification of lamins, building proteins of the nuclear lamina. Different tissues are affected in those disorders: striated muscle, adipose tissue, central or peripheral nervous system and aging process. Emery-Dreifuss muscular dystrophy and Hutchinson-Gildford Progery Syndrome are two examples of laminopathies. Other diseases, due to mutations in different genes, impair lamins function by a direct or an indirect way and they are frequently considered together. The last decade has seen an increasing interest and scientific advances on laminopathies that will allow us to answer key questions regarding metabolism, insulin resistance, sudden death and aging. Laminopathies are reviewed in this article from a molecular, pathogenic and clinical point of view.


Asunto(s)
Laminas/deficiencia , Membrana Nuclear/patología , Envejecimiento/genética , Envejecimiento/fisiología , Animales , Muerte Súbita , Modelos Animales de Enfermedad , Humanos , Resistencia a la Insulina , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Lamina Tipo A/deficiencia , Lamina Tipo A/genética , Lamina Tipo B/deficiencia , Lamina Tipo B/genética , Laminas/genética , Laminas/fisiología , Ratones , Ratones Noqueados , Modelos Biológicos , Distrofia Muscular de Emery-Dreifuss/genética , Especificidad de Órganos , Progeria/genética
9.
J Pathol ; 226(5): 735-45, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22025297

RESUMEN

The lamins are major determinants of nuclear shape and chromatin organization and these features are frequently altered in prostate cancer (CaP). Human CaP cell lines frequently have nuclear lobulations, which are enriched in A-type lamins but lack B-type lamins and have been defined as lamin B-deficient microdomains (LDMDs). LDMD frequency is correlated with CaP cell line aggressiveness and increased cell motility. In addition, LNCaP cells grown in the presence of dihydrotestosterone (DHT) show an increased frequency of LDMDs. The LDMDs are enriched in activated RNA polymerase II (Pol IIo) and androgen receptor (AR) and A-type lamins form an enlarged meshwork that appears to co-align with chromatin fibres and AR. Furthermore, fluorescence in situ hybridization and comparative genomic hybridization demonstrated that chromosomal regions associated with CaP susceptibility are preferentially localized to LDMDs. Surprisingly, these regions lack histone marks for transcript elongation and exhibit reduced BrU incorporation, suggesting that Pol II is stalled within LDMDs. Real-time PCR of genes near androgen response elements (AREs) was used to compare transcription between cells containing LDMDs and controls. Genes preferentially localized to LDMDs showed significantly decreased expression, while genes in the main nuclear body were largely unaffected. Furthermore, LDMDs were observed in human CaP tissue and the frequency was correlated with increased Gleason grade. These results imply that lamins are involved in chromatin organization and Pol II transcription, and provide insights into the development and progression of CaP.


Asunto(s)
Cromosomas Humanos/metabolismo , Regulación Neoplásica de la Expresión Génica , Lamina Tipo B/deficiencia , Microdominios de Membrana/metabolismo , Membrana Nuclear/metabolismo , Neoplasias de la Próstata/genética , Transcripción Genética , Sitios de Unión , Línea Celular Tumoral , Movimiento Celular , Forma del Núcleo Celular , Ensamble y Desensamble de Cromatina , Hibridación Genómica Comparativa , Regulación hacia Abajo , Activación Enzimática , Humanos , Hibridación Fluorescente in Situ , Lamina Tipo A/metabolismo , Lamina Tipo B/genética , Captura por Microdisección con Láser , Masculino , Microdominios de Membrana/patología , Microscopía Fluorescente , Clasificación del Tumor , Membrana Nuclear/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Polimerasa II/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Androgénicos/metabolismo , Factores de Tiempo , Análisis de Matrices Tisulares , Transfección
10.
Hum Mol Genet ; 20(18): 3537-44, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21659336

RESUMEN

Nuclear lamins are usually classified as A-type (lamins A and C) or B-type (lamins B1 and B2). A-type lamins have been implicated in multiple genetic diseases but are not required for cell growth or development. In contrast, B-type lamins have been considered essential in eukaryotic cells, with crucial roles in DNA replication and in the formation of the mitotic spindle. Knocking down the genes for B-type lamins (LMNB1, LMNB2) in HeLa cells has been reported to cause apoptosis. In the current study, we created conditional knockout alleles for mouse Lmnb1 and Lmnb2, with the goal of testing the hypothesis that B-type lamins are crucial for the growth and viability of mammalian cells in vivo. Using the keratin 14-Cre transgene, we bred mice lacking the expression of both Lmnb1 and Lmnb2 in skin keratinocytes (Lmnb1(Δ/Δ)Lmnb2(Δ/Δ)). Lmnb1 and Lmnb2 transcripts were absent in keratinocytes of Lmnb1(Δ/Δ)Lmnb2(Δ/Δ) mice, and lamin B1 and lamin B2 proteins were undetectable. But despite an absence of B-type lamins in keratinocytes, the skin and hair of Lmnb1(Δ/Δ)Lmnb2(Δ/Δ) mice developed normally and were free of histological abnormalities, even in 2-year-old mice. After an intraperitoneal injection of bromodeoxyuridine (BrdU), similar numbers of BrdU-positive keratinocytes were observed in the skin of wild-type and Lmnb1(Δ/Δ)Lmnb2(Δ/Δ) mice. Lmnb1(Δ/Δ)Lmnb2(Δ/Δ) keratinocytes did not exhibit aneuploidy, and their growth rate was normal in culture. These studies challenge the concept that B-type lamins are essential for proliferation and vitality of eukaryotic cells.


Asunto(s)
Proliferación Celular , Cabello/crecimiento & desarrollo , Queratinocitos/citología , Lamina Tipo B/deficiencia , Piel/crecimiento & desarrollo , Células 3T3 , Animales , Células Cultivadas , Femenino , Cabello/metabolismo , Células HeLa , Humanos , Queratinocitos/metabolismo , Lamina Tipo B/genética , Masculino , Ratones , Ratones Noqueados , Piel/metabolismo
12.
Proc Natl Acad Sci U S A ; 107(11): 5076-81, 2010 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-20145110

RESUMEN

Nuclear lamins are components of the nuclear lamina, a structural scaffolding for the cell nucleus. Defects in lamins A and C cause an array of human diseases, including muscular dystrophy, lipodystrophy, and progeria, but no diseases have been linked to the loss of lamins B1 or B2. To explore the functional relevance of lamin B2, we generated lamin B2-deficient mice and found that they have severe brain abnormalities resembling lissencephaly, with abnormal layering of neurons in the cerebral cortex and cerebellum. This neuronal layering abnormality is due to defective neuronal migration, a process that is dependent on the organized movement of the nucleus within the cell. These studies establish an essential function for lamin B2 in neuronal migration and brain development.


Asunto(s)
Cerebelo/anomalías , Cerebelo/embriología , Corteza Cerebral/anomalías , Corteza Cerebral/embriología , Lamina Tipo B/deficiencia , Animales , Movimiento Celular , Cerebelo/patología , Corteza Cerebral/patología , Silenciador del Gen , Lamina Tipo B/metabolismo , Ratones , Neuronas/patología
13.
Nucleus ; 1(5): 407-11, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21278813

RESUMEN

Nuclear lamins are major components of the nuclear lamina, and play essential roles in supporting the nucleus and organizing nuclear structures. While a large number of clinically important mutations have been mapped to the LMNA gene in humans, very few mutations have been associated with the B-type lamins. We have shown that lamin B2-deficiency in mice results in severe brain abnormalities. While the early stages of forebrain development in lamin B2-deficient mice appear to be normal, cortical neurons fail to migrate and organize into proper layers within the cerebral cortex. The morphogenesis of the hippocampus and cerebellum is also severely impaired. These phenotypes are reminiscent of lissencephaly, a human brain developmental disorder characterized by an abnormal neuronal migration. Most mutations in lissencephaly patients affect cytoplasmic regulators of nuclear translocation, which is a crucial step in neuronal migration. The phenotypes of lamin B2-deficient mice suggest that lamin B2 may also play a key role in nuclear translocation. Potential mechanisms for lamin B2 involvement, which include mechanical and non-mechanical roles, and participation in LINC complexes in the nuclear envelope, are discussed along with evidence that lamins B1 and B2 play distinct, cell-specific functions.


Asunto(s)
Movimiento Celular/fisiología , Cerebelo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hipocampo/metabolismo , Lamina Tipo B , Neuronas/fisiología , Animales , Cerebelo/crecimiento & desarrollo , Hipocampo/crecimiento & desarrollo , Lamina Tipo B/deficiencia , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Ratones , Ratones Noqueados , Mutación , Neurogénesis/fisiología , Lámina Nuclear/genética , Lámina Nuclear/metabolismo , Poro Nuclear/genética , Poro Nuclear/metabolismo
14.
J Biol Chem ; 282(27): 20015-26, 2007 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-17488709

RESUMEN

Mutations of the nuclear lamins cause a wide range of human diseases, including Emery-Dreifuss muscular dystrophy and Hutchinson-Gilford progeria syndrome. Defects in A-type lamins reduce nuclear structural integrity and affect transcriptional regulation, but few data exist on the biological role of B-type lamins. To assess the functional importance of lamin B1, we examined nuclear dynamics in fibroblasts from Lmnb1(Delta/Delta) and wild-type littermate embryos by time-lapse videomicroscopy. Here, we report that Lmnb1(Delta/Delta) cells displayed striking nuclear rotation, with approximately 90% of Lmnb1(Delta/Delta) nuclei rotating at least 90 degrees during an 8-h period. The rotation involved the nuclear interior as well as the nuclear envelope. The rotation of nuclei required an intact cytoskeletal network and was eliminated by expressing lamin B1 in cells. Nuclear rotation could also be abolished by expressing larger nesprin isoforms with long spectrin repeats. These findings demonstrate that lamin B1 serves a fundamental role within the nuclear envelope: anchoring the nucleus to the cytoskeleton.


Asunto(s)
Citoesqueleto/metabolismo , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Lamina Tipo B/metabolismo , Membrana Nuclear/metabolismo , Animales , Células Cultivadas , Citoesqueleto/genética , Citoesqueleto/patología , Embrión de Mamíferos/patología , Fibroblastos/patología , Humanos , Espacio Intranuclear/metabolismo , Espacio Intranuclear/patología , Lamina Tipo B/deficiencia , Ratones , Ratones Noqueados , Microscopía por Video , Membrana Nuclear/genética , Membrana Nuclear/patología , Progeria/genética , Progeria/metabolismo , Progeria/patología , Factores de Tiempo
15.
Rinsho Byori ; 53(1): 54-60, 2005 Jan.
Artículo en Japonés | MEDLINE | ID: mdl-15724491

RESUMEN

Gene abnormalities responsible for familial Pelger-Huet anomaly have been recently discovered. Abnormalities in sequence of Lamin B Receptor(LBR) gene results in a lack of LBR protein that is essential for chromatin-binding to nuclear membrane. In neutrophils lacking LBR protein shows abnormal bilobular or monolobular nuclear forms and hyper-condensed chromatin-aggregation. We re-analyzed distribution of such Pelger-Huet anomaly in other cell lineages; we found that not only neutrophils but erythroblasts, monocytes, lymphocytes, plasma cells, eosinophils and basophils are also carrying chromatin-hypercondensation. One third of megakaryocytes are also binucleated like neutrophils. We compared neutrophil morphology between familial Pelger-Huet anomaly and so called pseudo-Pelger-Huet anomaly observed in patients with myelodysplastic syndromes(MDS) and acute myeloid leukemia(AML). The neutrophils in MDS were much similar to those of the familial anomaly, but neutrophils of AML, such as t (8;21) M2-AML and t (15;17) M3-AML, showed more heterogeneous pattern in lobulation and chromatin-hypercondensation. Especially in M3, differentiation-induction by all-trans retinoic acid induced a marked neutrophilia with pseudo-Pelger-Huet anomaly without chromatin-hypercondensation. Lack of LBR protein in familial Pelger-Huet anomaly results in hypolobulation and chromatin-hypercondensation in neutrophils, but in other cells such as erythroblasts and lymphocytes only chromatin-hypercondensation can be observed. In contrast pseudo-Pelger-Huet anomaly are more heterogeneous in morphology compared to the familial anomaly. The lack of leukemic or MDS transformation in the familial anomaly is a sharp contrast to the neoplastic nature of the pseudo-Pelger-Huet anomaly. In conclusion, our morphological recognition of certain abnormality of cells shows an marked progression when genetic abnormality responsible for some of them are discovered, and often make us recognize a further heterogeneity in them. We, hematologists and technicians, must be well prepared to report our own observation of an un-explained morphological abnormality.


Asunto(s)
Núcleo Celular/genética , Núcleo Celular/patología , Neutrófilos/citología , Neutrófilos/patología , Anomalía de Pelger-Huët/sangre , Anomalía de Pelger-Huët/genética , Secuencia de Bases , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Humanos , Lamina Tipo B/deficiencia , Leucemia Mieloide Aguda/sangre , Mutación , Síndromes Mielodisplásicos/sangre , Receptores Citoplasmáticos y Nucleares/genética , Receptor de Lamina B
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA