Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Annu Rev Cell Dev Biol ; 31: 575-91, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26359775

RESUMEN

One of the major challenges in biology is to explain how complex tissues and organs arise from the collective action of individual polarized cells. The best-studied model of this process is the cross talk between individual epithelial cells during their polarization to form the multicellular epithelial lumen during tissue morphogenesis. Multiple mechanisms of apical lumen formation have been proposed. Some epithelial lumens form from preexisting polarized epithelial structures. However, de novo lumen formation from nonpolarized cells has recently emerged as an important driver of epithelial tissue morphogenesis, especially during the formation of small epithelial tubule networks. In this review, we discuss the latest findings regarding the mechanisms and regulation of de novo lumen formation in vitro and in vivo.


Asunto(s)
Polaridad Celular/fisiología , Células Epiteliales/fisiología , Morfogénesis/fisiología , Transporte de Proteínas/fisiología , Animales , Humanos
2.
Dev Dyn ; 243(12): 1511-23, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25130183

RESUMEN

BACKGROUND: Schwann cells, which arise from the neural crest, are the myelinating glia of the peripheral nervous system. During development neural crest and their Schwann cell derivatives engage in a sequence of events that comprise delamination from the neuroepithelium, directed migration, axon ensheathment, and myelin membrane synthesis. At each step neural crest and Schwann cells are polarized, suggesting important roles for molecules that create cellular asymmetries. In this work we investigated the possibility that one polarity protein, Pard3, contributes to the polarized features of neural crest and Schwann cells that are associated with directed migration and myelination. RESULTS: We analyzed mutant zebrafish embryos deficient for maternal and zygotic pard3 function. Time-lapse imaging revealed that neural crest delamination was normal but that migrating cells were disorganized with substantial amounts of overlapping membrane. Nevertheless, neural crest cells migrated to appropriate peripheral targets. Schwann cells wrapped motor axons and, although myelin gene expression was delayed, myelination proceeded to completion. CONCLUSIONS: Pard3 mediates contact inhibition between neural crest cells and promotes timely myelin gene expression but is not essential for neural crest migration or myelination.


Asunto(s)
Proteínas Portadoras/biosíntesis , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Cresta Neural/embriología , Células de Schwann/metabolismo , Proteínas de Pez Cebra/biosíntesis , Pez Cebra/embriología , Animales , Axones/metabolismo , Proteínas Portadoras/genética , Polaridad Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Cresta Neural/citología , Células de Schwann/citología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
3.
Dev Cell ; 27(4): 387-98, 2013 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-24239515

RESUMEN

Asymmetric self-renewing division of neural precursors is essential for brain development. Partitioning-defective (Par) proteins promote self-renewal, and their asymmetric distribution provides a mechanism for asymmetric division. Near the end of neural development, most asymmetric division ends and precursors differentiate. This correlates with Par protein disappearance, but mechanisms that cause downregulation are unknown. MicroRNAs can promote precursor differentiation but have not been linked to Par protein regulation. We tested a hypothesis that microRNA miR-219 promotes precursor differentiation by inhibiting Par proteins. Neural precursors in zebrafish larvae lacking miR-219 function retained apical proteins, remained in the cell cycle, and failed to differentiate. miR-219 inhibited expression via target sites within the 3' untranslated sequence of pard3 and prkci mRNAs, which encode Par proteins, and blocking miR-219 access to these sites phenocopied loss of miR-219 function. We propose that negative regulation of Par protein expression by miR-219 promotes cell-cycle exit and differentiation.


Asunto(s)
Diferenciación Celular , Polaridad Celular , MicroARNs/genética , Neurogénesis/genética , Neuronas/citología , Células Madre/citología , Proteínas de Pez Cebra/metabolismo , Animales , Western Blotting , Ciclo Celular , Proliferación Celular , Técnicas para Inmunoenzimas , Hibridación in Situ , Luciferasas/metabolismo , Neuronas/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética
4.
Artículo en Inglés | MEDLINE | ID: mdl-23576382

RESUMEN

The neural crest (NC) is first induced as an epithelial population of cells at the neural plate border requiring complex signaling between bone morphogenetic protein, Wnt, and fibroblast growth factors to differentiate the neural and NC fate from the epidermis. Remarkably, following induction, these cells undergo an epithelial-to-mesenchymal transition (EMT), delaminate from the neural tube, and migrate through various tissue types and microenvironments before reaching their final destination where they undergo terminal differentiation. This process is mirrored in cancer metastasis, where a primary tumor will undergo an EMT before migrating and invading other cell populations to create a secondary tumor site. In recent years, as our understanding of NC EMT and migration has deepened, important new insights into tumorigenesis and metastasis have also been achieved. These discoveries have been driven by the observation that many cancers misregulate developmental genes to reacquire proliferative and migratory states. In this review, we examine how the NC provides an excellent model for studying EMT and migration. These data are discussed from the perspective of the gene regulatory networks that control both NC and cancer cell EMT and migration. Deciphering these processes in a comparative manner will expand our knowledge of the underlying etiology and pathogenesis of cancer and promote the development of novel targeted therapeutic strategies for cancer patients.


Asunto(s)
Cresta Neural/citología , Adhesión Celular , Movimiento Celular , Transición Epitelial-Mesenquimal , Humanos , Metástasis de la Neoplasia , Neoplasias/metabolismo , Neoplasias/patología , Cresta Neural/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo
5.
Immunogenetics ; 61(3): 177-87, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19107381

RESUMEN

Cynomolgus macaques (Macaca fascicularis) provide increasingly common models for infectious disease research. Several geographically distinct populations of these macaques from Southeast Asia and the Indian Ocean island of Mauritius are available for pathogenesis studies. Though host genetics may profoundly impact results of such studies, similarities and differences between populations are often overlooked. In this study we identified 47 full-length MHC class I nucleotide sequences in 16 cynomolgus macaques of Filipino origin. The majority of MHC class I sequences characterized (39 of 47) were unique to this regional population. However, we discovered eight sequences with perfect identity and six sequences with close similarity to previously defined MHC class I sequences from other macaque populations. We identified two ancestral MHC haplotypes that appear to be shared between Filipino and Mauritian cynomolgus macaques, notably a Mafa-B haplotype that has previously been shown to protect Mauritian cynomolgus macaques against challenge with a simian/human immunodeficiency virus, SHIV(89.6P). We also identified a Filipino cynomolgus macaque MHC class I sequence for which the predicted protein sequence differs from Mamu-B*17 by a single amino acid. This is important because Mamu-B*17 is strongly associated with protection against simian immunodeficiency virus (SIV) challenge in Indian rhesus macaques. These findings have implications for the evolutionary history of Filipino cynomolgus macaques as well as for the use of this model in SIV/SHIV research protocols.


Asunto(s)
Genes MHC Clase I , Macaca fascicularis/genética , Animales , ADN Complementario , Haplotipos , Antígenos de Histocompatibilidad Clase I/química , Filipinas , Filogenia
6.
Immunogenetics ; 60(10): 575-84, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18629489

RESUMEN

Rhesus macaques (Macaca mulatta) provide well-established models for studying human disease pathogenesis and vaccine development. When challenged with infectious agents, macaques exhibit individual differences in susceptibility. An important determinant of these differences is the complement of major histocompatability complex (MHC) class I sequences expressed by each animal. Although previous studies have reported strong associations between MHC expression and disease outcome, a rapid, cost-effective method for high-resolution MHC genotyping in macaques is lacking. In this study, we adapted a modified heteroduplex assay, reference strand-mediated conformational analysis (RSCA) to an ABI 3130xl capillary electrophoresis genetic analyzer for macaque MHC class I genotyping. For validation, we investigated the concordance of RSCA genotyping for 14 MHC class I sequences in 12 Chinese rhesus macaques whose genotypes were established through complementary DNA cloning and sequencing of MHC class I sequences. We observed a concordance greater than 98% between RSCA and the cloning and sequencing data. Furthermore, RSCA confirmed the presence of MHC haplotype sharing between three macaques as predicted previously by microsatellite analysis. RSCA genotyping of an additional 25 Chinese rhesus macaques demonstrated that the frequency of these 14 MHC class I sequences ranged from 5% to 32%, with the Mamu-A1*2601 sequence being most common in this cohort. Capillary RSCA genotyping has the potential to enable researchers to rapidly evaluate MHC class I genotypes in rhesus macaques and associate specific MHC sequences with disease susceptibility.


Asunto(s)
Genes MHC Clase I/genética , Antígenos de Histocompatibilidad Clase I/genética , Macaca mulatta/genética , Polimorfismo Conformacional Retorcido-Simple , Animales , Electroforesis Capilar , Genotipo , Haplotipos
7.
PLoS One ; 3(6): e2384, 2008 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-18545705

RESUMEN

BACKGROUND: Thus far, live attenuated SIV has been the most successful method for vaccinating macaques against pathogenic SIV challenge; however, it is not clear what mechanisms are responsible for this protection. Adoptive transfer studies in mice have been integral to understanding live attenuated vaccine protection in models like Friend virus. Previous adoptive transfers in primates have failed as transferred cells are typically cleared within hours after transfer. METHODOLOGY/ PRINCIPAL FINDINGS: Here we describe adoptive transfer studies in Mauritian origin cynomolgus macaques (MCM), a non-human primate model with limited MHC diversity. Cells transferred between unrelated MHC-matched macaques persist for at least fourteen days but are rejected within 36 hours in MHC-mismatched macaques. Cells trafficked from the blood to peripheral lymphoid tissues within 12 hours of transfer. CONCLUSIONS/SIGNIFICANCE: MHC-matched MCM provide the first viable primate model for adoptive transfer studies. Because macaques infected with SIV are the best model for HIV/AIDS pathogenesis, we can now directly study the correlates of protective immune responses to AIDS viruses. For example, plasma viral loads following pathogenic SIV challenge are reduced by several orders of magnitude in macaques previously immunized with attenuated SIV. Adoptive transfer of lymphocyte subpopulations from vaccinated donors into SIV-naïve animals may define the immune mechanisms responsible for protection and guide future vaccine development.


Asunto(s)
Traslado Adoptivo , Animales Salvajes , Linfocitos T CD8-positivos/inmunología , Complejo Mayor de Histocompatibilidad , Animales , Secuencia de Bases , Cartilla de ADN , Haplotipos , Macaca fascicularis , Repeticiones de Microsatélite/genética , Virus de la Inmunodeficiencia de los Simios/inmunología
8.
Immunogenetics ; 60(7): 339-51, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18504574

RESUMEN

Cynomolgus macaques (Macaca fascicularis) are quickly becoming a useful model for infectious disease and transplantation research. Even though cynomolgus macaques from different geographic regions are used for these studies, there has been limited characterization of full-length major histocompatibility complex (MHC) class I immunogenetics of distinct geographic populations. Here, we identified 48 MHC class I cDNA nucleotide sequences in eleven Indonesian cynomolgus macaques, including 41 novel Mafa-A and Mafa-B sequences. We found seven MHC class I sequences in Indonesian macaques that were identical to MHC class I sequences identified in Malaysian or Mauritian macaques. Sharing of nucleotide sequences between these geographically distinct populations is also consistent with the hypothesis that Indonesia was a source of the Mauritian macaque population. In addition, we found that the Indonesian cDNA sequence Mafa-B7601 is identical throughout its peptide binding domain to Mamu-B03, an allele that has been associated with control of Simian immunodeficiency virus (SIV) viremia in Indian rhesus macaques. Overall, a better understanding of the MHC class I alleles present in Indonesian cynomolgus macaques improves their value as a model for disease research, and it better defines the biogeography of cynomolgus macaques throughout Southeast Asia.


Asunto(s)
Alelos , Genes MHC Clase I , Macaca fascicularis/genética , Animales , Genética de Población , Indonesia , Macaca fascicularis/inmunología
9.
Immunogenetics ; 60(1): 37-46, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18097659

RESUMEN

The rhesus macaque (Macaca mulatta) is an excellent model for human disease and vaccine research. Two populations exhibiting distinctive morphological and physiological characteristics, Indian- and Chinese-origin rhesus macaques, are commonly used in research. Genetic analysis has focused on the Indian macaque population, but the accessibility of these animals for research is limited. Due to their greater availability, Chinese rhesus macaques are now being used more frequently, particularly in vaccine and biodefense studies, although relatively little is known about their immunogenetics. In this study, we discovered major histocompatibility complex (MHC) class I cDNAs in 12 Chinese rhesus macaques and detected 41 distinct Mamu-A and Mamu-B sequences. Twenty-seven of these class I cDNAs were novel, while six and eight of these sequences were previously reported in Chinese and Indian rhesus macaques, respectively. We then performed microsatellite analysis on DNA from these 12 animals, as well as an additional 18 animals, and developed sequence specific primer PCR (PCR-SSP) assays for eight cDNAs found in multiple animals. We also examined our cohort for potential admixture of Chinese and Indian origin animals using a recently developed panel of single nucleotide polymorphisms (SNPs). The discovery of 27 novel MHC class I sequences in this analysis underscores the genetic diversity of Chinese rhesus macaques and contributes reagents that will be valuable for studying cellular immunology in this population.


Asunto(s)
Genes MHC Clase I , Antígenos de Histocompatibilidad Clase I/genética , Macaca mulatta/genética , Macaca mulatta/inmunología , Alelos , Animales , China , Humanos , Datos de Secuencia Molecular
10.
Immunogenetics ; 59(6): 449-62, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17384942

RESUMEN

There are currently no nonhuman primate models with fully defined major histocompatibility complex (MHC) class II genetics. We recently showed that six common MHC haplotypes account for essentially all MHC diversity in cynomolgus macaques (Macaca fascicularis) from the island of Mauritius. In this study, we employ complementary DNA cloning and sequencing to comprehensively characterize full length MHC class II alleles expressed at the Mafa-DPA, -DPB, -DQA, -DQB, -DRA, and -DRB loci on the six common haplotypes. We describe 34 full-length MHC class II alleles, 12 of which are completely novel. Polymorphism was evident at all six loci including DPA, a locus thought to be monomorphic in rhesus macaques. Similar to other Old World monkeys, Mauritian cynomolgus macaques (MCM) share MHC class II allelic lineages with humans at the DQ and DR loci, but not at the DP loci. Additionally, we identified extensive sharing of MHC class II alleles between MCM and other nonhuman primates. The characterization of these full-length-expressed MHC class II alleles will enable researchers to generate MHC class II transferent cell lines, tetramers, and other molecular reagents that can be used to explore CD4+ T lymphocyte responses in MCM.


Asunto(s)
Genes MHC Clase II , Variación Genética , Haplotipos , Macaca fascicularis/genética , Alelos , Secuencia de Aminoácidos , Animales , Humanos , Datos de Secuencia Molecular , Filogenia
11.
J Virol ; 81(1): 349-61, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17035320

RESUMEN

Nonhuman primates are widely used to study correlates of protective immunity in AIDS research. Successful cellular immune responses have been difficult to identify because heterogeneity within macaque major histocompatibility complex (MHC) genes results in quantitative and qualitative differences in immune responses. Here we use microsatellite analysis to show that simian immunodeficiency virus (SIV)-susceptible cynomolgus macaques (Macaca fascicularis) from the Indian Ocean island of Mauritius have extremely simple MHC genetics, with six common haplotypes accounting for two-thirds of the MHC haplotypes in feral animals. Remarkably, 39% of Mauritian cynomolgus macaques carry at least one complete copy of the most frequent MHC haplotype, and 8% of these animals are homozygous. In stark contrast, entire MHC haplotypes are rarely conserved in unrelated Indian rhesus macaques. After intrarectal infection with highly pathogenic SIVmac239 virus, a pair of MHC-identical Mauritian cynomolgus macaques mounted concordant cellular immune responses comparable to those previously reported for a pair of monozygotic twins infected with the same strain of human immunodeficiency virus. Our identification of relatively abundant SIV-susceptible, MHC-identical macaques will facilitate research into protective cellular immunity.


Asunto(s)
Genes MHC Clase II , Genes MHC Clase I , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Virus de la Inmunodeficiencia de los Simios/fisiología , Animales , Variación Genética , Haplotipos , Macaca , Mauricio , Repeticiones de Microsatélite , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...