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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Adv Exp Med Biol ; 1441: 145-153, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884709

RESUMEN

The development of the inflow tract is undoubtedly one of the most complex remodeling events in the formation of the four-chambered heart. It involves the creation of two separate atrial chambers, the formation of an atrial/atrioventricular (AV) septal complex, the incorporation of the caval veins and coronary sinus into the right atrium, and the remodeling events that result in pulmonary venous return draining into the left atrium. In these processes, the atrioventricular mesenchymal complex, consisting of the major atrioventricular (AV) cushions, the mesenchymal cap on the primary atrial septum (pAS), and the dorsal mesenchymal protrusion (DMP), plays a crucial role.


Asunto(s)
Atrios Cardíacos , Animales , Humanos , Seno Coronario/embriología , Seno Coronario/anomalías , Corazón/embriología , Mesodermo/embriología , Venas Pulmonares/anomalías
2.
Int J Mol Sci ; 24(2)2023 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-36675298

RESUMEN

The utilization of cardiac progenitor cells (CPCs) has been shown to induce favorable regenerative effects. While there are various populations of endogenous CPCs in the heart, there is no consensus regarding which population is ideal for cell-based regenerative therapy. Early-stage progenitor cells can be differentiated into all cardiovascular lineages, including cardiomyocytes and endothelial cells. Identifying an Islet-1+ (Isl-1+) early-stage progenitor population with enhanced stemness, multipotency and differentiation potential would be beneficial for the development of novel regenerative therapies. Here, we investigated the transcriptome of human neonatal Isl-1+ CPCs. Isl-1+ human neonatal CPCs exhibit enhanced stemness properties and were found to express Spalt-like transcription factor 4 (SALL4). SALL4 plays a role in embryonic development as well as proliferation and expansion of hematopoietic progenitor cells. SALL4, SOX2, EpCAM and TBX5 are co-expressed in the majority of Isl-1+ clones isolated from neonatal patients. The pre-mesendodermal transcript TFAP2C was identified in select Isl-1, SALL4, SOX2, EpCAM and TBX5 expressing clones. The ability to isolate and expand pre-mesendodermal stage cells from human patients is a novel finding that holds potential value for applications in regenerative medicine.


Asunto(s)
Células Endoteliales , Factores de Transcripción , Recién Nacido , Humanos , Factores de Transcripción/genética , Molécula de Adhesión Celular Epitelial , Diferenciación Celular/genética , Células Madre Hematopoyéticas , Células Clonales
3.
Int J Mol Sci ; 24(9)2023 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-37175796

RESUMEN

New stem cell and extracellular-vesicle-based therapies have the potential to improve outcomes for the increasing number of patients with heart failure. Since neonates have a significantly enhanced regenerative ability, we hypothesized that extracellular vesicles isolated from Islet-1+ expressing neonatal human cardiovascular progenitors (CPCs) will induce transcriptomic changes associated with improved regenerative capability when co-cultured with CPCs derived from adult humans. In order to test this hypothesis, we isolated extracellular vesicles from human neonatal Islet-1+ CPCs, analyzed the extracellular vesicle content using RNAseq, and treated adult CPCs with extracellular vesicles derived from neonatal CPCs to assess their functional effect. AKT, ERBB, and YAP1 transcripts were elevated in adult CPCs treated with neonatal CPC-derived extracellular vesicles. YAP1 is lost after the neonatal period but can stimulate cardiac regeneration. Our results demonstrate that YAP1 and additional transcripts associated with improved cardiovascular regeneration, as well as the activation of the cell cycle, can be achieved by the treatment of adult CPCs with neonatal CPC-derived extracellular vesicles. Progenitor cells derived from neonates secrete extracellular vesicles with the potential to stimulate and potentially improve functional effects in adult CPCs used for cardiovascular repair.


Asunto(s)
Células Madre Adultas , Vesículas Extracelulares , Recién Nacido , Humanos , Adulto , Miocitos Cardíacos/metabolismo , Células Cultivadas , Células Madre/metabolismo , Diferenciación Celular
4.
Cereb Cortex ; 31(11): 5206-5224, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34228108

RESUMEN

Cortical interneurons (GABAergic cells) arise during embryogenesis primarily from the medial and caudal ganglionic eminences (MGE and CGE, respectively) with a small population generated from the preoptic area (POA). Progenitors from the lateral ganglionic eminence (LGE) are thought to only generate GABAergic medium spiny neurons that populate the striatum and project to the globus pallidus. Here, we report evidence that neuronal precursors that express the LGE-specific transcription factor Islet1 (Isl1) can give rise to a small population of cortical interneurons. Lineage tracing and homozygous deletion of Nkx2.1 in Isl1 fate-mapped mice showed that neighboring MGE/POA-specific Nkx2.1 cells and LGE-specific Isl1 cells make both common and distinct lineal contributions towards cortical interneuron fate. Although the majority of cells had overlapping transcriptional domains between Nkx2.1 and Isl1, a population of Isl1-only derived cells also contributed to the adult cerebral cortex. The data indicate that Isl1-derived cells may originate from both the LGE and the adjacent LGE/MGE boundary regions to generate diverse neuronal progeny. Thus, a small population of neocortical interneurons appear to originate from Isl-1-positive precursors.


Asunto(s)
Neocórtex , Animales , Movimiento Celular/fisiología , Neuronas GABAérgicas , Regulación del Desarrollo de la Expresión Génica , Homocigoto , Interneuronas/fisiología , Ratones , Neocórtex/fisiología , Eliminación de Secuencia
5.
Food Microbiol ; 102: 103915, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34809941

RESUMEN

Listeria monocytogenes remains a threat to the food system and has led to numerous foodborne outbreaks worldwide. L. monocytogenes can establish itself in food production facilities by adhering to surfaces, resulting in increased resistance to environmental stressors. The aim of this study was to evaluate the adhesion ability of L. monocytogenes at 8 °C and to analyse associations between the observed phenotypes and genetic factors such as internalin A (inlA) genotypes, stress survival islet 1 (SSI-1) genotype, and clonal complex (CC). L. monocytogenes isolates (n = 184) were grown at 8 °C and 100% relative humidity for 15 days. The growth was measured by optical density at 600 nm every 24 h. Adherent cells were stained using crystal violet and quantified spectrophotometrically. Genotyping of inlA and SSI-1, multi-locus sequence typing, and a genome-wide association study (GWAS) were performed to elucidate the phenotype-genotype relationships in L. monocytogenes cold adhesion. Among all inlA genotypes, truncated inlA isolates had the highest mean adhered cells, ABS595nm = 0.30 ± 0.15 (Tukey HSD; P < 0.05), while three-codon deletion inlA isolates had the least mean adhered cells (Tukey HSD; P < 0.05). When SSI-1 was present, more cells adhered; less cells adhered when SSI-1 was absent (Welch's t-test; P < 0.05). Adhesion was associated with clonal complexes which have low clinical frequency, while reduced adhesion was associated with clonal complexes which have high frequency. The results of this study support that premature stop codons in the virulence gene inlA are associated with increased cold adhesion and that an invasion enhancing deletion in inlA is associated with decreased cold adhesion. This study also provides evidence to suggest that there is an evolutionary trade off between virulence and adhesion in L. monocytogenes. These results provide a greater understanding of L. monocytogenes adhesion which will aid in the development of strategies to reduce L. monocytogenes in the food system.


Asunto(s)
Adhesión Bacteriana , Listeria monocytogenes , Poliestirenos , Proteínas Bacterianas/genética , Microbiología de Alimentos , Estudios de Asociación Genética , Genómica , Listeria monocytogenes/clasificación , Listeria monocytogenes/genética , Tipificación de Secuencias Multilocus , Mutación
6.
Histochem Cell Biol ; 155(1): 133-143, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33070205

RESUMEN

In mammalian hearts, cardiomyocytes retain a transient capacity to proliferate and regenerate following injury before birth, whereas they lose proliferative capacity immediately after birth. It has also been known that cardiac progenitor cells including islet1-positive cells do not contribute to the cardiac repair and regeneration in mammals. In contrast, hearts of zebrafish, amphibians and reptiles maintain a regenerative ability throughout life. Here, we analyzed proliferative capacity of cardiac cells during cardiac development and post-ventricular resection using Xenopus laevis, especially focusing on islet1. Immunohistochemical examination showed that islet1-positive cells were present in a wide range of the ventricle and maintained high dividing ability after metamorphosis. Interestingly, the islet1-positive cells were preserved even at 1 year after metamorphosis, some of which showed tropomyosin expression. To assess the possibility of islet1-positive cells as a cellular resource, islet1 response to cardiac resection was analyzed, using adult hearts of 3 months after metamorphosis. Transient gene activation of islet1 in apical region was detected within 1 day after amputation. Histological analyses revealed that islet1-positive cells appeared in the vicinity of resection plane at 1 day post-amputation (dpa) and increased at 3 dpa in both tropomyosin-positive and tropomyosin-negative regions. Vascular labeling analysis by biotinylated dextran amine (BDA) indicated that the islet1-positive cells in a tropomyosin-negative region were closely associated with cardiac vessels. Moreover, dividing ability at this time point was peaked. The resected region was healed with tropomyosin-positive cardiomyocytes until 3 months post-amputation. These results suggest a role of islet1-positive cells as a cellular resource for vascularization and cardiogenesis in Xenopus laevis.


Asunto(s)
Proteínas con Homeodominio LIM/genética , Metamorfosis Biológica/genética , Factores de Transcripción/genética , Cicatrización de Heridas/genética , Animales , Células Cultivadas , Proteínas con Homeodominio LIM/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Transcripción/metabolismo , Xenopus laevis
7.
Int J Mol Sci ; 22(17)2021 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-34502291

RESUMEN

Early-stage mammalian embryos survive within a low oxygen tension environment and develop into fully functional, healthy organisms despite this hypoxic stress. This suggests that hypoxia plays a regulative role in fetal development that influences cell mobilization, differentiation, proliferation, and survival. The long-term hypoxic environment is sustained throughout gestation. Elucidation of the mechanisms by which cardiovascular stem cells survive and thrive under hypoxic conditions would benefit cell-based therapies where stem cell survival is limited in the hypoxic environment of the infarcted heart. The current study addressed the impact of long-term hypoxia on fetal Islet-1+ cardiovascular progenitor cell clones, which were isolated from sheep housed at high altitude. The cells were then cultured in vitro in 1% oxygen and compared with control Islet-1+ cardiovascular progenitor cells maintained at 21% oxygen. RT-PCR, western blotting, flow cytometry, and migration assays evaluated adaptation to long term hypoxia in terms of survival, proliferation, and signaling. Non-canonical Wnt, Notch, AKT, HIF-2α and Yap1 transcripts were induced by hypoxia. The hypoxic niche environment regulates these signaling pathways to sustain the dedifferentiation and survival of fetal cardiovascular progenitor cells.


Asunto(s)
Sistema Cardiovascular/embriología , Hipoxia de la Célula/fisiología , Células Madre/citología , Animales , Sistema Cardiovascular/citología , Ciclo Celular , Diferenciación Celular , Movimiento Celular , Supervivencia Celular , Femenino , Hipoxia/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ovinos , Células Madre/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Development ; 141(19): 3761-71, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25209244

RESUMEN

The T-box transcription factor Tbx1 and the LIM-homeodomain transcription factor Islet1 are key components in regulatory circuits that generate myogenic and cardiogenic lineage diversity in chordates. We show here that Org-1 and Tup, the Drosophila orthologs of Tbx1 and Islet1, are co-expressed and required for formation of the heart-associated alary muscles (AMs) in the abdomen. The same holds true for lineage-related muscles in the thorax that have not been described previously, which we name thoracic alary-related muscles (TARMs). Lineage analyses identified the progenitor cell for each AM and TARM. Three-dimensional high-resolution analyses indicate that AMs and TARMs connect the exoskeleton to the aorta/heart and to different regions of the midgut, respectively, and surround-specific tracheal branches, pointing to an architectural role in the internal anatomy of the larva. Org-1 controls tup expression in the AM/TARM lineage by direct binding to two regulatory sites within an AM/TARM-specific cis-regulatory module, tupAME. The contributions of Org-1 and Tup to the specification of Drosophila AMs and TARMs provide new insights into the transcriptional control of Drosophila larval muscle diversification and highlight new parallels with gene regulatory networks involved in the specification of cardiopharyngeal mesodermal derivatives in chordates.


Asunto(s)
Músculos Abdominales/citología , Linaje de la Célula/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/anatomía & histología , Regulación del Desarrollo de la Expresión Génica/fisiología , Modelos Anatómicos , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Músculos Abdominales/fisiología , Animales , Animales Modificados Genéticamente , Inmunoprecipitación de Cromatina , Drosophila/genética , Drosophila/fisiología , Inmunohistoquímica , Larva/anatomía & histología , Larva/fisiología , Imagen de Lapso de Tiempo , Vísceras/anatomía & histología
9.
J Appl Microbiol ; 123(1): 262-270, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28452154

RESUMEN

AIMS: Listeria monocytogenes nisin resistance increases when first exposed to NaCl and other stresses, such as low pH. In addition to environmental stressors, specific genomic elements can confer nisin resistance, such as the stress survival islet (SSI-1). As SSI-1 is variably present among L. monocytogenes strains, we wanted to determine if SSI-1 was associated with salt-induced nisin resistance. METHODS AND RESULTS: The presence of SSI-1 was determined using PCR for 48 strains of L. monocytogenes. When combined with multilocus sequence typing data, we found that the distribution of SSI-1 is clonal, where strains from clonal complexes (CC) 2, 6 and 11 do not have SSI-1, while strains from CCs 3, 5, 7 and 9 contain SSI-1. The impact of SSI-1 on salt-induced nisin resistance was dependent on CC. The average log decrease after 24 h of exposure to nisin at 7°C under salt-inducing conditions was 2·6 ± 1·1 for CC 9 strains and 2·3 ± 0·7 for CC 11 strains, which had significantly lower survival compared to the other CCs, such as 1·3 ± 0·3 for CC 6. Deletion of SSI-1 from a CC 7 strain demonstrated the role SSI-1 plays in salt-induced nisin resistance, as the deletion mutant had lower resistance compared to the parent strain. CONCLUSIONS: These data suggest that inducible nisin resistance in L. monocytogenes can be influenced by environmental conditions as well as the genetic composition of the strain, which should be considered when selecting control measures for ready-to-eat foods. SIGNIFICANCE AND IMPACT OF THE STUDY: The foodborne pathogen L. monocytogenes can grow in suboptimal conditions, including low temperature and high osmolarity, which makes it a safety concern for ready-to-eat foods. When using antimicrobial peptide inhibitors such as nisin, it is important to understand how food components can impact antimicrobial resistance across the genetic diversity of L. monocytogenes.

10.
Development ; 140(22): 4499-509, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24131630

RESUMEN

In contrast to adult vertebrates, which have limited capacities for neurogenesis, adult planarians undergo constitutive cellular turnover during homeostasis and are even able to regenerate a whole brain after decapitation. This enormous plasticity derives from pluripotent stem cells residing in the planarian body in large numbers. It is still obscure how these stem cells are programmed for differentiation into specific cell lineages and how lineage identity is maintained. Here we identify a Pitx transcription factor of crucial importance for planarian regeneration. In addition to patterning defects that are co-dependent on the LIM homeobox transcription factor gene islet1, which is expressed with pitx at anterior and posterior regeneration poles, RNAi against pitx results in islet1-independent specific loss of serotonergic (SN) neurons during regeneration. Besides its expression in terminally differentiated SN neurons we found pitx in stem cell progeny committed to the SN fate. Also, intact pitx RNAi animals gradually lose SN markers, a phenotype that depends neither on increased apoptosis nor on stem cell-based turnover or transdifferentiation into other neurons. We propose that pitx is a terminal selector gene for SN neurons in planarians that controls not only their maturation but also their identity by regulating the expression of the Serotonin production and transport machinery. Finally, we made use of this function of pitx and compared the transcriptomes of regenerating planarians with and without functional SN neurons, identifying at least three new neuronal targets of Pitx.


Asunto(s)
Linaje de la Célula , Factores de Transcripción Paired Box/metabolismo , Planarias/citología , Planarias/metabolismo , Neuronas Serotoninérgicas/citología , Neuronas Serotoninérgicas/metabolismo , Animales , Apoptosis/genética , Biomarcadores/metabolismo , Tipificación del Cuerpo/genética , Diferenciación Celular/genética , Transdiferenciación Celular/genética , Regulación de la Expresión Génica , Proteínas con Homeodominio LIM/metabolismo , Datos de Secuencia Molecular , Planarias/genética , Interferencia de ARN , Regeneración/genética , Serotonina/biosíntesis , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma/genética
11.
Biochem Biophys Res Commun ; 459(3): 504-8, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25747715

RESUMEN

Cadherin-6B induces bone morphogenetic protein (BMP) signaling to promote the epithelial mesenchymal transition (EMT) in the neural crest. We have previously found that knockdown of Cadherin-6B inhibits both BMP signaling and the emigration of the early pre-migratory neural crest cells from the dorsal neural tube. In this study, we found that inhibition of BMP signaling in the neural tube, mediated by the ectopic expression of Smad-6 or Noggin, decreased the size of the Islet-1-positive dorsal cell population. Knockdown or loss of function of Cadherin-6B suppressed the generation of Islet-1-expressing cells in the dorsal neural tube, but not the Lim-1/2 positive dorsal cell population. Our results thus indicate that Cadherin-6B is necessary for the generation of Islet-1-positive dorsal interneurons, as well as the initiation of pre-migratory neural crest cell emigration.


Asunto(s)
Proteínas Aviares/metabolismo , Cadherinas/metabolismo , Interneuronas/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas Aviares/antagonistas & inhibidores , Proteínas Aviares/genética , Proteínas Morfogenéticas Óseas/metabolismo , Cadherinas/antagonistas & inhibidores , Cadherinas/genética , Proteínas Portadoras/metabolismo , Movimiento Celular , Embrión de Pollo , Transición Epitelial-Mesenquimal , Técnicas de Silenciamiento del Gen , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/metabolismo , Tubo Neural/citología , Tubo Neural/embriología , Tubo Neural/metabolismo , Transducción de Señal , Proteína smad6/metabolismo
12.
Histopathology ; 66(2): 182-91, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24766278

RESUMEN

AIMS: Neuroendocrine middle ear adenoma (MEA) is a rare epithelial neoplasm of uncertain histogenesis that frequently shows neuroendocrine features. To date, <120 cases have been reported. The aims of the current study were to describe our experience with neuroendocrine MEA, to assess the frequency of specific neuroendocrine differentiation, and to test these lesions for transcription factors known to be expressed in a variety of other neuroendocrine tumours. METHODS AND RESULTS: We investigated six cases of neuroendocrine MEA, and stained them, for the first time, for the transcription factors CDX2, TTF1, PAX8, and ISL1 (islet-1). The patients were four men and two women (mean age, 39 years; range, 27-53 years). Two of four patients with extended follow-up (4.5-22 years) experienced recurrence at 92 months, and at 9 and 22 years, respectively. One case extending into the external ear coexisted with cholesteatoma. Histological examination showed trabecular, solid, acinar, glandular, cribriform, organoid, nested, diffuse non-cohesive plasmacytoid and pseudoalveolar patterns in varying combinations. Immunohistochemistry showed consistent expression of vimentin (4/4), pancytokeratin (6/6), synaptophysin (6/6), CD56 (4/4), and ISL1 (6/6). A CK7 antibody stained scattered cells in two of five cases. The myoepithelial markers and transcription factors TTF1, CDX2 or PAX8 were not expressed in any of the cases. CONCLUSIONS: Middle ear adenoma is an indolent, locally recurring, but generally non-metastasizing neoplasm with uniform expression of synaptophysin and ISL1, indicating true neuroendocrine differentiation. Because of its highly varied cellular and architectural appearance, MEA should be distinguished from tympanic paraganglioma and a variety of rare benign and malignant lesions at this site.


Asunto(s)
Adenoma/patología , Neoplasias del Oído/patología , Oído Medio/patología , Tumores Neuroendocrinos/patología , Adulto , Biomarcadores de Tumor/análisis , Femenino , Humanos , Inmunohistoquímica , Proteínas con Homeodominio LIM/biosíntesis , Masculino , Persona de Mediana Edad , Factores de Transcripción/biosíntesis
13.
Stem Cells ; 32(7): 1843-54, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24578274

RESUMEN

The LIM-homeobox transcription factor islet-1 (ISL1) has been proposed to mark a cardiovascular progenitor cell lineage that gives rise to cardiomyocytes, endothelial cells, and smooth muscle cells. The aim of this study was to investigate whether forced expression of ISL1 in human mesenchymal stem cells (hMSCs) influenced the differentiation capacity and angiogenic properties of hMSCs. The lentiviral vector, EF1α-ISL1, was constructed using the Multisite Gateway System and used to transduce hMSCs. We found that ISL1 overexpression did not alter the proliferation, migration, or survival of hMSCs or affect their ability to differentiate into osteoblasts, adipocytes, cardiomyocytes, or endotheliocytes. However, ISL1-hMSCs differentiated into smooth muscle cells more efficiently than control hMSCs. Furthermore, conditioned medium from ISL1-hMSCs greatly enhanced the survival, migration, and tube-formation ability of human umbilical vein endothelial cells (HUVECs) in vitro. In vivo angiogenesis assays also showed much more vascular-like structures in the group cotransplanted with ISL1-hMSCs and HUVECs than in the group cotransplanted with control hMSCs and HUVECs. Quantitative RT-PCR and antibody arrays detected monocyte chemoattractant protein-3 (MCP3) at a higher level in conditioned medium from ISL1-hMSCs cultures than in conditioned medium from control hMSCs. Neutralization assays showed that addition of an anti-MCP3 antibody to ISL1-hMSCs-conditioned medium efficiently abolished the angiogenesis-promoting effect of ISL1-hMSCs. Our data suggest that overexpression of ISL1 in hMSCs promotes angiogenesis in vitro and in vivo through increasing secretion of paracrine factors, smooth muscle differentiation ability, and enhancing the survival of HUVECs.


Asunto(s)
Quimiocina CCL7/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Proteínas con Homeodominio LIM/genética , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica , Factores de Transcripción/genética , Animales , Diferenciación Celular , Células Cultivadas , Quimiocina CCL7/genética , Expresión Génica , Humanos , Proteínas con Homeodominio LIM/metabolismo , Ratones SCID , Miocitos del Músculo Liso/fisiología , Factores de Transcripción/metabolismo
14.
Exp Eye Res ; 138: 22-31, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26122047

RESUMEN

The LIM-homeodomain transcription factor Islet-1 (Isl1) has been widely used as a marker of different subtypes of neurons in the developing and mature retina of vertebrates. During retinal neurogenesis, early Isl1 expression is detected in the nuclei of neuroblasts that give rise to ganglion, amacrine, bipolar, and horizontal cells. In the mature retina, Isl1 expression is restricted to the nuclei of ganglion cells, cholinergic amacrine cells, ON-bipolar cells, and subpopulations of horizontal cells. Recent studies have explored the functional mechanisms of Isl1 during specification and differentiation of these retinal cell types. Thus, conditional inactivation of Isl1 in the developing mouse retina disrupts retinal function, and also results in optic nerve hypoplasia, marked reductions in mature ganglion, amacrine, and bipolar cells, and a substantial increase in horizontal cells. Furthermore, conditional knockout shows delayed ganglion cell axon growth, ganglion cell axon guidance error, and ganglion cell nerve fiber defasciculation. These data together suggest a possible role for Isl1 in the early differentiation and maintenance of different vertebrate retinal cell types. This review examines whether the expression pattern of Isl1 during vertebrate retinal development is conserved across vertebrate species, and discusses current understanding of the developmental functions of Isl1 in retinogenesis.


Asunto(s)
Biomarcadores/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Neurogénesis/fisiología , Retina/embriología , Neuronas Retinianas/fisiología , Factores de Transcripción/metabolismo , Animales , Ojo/embriología , Humanos , Retina/fisiología
15.
Dev Biol ; 380(2): 286-98, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23648511

RESUMEN

Islet-1 is a LIM-Homeodomain transcription factor with important functions for the development of distinct neuronal and non-neuronal cell populations. We show here that Islet-1 acts genetically downstream of Phox2B in cells of the sympathoadrenal cell lineage and that the development of sympathetic neurons and chromaffin cells is impaired in mouse embryos with a conditional deletion of Islet-1 controlled by the wnt1 promotor. Islet-1 is not essential for the initial differentiation of sympathoadrenal cells, as indicated by the correct expression of pan-neuronal and catecholaminergic subtype specific genes in primary sympathetic ganglia of Islet-1 deficient mouse embryos. However, our data indicate that the subsequent survival of sympathetic neuron precursors and their differentiation towards TrkA expressing neurons depends on Islet-1 function. In contrast to spinal sensory neurons, sympathetic neurons of Islet-1 deficient mice did not display ectopic expression of genes normally present in the CNS. In Islet-1 deficient mouse embryos the numbers of chromaffin cells were only mildly reduced, in contrast to that of sympathetic neurons, but the initiation of the adrenaline synthesizing enzyme PNMT was abrogated and the expression level of chromogranin A was diminished. Microarray analysis revealed that developing chromaffin cells of Islet-1 deficient mice displayed normal expression levels of TH, DBH and the transcription factors Phox2B, Mash-1, Hand2, Gata3 and Insm1, but the expression levels of the transcription factors Gata2 and Hand1, and AP-2ß were significantly reduced. Together our data indicate that Islet-1 is not essentially required for the initial differentiation of sympathoadrenal cells, but has an important function for the correct subsequent development of sympathetic neurons and chromaffin cells.


Asunto(s)
Diferenciación Celular , Células Cromafines/citología , Ganglios Simpáticos/citología , Proteínas con Homeodominio LIM/fisiología , Factores de Transcripción/fisiología , Animales , Linaje de la Célula , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-ret/análisis , Receptor trkA/análisis
16.
Dev Biol ; 383(2): 253-63, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24055866

RESUMEN

Cardiovascular disease is a global scourge to society, with novel therapeutic approaches required in order to alleviate the suffering caused by sustained cardiac damage. MicroRNAs (miRNAs) are being touted as one such approach in the fight against heart disease, acting as possible post-transcriptional molecular triggers responsible for invoking cardiac regeneration. To further ones understanding of miRNAs and cardiac regeneration, it is prudent to learn from organisms that can intrinsically regenerate their hearts following injury. Using the red-spotted newt, an adult chordate capable of cardiac regeneration, we decided to delve deeper into the role miRNAs play during this process. RNA isolated from regenerating newt heart samples, was used in a microarray screen, to identify significantly expressed candidate miRNAs during newt cardiac regeneration. We performed quantitative qPCR analysis on several conserved miRNAs and found one in particular, miR-128, to be significantly elevated when cardiac hyperplasia is at its peak following injury. In-situ hybridisation techniques revealed a localised expression pattern for miR-128 in the cardiomyocytes and non-cardiomyocytes in close proximity to the regeneration zone and in vivo knockdown studies revealed a regulatory role for miR-128 in proliferating non-cardiomyocyte populations and extracellular matrix deposition. Finally, 3'UTR reporter assays revealed Islet1 as a biological target for miR-128, which was confirmed further through in vivo Islet1 transcriptional and translational expression analysis in regenerating newt hearts. From these studies we conclude that miR-128 regulates both cardiac hyperplasia and Islet1 expression during newt heart regeneration and that this information could be translated into future mammalian cardiac studies.


Asunto(s)
Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Proteínas con Homeodominio LIM/genética , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Regeneración/genética , Factores de Transcripción/genética , Animales , Secuencia de Bases , Regulación hacia Abajo , Fibrina/metabolismo , Hiperplasia , Proteínas con Homeodominio LIM/metabolismo , MicroARNs/genética , Datos de Secuencia Molecular , Miocardio/metabolismo , Miocardio/patología , Transporte de ARN/genética , Salamandridae , Factores de Transcripción/metabolismo , Transcripción Genética
17.
Am J Physiol Gastrointest Liver Physiol ; 307(10): G979-91, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25214396

RESUMEN

Enteroendocrine cells secrete over a dozen different hormones responsible for coordinating digestion, absorption, metabolism, and gut motility. Loss of enteroendocrine cells is a known cause of severe congenital diarrhea. Furthermore, enteroendocrine cells regulate glucose metabolism, with the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) playing critical roles in stimulating insulin release by pancreatic ß-cells. Islet1 (Isl1) is a LIM-homeodomain transcription factor expressed specifically in an array of intestinal endocrine cells, including incretin-expressing cells. To examine the impact of intestinal Isl1 on glycemic control, we set out to explore the role of intestinal Isl1 in hormone cell specification and organismal physiology. Mice with intestinal epithelial-specific ablation of Isl1 were obtained by crossing Villin-Cre transgenic animals with mice harboring a Isl1(loxP) allele (Isl1(int) model). Gene ablation of Isl1 in the intestine results in loss of GLP-1, GIP, cholecystokinin (CCK), and somatostatin-expressing cells and an increase in 5-HT (serotonin)-producing cells, while the chromogranin A population was unchanged. This dramatic change in hormonal milieu results in animals with lipid malabsorption and females smaller than their littermate controls. Interestingly, when challenged with oral, not intraperitoneal glucose, the Isl-1 intestinal-deficient animals (Isl1(int)) display impaired glucose tolerance, indicating loss of the incretin effect. Thus the Isl1(int) model confirms that intestinal biology is essential for organism physiology in glycemic control and susceptibility to diabetes.


Asunto(s)
Glucemia/metabolismo , Células Enteroendocrinas/metabolismo , Trastornos del Metabolismo de la Glucosa/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Proteínas con Homeodominio LIM/deficiencia , Factores de Transcripción/deficiencia , Factores de Edad , Animales , Animales Recién Nacidos , Biomarcadores/sangre , Colecistoquinina/metabolismo , Cromogranina A/metabolismo , Diarrea/genética , Diarrea/metabolismo , Grasas de la Dieta/metabolismo , Células Enteroendocrinas/patología , Femenino , Polipéptido Inhibidor Gástrico/metabolismo , Gastrinas/metabolismo , Genotipo , Ghrelina/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Trastornos del Metabolismo de la Glucosa/sangre , Trastornos del Metabolismo de la Glucosa/genética , Prueba de Tolerancia a la Glucosa , Integrasas/genética , Absorción Intestinal , Mucosa Intestinal/patología , Intestino Delgado/patología , Proteínas con Homeodominio LIM/genética , Síndromes de Malabsorción/genética , Síndromes de Malabsorción/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Fenotipo , Serotonina/metabolismo , Somatostatina/metabolismo , Factores de Transcripción/genética , Aumento de Peso
18.
Stem Cells Dev ; 33(17-18): 496-504, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38943285

RESUMEN

Prostaglandin E2 (PGE2) has recently gained attention in the field of regenerative medicine because of the beneficial effects of this molecule on stem cell proliferation and migration. Furthermore, PGE2 has the ability to mitigate immune rejection and fibrosis. In the colon and kidney, PGE2 induces YAP1, a transcription factor critical for cardiac regeneration. Establishing a similar connection in stem cells that can be transplanted in the heart could lead to the development of more effective therapeutics. In this report, we identify the effects of PGE2 on neonatal Islet-1+ stem cells. These stem cells synthesize PGE2, which functions by stimulating the transcription of the extracellular matrix protein Agrin. Agrin upregulates YAP1. Consequently, both YAP1 and Agrin are induced by PGE2 treatment. Our study shows that PGE2 upregulated the expression of both YAP1 and Agrin in Islet-1+ stem cells through the EP4 receptor and stimulated proliferation using the same mechanisms. PGE2 administration further elevated the expression of stemness markers and the matrix metalloproteinase MMP9, a key regulator of remodeling in the extracellular matrix post-injury. The expression of PGE2 in neonatal Islet-1+ cells is a factor which contributes to improving the functional efficacy of these cells for cardiac repair.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Agrina , Proliferación Celular , Dinoprostona , Subtipo EP4 de Receptores de Prostaglandina E , Células Madre , Proteínas Señalizadoras YAP , Proteínas Señalizadoras YAP/metabolismo , Animales , Dinoprostona/farmacología , Dinoprostona/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/genética , Agrina/metabolismo , Agrina/farmacología , Células Madre/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Ratones , Proliferación Celular/efectos de los fármacos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Animales Recién Nacidos , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética
19.
Cureus ; 16(5): e60136, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38864046

RESUMEN

Metaplastic thymoma is a rare biphasic thymic tumor with indolent behavior and recurrent YAP1::MAML2 gene rearrangement. Although the diagnosis of this tumor is usually straightforward based on hematoxylin and eosin (H&E) findings alone, cases with scant spindle-cell ("pseudosarcomatous stroma") components can be easily confused with more commonly occurring type A thymoma. We present a case of metaplastic thymoma with a sparse stroma-like spindle-cell component, discussing its histological and immunohistochemical hints and drawing attention to the visual similarity to type A thymoma. This is also the first published case of metaplastic thymoma with associated psoriasis.

20.
Pathol Res Pract ; 260: 155469, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39018925

RESUMEN

ISLET-1 (ISL1) is a LIM-homeodomain transcription factor. Selective ISL1 expression is shown in neuroendocrine, non-neuroendocrine, and some soft tissue tumors including desmoplastic small round cell tumor (DSRCT). We assessed the specificity of ISL1 (clone EP283, 1:500, Cell Marque) in 288 soft tissue tumors, which included 17 DSRCTs and other histologic mimics. Positive staining threshold for ISL1 was set to >10 % of neoplastic cell nuclei at moderate intensity. ISL1 IHC was positive in 15/16 (94 %) DSRCTs with 75 % showing diffuse (>50 %) expression. ISL1 was positive in 1/10 (10 %) Ewing sarcomas (EWS), 7/13 (54 %) alveolar rhabdomyosarcoma (RMS), 14/22 (63 %) embryonal RMS, 7/14 (50 %) synovial sarcomas, 15/16 (93 %) neuroblastoma, 1/5 (20 %) Wilms tumor, 2/4 (50 %) olfactory neuroblastoma, and all 9 Merkel cell carcinomas. Other tumors, including all CIC::DUX4 sarcomas, were negative except 3/27 leiomyosarcomas, and 1 each of angiosarcoma, myxoid liposarcomas, inflammatory myofibroblastic tumor, malignant peripheral nerve sheath tumor, tenosynovial giant cell tumor, dedifferentiated LPS, and 1 ectomesenchymoma. In summary, among the soft tissue tumors tested, ISL1 is a highly sensitive but moderately specific marker for DSRCT and may be useful to distinguish from round cell mimics including EWS and CIC::DUX4 sarcomas. The oncogenic role of ISL1 in these tumors warrants further investigation.


Asunto(s)
Biomarcadores de Tumor , Tumor Desmoplásico de Células Pequeñas Redondas , Proteínas con Homeodominio LIM , Neoplasias de los Tejidos Blandos , Factores de Transcripción , Humanos , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Tumor Desmoplásico de Células Pequeñas Redondas/patología , Tumor Desmoplásico de Células Pequeñas Redondas/diagnóstico , Tumor Desmoplásico de Células Pequeñas Redondas/metabolismo , Neoplasias de los Tejidos Blandos/patología , Neoplasias de los Tejidos Blandos/diagnóstico , Neoplasias de los Tejidos Blandos/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Proteínas con Homeodominio LIM/análisis , Factores de Transcripción/metabolismo , Factores de Transcripción/análisis , Sensibilidad y Especificidad , Inmunohistoquímica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA