Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 150(8)2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-37039233

RESUMEN

The gastrointestinal tract is innervated by an intrinsic neuronal network, known as the enteric nervous system (ENS), and by extrinsic axons arising from peripheral ganglia. The nerve of Remak (NoR) is an avian-specific sacral neural crest-derived ganglionated structure that extends from the cloaca to the proximal midgut and, similar to the pelvic plexus, provides extrinsic innervation to the distal intestine. The molecular mechanisms controlling extrinsic nerve fiber growth into the gut is unknown. In vertebrates, CXCR4, a cell-surface receptor for the CXCL12 chemokine, regulates migration of neural crest cells and axon pathfinding. We have employed chimeric tissue recombinations and organ culture assays to study the role of CXCR4 and CXCL12 molecules in the development of colorectal innervation. CXCR4 is specifically expressed in nerve fibers arising from the NoR and pelvic plexus, while CXCL12 is localized to the hindgut mesenchyme and enteric ganglia. Overexpression of CXCL12 results in significantly enhanced axonal projections to the gut from the NoR, while CXCR4 inhibition disrupts nerve fiber extension, supporting a previously unreported role for CXCR4 and CXCL12 signaling in extrinsic innervation of the colorectum.


Asunto(s)
Sistema Nervioso Entérico , Tracto Gastrointestinal , Animales , Tracto Gastrointestinal/inervación , Colon , Neuronas/fisiología , Transducción de Señal , Cresta Neural
2.
Development ; 150(5)2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36779913

RESUMEN

Enteric nervous system development relies on intestinal colonization by enteric neural crest-derived cells (ENCDCs). This is driven by a population of highly migratory and proliferative ENCDCs at the wavefront, but the molecular characteristics of these cells are unknown. ENCDCs from the wavefront and the trailing region were isolated and subjected to RNA-seq. Wavefront-ENCDCs were transcriptionally distinct from trailing ENCDCs, and temporal modelling confirmed their relative immaturity. This population of ENCDCs exhibited altered expression of ECM and cytoskeletal genes, consistent with a migratory phenotype. Unlike trailing ENCDCs, the wavefront lacked expression of genes related to neuronal or glial maturation. As wavefront ENCDC genes were associated with migration and developmental immaturity, the genes that remain expressed in later progenitor populations may be particularly pertinent to understanding the maintenance of ENCDC progenitor characteristics. Dusp6 expression was specifically upregulated at the wavefront. Inhibiting DUSP6 activity prevented wavefront colonization of the hindgut, and inhibited the migratory ability of post-colonized ENCDCs from midgut and postnatal neurospheres. These effects were reversed by simultaneous inhibition of ERK signaling, indicating that DUSP6-mediated ERK inhibition is required for ENCDC migration in mouse and chick.


Asunto(s)
Sistema Nervioso Entérico , Ratones , Animales , Cresta Neural/metabolismo , Transcriptoma , Movimiento Celular/fisiología , Intestinos
3.
Development ; 148(22)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34792104

RESUMEN

The enteric nervous system (ENS), which is derived from enteric neural crest cells (ENCCs), represents the neuronal innervation of the intestine. Compromised ENCC migration can lead to Hirschsprung disease, which is characterized by an aganglionic distal bowel. During the craniocaudal migration of ENCCs along the gut, we find that their proliferation is greatest as the ENCC wavefront passes through the ceca, a pair of pouches at the midgut-hindgut junction in avian intestine. Removal of the ceca leads to hindgut aganglionosis, suggesting that they are required for ENS development. Comparative transcriptome profiling of the cecal buds compared with the interceca region shows that the non-canonical Wnt signaling pathway is preferentially expressed within the ceca. Specifically, WNT11 is highly expressed, as confirmed by RNA in situ hybridization, leading us to hypothesize that cecal expression of WNT11 is important for ENCC colonization of the hindgut. Organ cultures using embryonic day 6 avian intestine show that WNT11 inhibits enteric neuronal differentiation. These results reveal an essential role for the ceca during hindgut ENS formation and highlight an important function for non-canonical Wnt signaling in regulating ENCC differentiation.


Asunto(s)
Sistema Nervioso Entérico/metabolismo , Cresta Neural/metabolismo , Neuronas/metabolismo , Proteínas Wnt/genética , Animales , Diferenciación Celular/genética , Movimiento Celular/genética , Embrión de Pollo , Pollos/genética , Pollos/crecimiento & desarrollo , Sistema Digestivo/crecimiento & desarrollo , Sistema Digestivo/metabolismo , Sistema Nervioso Entérico/crecimiento & desarrollo , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/patología , Humanos , Intestinos/inervación , Cresta Neural/citología , ARN/genética , RNA-Seq , Transcriptoma/genética , Vía de Señalización Wnt/genética
4.
J Anat ; 244(1): 120-132, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37626442

RESUMEN

Myocardial sleeve around human pulmonary veins plays a critical role in the pathomechanism of atrial fibrillation. Besides the well-known arrhythmogenicity of these veins, there is evidence that myocardial extensions into caval veins and coronary sinus may exhibit similar features. However, studies investigating histologic properties of these structures are limited. We aimed to investigate the immunoreactivity of myocardial sleeves for intermediate filament desmin, which was reported to be more abundant in Purkinje fibers than in ventricular working cardiomyocytes. Sections of 16 human (15 adult and 1 fetal) hearts were investigated. Specimens of atrial and ventricular myocardium, sinoatrial and atrioventricular nodes, pulmonary veins, superior caval vein and coronary sinus were stained with anti-desmin monoclonal antibody. Intensity of desmin immunoreactivity in different areas was quantified by the ImageJ program. Strong desmin labeling was detected at the pacemaker and conduction system as well as in the myocardial sleeves around pulmonary veins, superior caval vein, and coronary sinus of adult hearts irrespective of sex, age, and medical history. In the fetal heart, prominent desmin labeling was observed at the sinoatrial nodal region and in the myocardial extensions around the superior caval vein. Contrarily, atrial and ventricular working myocardium exhibited low desmin immunoreactivity in both adults and fetuses. These differences were confirmed by immunohistochemical quantitative analysis. In conclusion, this study indicates that desmin is abundant in the conduction system and venous myocardial sleeves of human hearts.


Asunto(s)
Seno Coronario , Desmina , Venas Pulmonares , Adulto , Humanos , Miocardio/patología , Miocitos Cardíacos , Venas Pulmonares/patología , Vena Cava Superior
5.
Development ; 147(21)2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-32994173

RESUMEN

Appropriately balanced RET signaling is of crucial importance during embryonic neural crest cell migration, proliferation and differentiation. RET deficiency, for example, leads to intestinal aganglionosis (Hirschsprung disease), whereas overactive RET can lead to multiple endocrine neoplasia (MEN) syndromes. Some RET mutations are associated with both intestinal aganglionosis and MEN-associated tumors. This seemingly paradoxical occurrence has led to speculation of a 'Janus mutation' in RET that causes overactivation or impairment of RET activity depending on the cellular context. Using an intestinal catenary culture system to test the effects of GDNF-mediated RET activation, we demonstrate the concurrent development of distal colonic aganglionosis and intestinal ganglioneuromas. Interestingly, the tumors induced by GDNF stimulation contain enteric neuronal progenitors capable of reconstituting an enteric nervous system when transplanted into a normal developmental environment. These results suggest that a Janus mutation may not be required to explain co-existing Hirschsprung disease and MEN-associated tumors, but rather that RET overstimulation alone is enough to cause both phenotypes. The results also suggest that reprogramming tumor cells toward non-pathological fates may represent a possible therapeutic avenue for MEN-associated neoplasms.


Asunto(s)
Ganglioneuroma/patología , Enfermedad de Hirschsprung/patología , Intestinos/patología , Proteínas Proto-Oncogénicas c-ret/metabolismo , Animales , Agregación Celular , Diferenciación Celular , Embrión de Pollo , Pollos , Sistema Nervioso Entérico/patología , Ganglioneuroma/metabolismo , Factores Neurotróficos Derivados de la Línea Celular Glial/metabolismo , Enfermedad de Hirschsprung/metabolismo , Ratones Endogámicos C57BL , Cresta Neural/patología , Neuronas/metabolismo , Neuronas/patología , Nervio Vago/patología
6.
Int J Mol Sci ; 24(21)2023 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-37958648

RESUMEN

The enteric nervous system (ENS) is principally derived from vagal neural crest cells that migrate caudally along the entire length of the gastrointestinal tract, giving rise to neurons and glial cells in two ganglionated plexuses. Incomplete migration of enteric neural crest-derived cells (ENCDC) leads to Hirschsprung disease, a congenital disorder characterized by the absence of enteric ganglia along variable lengths of the colorectum. Our previous work strongly supported the essential role of the avian ceca, present at the junction of the midgut and hindgut, in hindgut ENS development, since ablation of the cecal buds led to incomplete ENCDC colonization of the hindgut. In situ hybridization shows bone morphogenetic protein-4 (BMP4) is highly expressed in the cecal mesenchyme, leading us to hypothesize that cecal BMP4 is required for hindgut ENS development. To test this, we modulated BMP4 activity using embryonic intestinal organ culture techniques and retroviral infection. We show that overexpression or inhibition of BMP4 in the ceca disrupts hindgut ENS development, with GDNF playing an important regulatory role. Our results suggest that these two important signaling pathways are required for normal ENCDC migration and enteric ganglion formation in the developing hindgut ENS.


Asunto(s)
Neoplasias Colorrectales , Sistema Nervioso Entérico , Humanos , Transducción de Señal/fisiología , Diferenciación Celular/fisiología , Sistema Nervioso Entérico/metabolismo , Movimiento Celular/fisiología , Neoplasias Colorrectales/metabolismo , Cresta Neural/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo
7.
J Mol Cell Cardiol ; 165: 19-30, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34959166

RESUMEN

BACKGROUND: Cardiac cell lines and primary cells are widely used in cardiovascular research. Despite increasing number of publications using these models, comparative characterization of these cell lines has not been performed, therefore, their limitations are undetermined. We aimed to compare cardiac cell lines to primary cardiomyocytes and to mature cardiac tissues in a systematic manner. METHODS AND RESULTS: Cardiac cell lines (H9C2, AC16, HL-1) were differentiated with widely used protocols. Left ventricular tissue, neonatal primary cardiomyocytes, and human induced pluripotent stem cell-derived cardiomyocytes served as reference tissue or cells. RNA expression of cardiac markers (e.g. Tnnt2, Ryr2) was markedly lower in cell lines compared to references. Differentiation induced increase in cardiac- and decrease in embryonic markers however, the overall transcriptomic profile and annotation to relevant biological processes showed consistently less pronounced cardiac phenotype in all cell lines in comparison to the corresponding references. Immunocytochemistry confirmed low expressions of structural protein sarcomeric alpha-actinin, troponin I and caveolin-3 in cell lines. Susceptibility of cell lines to sI/R injury in terms of viability as well as mitochondrial polarization differed from the primary cells irrespective of their degree of differentiation. CONCLUSION: Expression patterns of cardiomyocyte markers and whole transcriptomic profile, as well as response to sI/R, and to hypertrophic stimuli indicate low-to-moderate similarity of cell lines to primary cells/cardiac tissues regardless their differentiation. Low resemblance of cell lines to mature adult cardiac tissue limits their potential use. Low translational value should be taken into account while choosing a particular cell line to model cardiomyocytes.


Asunto(s)
Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Animales , Biomarcadores/metabolismo , Diferenciación Celular/genética , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Fenotipo , Transcriptoma
8.
Stem Cells ; 39(9): 1236-1252, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33938072

RESUMEN

Interplay between embryonic enteric neural stem cells (ENSCs) and enteric mesenchymal cells (EMCs) in the embryonic gut is essential for normal development of the enteric nervous system. Disruption of these interactions underlies the pathogenesis of intestinal aganglionosis in Hirschsprung disease (HSCR). ENSC therapy has been proposed as a possible treatment for HSCR, but whether the survival and development of postnatal-derived ENSCs similarly rely on signals from the mesenchymal environment is unknown and has important implications for developing protocols to expand ENSCs for cell transplantation therapy. Enteric neural crest-derived cells (ENCDCs) and EMCs were cultured from the small intestine of Wnt1-Rosa26-tdTomato mice. EMCs promoted the expansion of ENCDCs 9.5-fold by inducing ENSC properties, including expression of Nes, Sox10, Sox2, and Ngfr. EMCs enhanced the neurosphere-forming ability of ENCDCs, and this persisted after withdrawal of the EMCs. These effects were mediated by paracrine factors and several ligands known to support neural stem cells were identified in EMCs. Using the optimized expansion procedures, neurospheres were generated from small intestine of the Ednrb-/- mouse model of HSCR. These ENSCs had similar proliferative and migratory capacity to Ednrb+/+ ENSCs, albeit neurospheres contained fewer neurons. ENSCs derived from Ednrb-/- mice generated functional neurons with similar calcium responses to Ednrb+/+ ENSCs and survived after transplantation into the aganglionic colon of Ednrb-/- recipients. EMCs act as supporting cells to ENSCs postnatally via an array of synergistically acting paracrine signaling factors. These properties can be leveraged to expand autologous ENSCs from patients with HSCR mutations for therapeutic application.


Asunto(s)
Sistema Nervioso Entérico , Enfermedad de Hirschsprung , Células-Madre Neurales , Animales , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/metabolismo , Enfermedad de Hirschsprung/terapia , Humanos , Intestino Delgado/metabolismo , Ratones , Ratones Endogámicos C57BL , Cresta Neural/metabolismo , Células-Madre Neurales/metabolismo
9.
J Immunol ; 204(1): 23-36, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31767783

RESUMEN

The cellular homeostasis of lymphoid tissues is determined by the continuous interactions of mobile hematopoietic cells within specialized microenvironments created by sessile stromal cells. In contrast to the lymph nodes and mucosal lymphoid tissues with well-defined entry and exit routes, the movement of leukocytes in the peritoneal cavity is largely unknown. In this study, we report that, in addition to the omental milky spots and fat-associated lymphoid clusters, in mice, the serous surface of the mesenteric adipose streaks contains lymphocyte-rich organoids comprised of a highly compacted leaf-like part connected to the adipose tissue that can also efficiently bind B cells and high-grade B cell lymphoma (diffuse large B cell lymphoma) cells. Denoted as foliate lymphoid aggregates (FLAgs), these structures show incomplete T/B segregation and a partially differentiated stromal architecture. LYVE-1-positive macrophages covering FLAgs efficiently bind i.p. injected normal B cells as well as different types of diffuse large B cell lymphoma cells. Within FLAgs, the lymphocytes compartmentalize according to their chemokine receptor pattern and subsequently migrate toward the mesenteric lymph nodes via the mesenteric lymphatic capillaries. The blood supply of FLAgs includes short vascular segments displaying peripheral lymph node addressin, and the extravasation of lymphocytes to the omental and mesenteric adipose tissues is partly mediated by L-selectin. The appearance of i.p. injected cells in mesenteric lymph nodes suggests that the mesentery-associated lymphatics may also collect leukocytes from the fat-associated lymphoid clusters and FLAgs, thus combining the mucosal and serous exit of mobile leukocytes and increasing the range of drainage sites for the peritoneal expansion of lymphoid malignancies.


Asunto(s)
Linfocitos B/inmunología , Movimiento Celular/inmunología , Linfoma de Células B Grandes Difuso/patología , Mesenterio/citología , Cavidad Peritoneal/citología , Animales , Línea Celular , Selectina L/metabolismo , Leucocitos/inmunología , Ganglios Linfáticos/citología , Vasos Linfáticos/metabolismo , Linfoma de Células B Grandes Difuso/inmunología , Macrófagos/inmunología , Proteínas de Transporte de Membrana/metabolismo , Mesenterio/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Microambiente Tumoral/inmunología
10.
Development ; 145(9)2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29678817

RESUMEN

The enteric nervous system (ENS) arises from neural crest cells that migrate, proliferate, and differentiate into enteric neurons and glia within the intestinal wall. Many extracellular matrix (ECM) components are present in the embryonic gut, but their role in regulating ENS development is largely unknown. Here, we identify heparan sulfate proteoglycan proteins, including collagen XVIII (Col18) and agrin, as important regulators of enteric neural crest-derived cell (ENCDC) development. In developing avian hindgut, Col18 is expressed at the ENCDC wavefront, while agrin expression occurs later. Both proteins are normally present around enteric ganglia, but are absent in aganglionic gut. Using chick-mouse intestinal chimeras and enteric neurospheres, we show that vagal- and sacral-derived ENCDCs from both species secrete Col18 and agrin. Whereas glia express Col18 and agrin, enteric neurons only express the latter. Functional studies demonstrate that Col18 is permissive whereas agrin is strongly inhibitory to ENCDC migration, consistent with the timing of their expression during ENS development. We conclude that ENCDCs govern their own migration by actively remodeling their microenvironment through secretion of ECM proteins.


Asunto(s)
Agrina/metabolismo , Proteínas Aviares/metabolismo , Pollos/metabolismo , Colágeno/metabolismo , Sistema Digestivo , Cresta Neural/embriología , Nicho de Células Madre/fisiología , Agrina/genética , Animales , Proteínas Aviares/genética , Movimiento Celular/fisiología , Embrión de Pollo , Pollos/genética , Colágeno/genética , Sistema Digestivo/citología , Sistema Digestivo/embriología , Sistema Digestivo/inervación , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , Cresta Neural/citología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo
11.
Dev Biol ; 446(1): 34-42, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30529057

RESUMEN

Cells of the vagal neural crest (NC) form most of the enteric nervous system (ENS) by a colonising wave in the embryonic gut, with high cell proliferation and differentiation. Enteric neuropathies have an ENS deficit and cell replacement has been suggested as therapy. This would be performed post-natally, which raises the question of whether the ENS cell population retains its initial ENS-forming potential with age. We tested this on the avian model in organ culture in vitro (3 days) using recipient aneural chick midgut/hindgut combined with ENS-donor quail midgut or hindgut of ages QE5 to QE10. ENS cells from young donor tissues (≤ QE6) avidly colonised the aneural recipient, but this capacity dropped rapidly 2-3 days after the transit of the ENS cell wavefront. This loss in capability was autonomous to the ENS population since a similar decline was observed in ENS cells isolated by HNK1 FACS. Using QE5, 6, 8 and 10 midgut donors and extending the time of assay to 8 days in chorio-allantoic membrane grafts did not produce 'catch up' colonisation. NC-derived cells were counted in dissociated quail embryo gut and in transverse sections of chick embryo gut using NC, neuron and glial marker antibodies. This showed that the decline in ENS-forming ability correlated with a decrease in proportion of ENS cells lacking both neuronal and glial differentiation markers, but there were still large numbers of such cells even at stages with low colonisation ability. Moreover, ENS cells in small numbers from young donors were far superior in colonisation ability to larger numbers of apparently undifferentiated cells from older donors. This suggests that the decline of ENS-forming ability has both quantitative and qualitative aspects. In this case, ENS cells for cell therapies should aim to replicate the embryonic ENS stage rather than using post-natal ENS stem/progenitor cells.


Asunto(s)
Sistema Digestivo/embriología , Sistema Nervioso Entérico/embriología , Intestino Delgado/embriología , Cresta Neural/embriología , Animales , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Embrión de Pollo , Pollos , Membrana Corioalantoides/trasplante , Coturnix , Sistema Digestivo/citología , Sistema Digestivo/metabolismo , Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/metabolismo , Intestino Delgado/citología , Intestino Delgado/inervación , Cresta Neural/citología , Cresta Neural/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Técnicas de Cultivo de Órganos
12.
Semin Cell Dev Biol ; 66: 94-106, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28087321

RESUMEN

The enteric nervous system (ENS) is comprised of a network of neurons and glial cells that are responsible for coordinating many aspects of gastrointestinal (GI) function. These cells arise from the neural crest, migrate to the gut, and then continue their journey to colonize the entire length of the GI tract. Our understanding of the molecular and cellular events that regulate these processes has advanced significantly over the past several decades, in large part facilitated by the use of rodents, avians, and zebrafish as model systems to dissect the signals and pathways involved. These studies have highlighted the highly dynamic nature of ENS development and the importance of carefully balancing migration, proliferation, and differentiation of enteric neural crest-derived cells (ENCCs). Proliferation, in particular, is critically important as it drives cell density and speed of migration, both of which are important for ensuring complete colonization of the gut. However, proliferation must be tempered by differentiation among cells that have reached their final destination and are ready to send axonal extensions, connect to effector cells, and begin to produce neurotransmitters or other signals. Abnormalities in the normal processes guiding ENCC development can lead to failure of ENS formation, as occurs in Hirschsprung disease, in which the distal intestine remains aganglionic. This review summarizes our current understanding of the factors involved in early development of the ENS and discusses areas in need of further investigation.


Asunto(s)
Sistema Nervioso Entérico/crecimiento & desarrollo , Tracto Gastrointestinal/crecimiento & desarrollo , Humanos
13.
Development ; 143(2): 264-75, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26674309

RESUMEN

The enteric nervous system (ENS) develops from neural crest cells that migrate along the intestine, differentiate into neurons and glia, and pattern into two plexuses within the gut wall. Inductive interactions between epithelium and mesenchyme regulate gut development, but the influence of these interactions on ENS development is unknown. Epithelial-mesenchymal recombinations were constructed using avian hindgut mesenchyme and non-intestinal epithelium from the bursa of Fabricius. These recombinations led to abnormally large and ectopically positioned ganglia. We hypothesized that sonic hedgehog (Shh), a secreted intestinal epithelial protein not expressed in the bursa, mediates this effect. Inhibition of Shh signaling, by addition of cyclopamine or a function-blocking antibody, resulted in large, ectopic ganglia adjacent to the epithelium. Shh overexpression, achieved in ovo using Shh-encoding retrovirus and in organ culture using recombinant protein, led to intestinal aganglionosis. Shh strongly induced the expression of versican and collagen type IX, whereas cyclopamine reduced expression of these chondroitin sulfate proteoglycans that are known to be inhibitory to neural crest cell migration. Shh also inhibited enteric neural crest-derived cell (ENCC) proliferation, promoted neuronal differentiation, and reduced expression of Gdnf, a key regulator of ENS formation. Ptc1 and Ptc2 were not expressed by ENCCs, and migration of isolated ENCCs was not inhibited by Shh protein. These results suggest that epithelial-derived Shh acts indirectly on the developing ENS by regulating the composition of the intestinal microenvironment.


Asunto(s)
Sistema Nervioso Entérico/metabolismo , Matriz Extracelular/metabolismo , Proteínas Hedgehog/metabolismo , Animales , Apoptosis/fisiología , Movimiento Celular , Pollos , Proteínas Hedgehog/genética , Inmunohistoquímica , Hibridación in Situ , Ratones , Receptores Patched , Receptor Patched-1 , Codorniz , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal/fisiología
14.
J Anat ; 233(4): 401-410, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30022489

RESUMEN

The enteric nervous system shares embryological, morphological, neurochemical, and functional features with the central nervous system. In addition to neurons and glia, the CNS includes a third component, microglia, which are functionally and immunophenotypically similar to macrophages, but a similar cell type has not previously been identified in enteric ganglia. In this study we identify a population of macrophages in the enteric ganglia, intermingling with the neurons and glia. These intraganglionic macrophages (IMs) are highly ramified and express the hematopoietic marker CD45, major histocompatibility complex (MHC) class II antigen, and chB6, a marker specific for B cells and microglia in avians. These IMs do not express antigens typically associated with T cells or dendritic cells. The CD45+ /ChB6+ /MHCII+ signature supports a hematopoietic origin and this was confirmed using intestinal chimeras in GFP-transgenic chick embryos. The presence of green fluorescent protein positive (GFP+) /CD45+ cells in the intestinal graft ENS confirms that IMs residing within enteric ganglia have a hematopoietic origin. IMs are also found in the ganglia of CSF1RGFP chicken and CX3CR1GFP mice. Based on the expression pattern and location of IMs in avians and rodents, we conclude that they represent a novel non-neural crest-derived microglia-like cell population within the enteric ganglia.


Asunto(s)
Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/inmunología , Macrófagos/citología , Macrófagos/inmunología , Animales , Embrión de Pollo , Ganglios/citología , Ganglios/inmunología , Neuroinmunomodulación/fisiología
15.
Dev Biol ; 409(2): 473-88, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26586201

RESUMEN

Hirschsprung Disease (HSCR) is a potentially deadly birth defect characterized by the absence of the enteric nervous system (ENS) in distal bowel. Although HSCR has clear genetic causes, no HSCR-associated mutation is 100% penetrant, suggesting gene-gene and gene-environment interactions determine HSCR occurrence. To test the hypothesis that certain medicines might alter HSCR risk we treated zebrafish with medications commonly used during early human pregnancy and discovered that ibuprofen caused HSCR-like absence of enteric neurons in distal bowel. Using fetal CF-1 mouse gut slice cultures, we found that ibuprofen treated enteric neural crest-derived cells (ENCDC) had reduced migration, fewer lamellipodia and lower levels of active RAC1/CDC42. Additionally, inhibiting ROCK, a RHOA effector and known RAC1 antagonist, reversed ibuprofen effects on migrating mouse ENCDC in culture. Ibuprofen also inhibited colonization of Ret+/- mouse bowel by ENCDC in vivo and dramatically reduced bowel colonization by chick ENCDC in culture. Interestingly, ibuprofen did not affect ENCDC migration until after at least three hours of exposure. Furthermore, mice deficient in Ptgs1 (COX 1) and Ptgs2 (COX 2) had normal bowel colonization by ENCDC and normal ENCDC migration in vitro suggesting COX-independent effects. Consistent with selective and strain specific effects on ENCDC, ibuprofen did not affect migration of gut mesenchymal cells, NIH3T3, or WT C57BL/6 ENCDC, and did not affect dorsal root ganglion cell precursor migration in zebrafish. Thus, ibuprofen inhibits ENCDC migration in vitro and bowel colonization by ENCDC in vivo in zebrafish, mouse and chick, but there are cell type and strain specific responses. These data raise concern that ibuprofen may increase Hirschsprung disease risk in some genetically susceptible children.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Sistema Nervioso Entérico/citología , Ibuprofeno/farmacología , Intestinos/citología , Células-Madre Neurales/citología , Citoesqueleto de Actina/metabolismo , Animales , Caspasa 3/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Pollos , Ciclooxigenasa 1/deficiencia , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/deficiencia , Ciclooxigenasa 2/metabolismo , Activación Enzimática/efectos de los fármacos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Mesodermo/citología , Ratones , Modelos Biológicos , Células 3T3 NIH , Células-Madre Neurales/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , PPAR gamma/metabolismo , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Pez Cebra , Proteína de Unión al GTP rac1/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
16.
Dev Biol ; 417(2): 229-51, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27059883

RESUMEN

Over the last 20 years, there has been increasing focus on the development of novel stem cell based therapies for the treatment of disorders and diseases affecting the enteric nervous system (ENS) of the gastrointestinal tract (so-called enteric neuropathies). Here, the idea is that ENS progenitor/stem cells could be transplanted into the gut wall to replace the damaged or absent neurons and glia of the ENS. This White Paper sets out experts' views on the commonly used methods and approaches to identify, isolate, purify, expand and optimize ENS stem cells, transplant them into the bowel, and assess transplant success, including restoration of gut function. We also highlight obstacles that must be overcome in order to progress from successful preclinical studies in animal models to ENS stem cell therapies in the clinic.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Sistema Nervioso Entérico/patología , Tracto Gastrointestinal/patología , Enfermedad de Hirschsprung/terapia , Seudoobstrucción Intestinal/terapia , Células-Madre Neurales/trasplante , Trasplante de Células Madre , Animales , Modelos Animales de Enfermedad , Tracto Gastrointestinal/inervación , Guías como Asunto , Enfermedad de Hirschsprung/patología , Humanos , Seudoobstrucción Intestinal/patología
17.
Infect Immun ; 85(11)2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28784929

RESUMEN

Enteropathogenic Escherichia coli (EPEC) is a leading cause of severe intestinal disease and infant mortality in developing countries. Virulence is mediated by a type three secretion system (T3SS), causing the hallmark attaching and effacing (AE) lesions and actin-rich pedestal formation beneath the infecting bacteria on the apical surface of enterocytes. EPEC is a human-specific pathogen whose pathogenesis cannot be studied in animal models. We therefore established an EPEC infection model in human gut xenografts in SCID mice and used it to study the role of T3SS in the pathogenesis of the disease. Following EPEC O127:H6 strain E2348/69 infection, T3SS-dependent AE lesions and pedestals were demonstrated in all infected xenografts. We report here the development of T3SS-dependent intestinal thrombotic microangiopathy (iTMA) and ischemic enteritis in ∼50% of infected human gut xenografts. Using species-specific CD31 immunostaining, we showed that iTMA was limited to the larger human-mouse chimeric blood vessels, which are located between the muscularis mucosa and circular muscular layer of the human gut. These blood vessels were massively invaded by bacteria, which adhered to and formed pedestals on endothelial cells and aggregated with mouse neutrophils in the lumen. We conclude that endothelial infection, iTMA, and ischemic enteritis might be central mechanisms underlying severe EPEC-mediated disease.

18.
Cell Tissue Res ; 368(2): 353-370, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28353134

RESUMEN

Embryonic tissues contain highly ramified stellate-shaped cells expressing CD45 and MHC II antigens but their origin and immunophenotype are unknown. Using staged avian embryos and cell-type-specific antibodies, we establish a detailed spatiotemporal ontogeny of cells that express CD45, the earliest marker of hematopoietic stem cells in the chick. CD45 immunostaining marks three distinct embryonic cell populations: round, ramified and amoeboid cells. The round and ramified CD45+ cells appear first in yolk-sac blood islands before the onset of circulation. A subpopulation of round cells co-expresses the thrombocyte-specific CD51/CD61 antigen. Amoeboid cells express macrophage-specific antigens and frequently occur in regions of apoptosis. Ramified cells are distributed uniformly in the embryonic mesenchyme, colonize lymphoid and non-lymphoid organs and later express MHC II. To study the origin of CD45+ cells, 2-day-old chick embryos were ablated from the yolk sac before the establishment of circulation and incubated for 2-5 days. Large numbers of CD45+MHC II+ ramified cells differentiated in the yolk sac. Yolk-sac chimeras were generated by grafting embryos into GFP-expressing de-embryonated yolk sacs. GFP/CD45 co-expressing ramified and amoeboid cells colonized all organ primordia in the donor embryo. We also recombined GFP+ yolk sac with the bursa of Fabricius and found ramified GFP+CD45+ cells in the bursa where they differentiated into dendritic cells. Thus, CD45 cells are first present in the yolk sac during primitive hematopoiesis and then migrate from the extra-embryonic yolk sac to give rise to cells throughout all organ primordia, including dendritic cells in the bursa of Fabricius.


Asunto(s)
Bolsa de Fabricio/citología , Células Dendríticas/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Animales , Diferenciación Celular , Embrión de Pollo , Células Dendríticas/citología , Células Madre Hematopoyéticas , Linfocitos/citología , Células Mieloides/citología , Fenotipo , Saco Vitelino/citología , Saco Vitelino/metabolismo
19.
J Anat ; 230(4): 567-574, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28116763

RESUMEN

The development of the enteric nervous system (ENS) and intestinal smooth muscle occurs in a spatially and temporally correlated manner, but how they influence each other is unknown. In the developing mid-gut of the chick embryo, we find that α-smooth muscle actin expression, indicating early muscle differentiation, occurs after the arrival of migrating enteric neural crest-derived cells (ENCCs). In contrast, hindgut smooth muscle develops prior to ENCC arrival. Smooth muscle development is normal in experimentally aganglionic hindguts, suggesting that proper development and patterning of the muscle layers does not rely on the ENS. However, inhibiting early smooth muscle development severely disrupts ENS patterning without affecting ENCC proliferation or apoptosis. Our results demonstrate that early intestinal smooth muscle differentiation is required for patterning the developing ENS.


Asunto(s)
Sistema Nervioso Entérico/embriología , Intestinos/embriología , Músculo Liso/embriología , Animales , Embrión de Pollo , Pollos , Sistema Nervioso Entérico/anatomía & histología , Intestinos/anatomía & histología , Músculo Liso/anatomía & histología , Técnicas de Cultivo de Órganos
20.
Pediatr Res ; 81(5): 838-846, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28060794

RESUMEN

BACKGROUND: Enteric neural stem/progenitor cells (ENSCs) offer an innovative approach to treating Hirschsprung disease (HSCR) and other enteric neuropathies. However, postnatal-derived human ENSCs have not been thoroughly characterized and their behavior in the embryonic and postnatal intestinal environment is unknown. METHODS: ENSCs were isolated from the intestines of 25 patients undergoing bowel resection, including 7 children with HSCR. Neuronal differentiation and proliferation of ENSCs from submucosal and myenteric plexuses from patients with and without HSCR were characterized. ENSC migration and differentiation were studied following transplantation into embryonic chick neural crest, embryonic chick hindgut, and postnatal mouse aganglionic colon. RESULTS: The proliferative and neurogenic potential of ENSCs from HSCR intestine is equivalent to that of non-HSCR controls. Similarly, no difference was observed between myenteric- and submucosal-derived ENSCs. Postnatal ENSCs transplanted to embryonic neural crest pathways and to aneural hindgut migrate normally and differentiate into appropriate neural crest-derived cell types. ENSCs in postnatal mouse aganglionic colon differentiate into neurons and glia both ex vivo and in vivo. CONCLUSIONS: ENSCs isolated from the postnatal intestine of patients with and without HSCR can behave like embryonic neural crest-derived cells. These results support the feasibility of cell-based therapy for future treatment of neurointestinal disease.


Asunto(s)
Movimiento Celular , Proliferación Celular , Enfermedad de Hirschsprung/patología , Intestino Grueso/inervación , Plexo Mientérico/patología , Células-Madre Neurales/patología , Neurogénesis , Nicho de Células Madre , Plexo Submucoso/patología , Adolescente , Animales , Células Cultivadas , Embrión de Pollo , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Enfermedad de Hirschsprung/cirugía , Humanos , Lactante , Recién Nacido , Masculino , Ratones Endogámicos C57BL , Células-Madre Neurales/trasplante , Esferoides Celulares , Trasplante de Células Madre , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA