Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 262
Filter
1.
Proc Natl Acad Sci U S A ; 116(27): 13414-13423, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31196952

ABSTRACT

The molecular mechanisms regulating sympathetic innervation of the heart during embryogenesis and its importance for cardiac development and function remain to be fully elucidated. We generated mice in which conditional knockout (CKO) of the Hif1a gene encoding the transcription factor hypoxia-inducible factor 1α (HIF-1α) is mediated by an Islet1-Cre transgene expressed in the cardiac outflow tract, right ventricle and atrium, pharyngeal mesoderm, peripheral neurons, and hindlimbs. These Hif1aCKO mice demonstrate significantly decreased perinatal survival and impaired left ventricular function. The absence of HIF-1α impaired the survival and proliferation of preganglionic and postganglionic neurons of the sympathetic system, respectively. These defects resulted in hypoplasia of the sympathetic ganglion chain and decreased sympathetic innervation of the Hif1aCKO heart, which was associated with decreased cardiac contractility. The number of chromaffin cells in the adrenal medulla was also decreased, indicating a broad dependence on HIF-1α for development of the sympathetic nervous system.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Sympathetic Nervous System/growth & development , Adrenal Medulla/embryology , Adrenal Medulla/innervation , Animals , Chromaffin Cells , Coronary Vessel Anomalies/embryology , Coronary Vessels/embryology , Female , Ganglia, Sympathetic/embryology , Ganglia, Sympathetic/growth & development , Heart/embryology , Heart/innervation , Male , Mice , Mice, Knockout , Mice, Transgenic , Sympathetic Nervous System/enzymology
2.
Surg Radiol Anat ; 41(8): 973-976, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30820646

ABSTRACT

The spermatic ganglia are collections of sympathetic neuron cell bodies located within the cords of the infrarenal aortic plexus, positioned at the origin of the testicular arteries in males. During routine dissection of the aortic plexus at our institution, one specimen exhibited a second (accessory) testicular artery on the right side that coursed retrocaval. Histology was used to confirm the presence of an accessory right spermatic ganglion at the base of the accessory retrocaval testicular artery. Interestingly, the accessory spermatic ganglion was also supplied by its own right lumbar splanchnic nerve. This is the first case to describe the anatomy of an accessory spermatic ganglion in a specimen that exhibits an accessory testicular artery on the right side. This neurovascular variation is of interest to surgeons who aim to perform nerve-sparing retroperitoneal lymph node dissections for malignancy.


Subject(s)
Ganglia, Sympathetic/abnormalities , Lumbosacral Plexus/abnormalities , Testis/innervation , Aged , Anatomic Variation , Cadaver , Dissection , Ganglia, Sympathetic/embryology , Humans , Lumbosacral Plexus/embryology , Lymph Node Excision/methods , Male , Retroperitoneal Space/surgery , Testis/blood supply
3.
Anat Rec (Hoboken) ; 302(2): 278-287, 2019 02.
Article in English | MEDLINE | ID: mdl-30290083

ABSTRACT

Although the embryonic kidney's ascent is well established, the intermediate morphological changes that occur during the process are unclear. To evaluate the morphological events that accompany the kidney's ascent, we examined serial sagittal sections from 24 embryos at 5-7 weeks gestation. Six specimens had bilaterally ascending kidneys that were between the levels of the second to fifth lumbar vertebrae, and each kidney had a primitive renal cortex surrounding clusters of ampullae, which branched from the pelvis, and a dense tissue band that connected the renal cortex with the embryonic adrenal cortex or celiac ganglia, and there was no adipose capsule or renal artery. The tissue band contained abundant nerve twigs from the major splanchnic nerve; thus, it was conceivable that it was sufficiently rigid to support the length of the retroperitoneal tissue mass that included the embryonic adrenal cortex, celiac ganglia, and kidney. The lumbar vertebral body's height was much shorter than that of the ascending kidney. However, the lower vertebral column's curvature was often maintained, even when the kidneys had ascended. Therefore, vertebral column straightening was not the only factor required to drive the ascent. Together with the growth of the thorax and liver, the adrenal cortex, ganglia, and kidney appeared to change simultaneously at a position relative to the vertebrae. The renal artery established a connection to the renal cortex after the ascent. Evaluations of frontal sections from five additional specimens suggested that from its initial position, the kidney extended upwards between bilateral umbilical arteries. Anat Rec, 302:278-287, 2019. © 2018 The Authors. The Anatomical Record: Advances in Integrative Anatomy and Evolutionary Biology published by Wiley Periodicals, Inc. on behalf of Wiley-Liss, Inc.


Subject(s)
Fetal Development , Ganglia, Sympathetic/embryology , Kidney/embryology , Kidney/innervation , Lumbar Vertebrae/embryology , Renal Artery/embryology , Female , Ganglia, Sympathetic/anatomy & histology , Gestational Age , Humans , Kidney/anatomy & histology , Lumbar Vertebrae/anatomy & histology , Male , Pregnancy , Renal Artery/anatomy & histology
4.
Science ; 354(6314): 893-897, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27856909

ABSTRACT

A kinship between cranial and pelvic visceral nerves of vertebrates has been accepted for a century. Accordingly, sacral preganglionic neurons are considered parasympathetic, as are their targets in the pelvic ganglia that prominently control rectal, bladder, and genital functions. Here, we uncover 15 phenotypic and ontogenetic features that distinguish pre- and postganglionic neurons of the cranial parasympathetic outflow from those of the thoracolumbar sympathetic outflow in mice. By every single one, the sacral outflow is indistinguishable from the thoracolumbar outflow. Thus, the parasympathetic nervous system receives input from cranial nerves exclusively and the sympathetic nervous system from spinal nerves, thoracic to sacral inclusively. This simplified, bipartite architecture offers a new framework to understand pelvic neurophysiology as well as development and evolution of the autonomic nervous system.


Subject(s)
Ganglia, Sympathetic/physiology , Neurons/physiology , Sacrum/innervation , Sympathetic Nervous System/physiology , Animals , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/embryology , Mice , Neurons/metabolism , Nitric Oxide Synthase Type I/metabolism , Parasympathetic Nervous System/anatomy & histology , Parasympathetic Nervous System/embryology , Parasympathetic Nervous System/physiology , Pelvis/anatomy & histology , Pelvis/embryology , Pelvis/innervation , Sacrum/anatomy & histology , Sacrum/embryology , Spinal Nerves/physiology , Sympathetic Nervous System/anatomy & histology , Sympathetic Nervous System/embryology , Thorax/innervation , Transcription, Genetic , Vesicular Acetylcholine Transport Proteins/metabolism
5.
PLoS One ; 11(2): e0147535, 2016.
Article in English | MEDLINE | ID: mdl-26859889

ABSTRACT

The heterotrimeric G protein subunit Gsα couples receptors to activate adenylyl cyclase and is required for the intracellular cAMP response and protein kinase A (PKA) activation. Gsα is ubiquitously expressed in many cell types; however, the role of Gsα in neural crest cells (NCCs) remains unclear. Here we report that NCCs-specific Gsα knockout mice die within hours after birth and exhibit dramatic craniofacial malformations, including hypoplastic maxilla and mandible, cleft palate and craniofacial skeleton defects. Histological and anatomical analysis reveal that the cleft palate in Gsα knockout mice is a secondary defect resulting from craniofacial skeleton deficiencies. In Gsα knockout mice, the morphologies of NCCs-derived cranial nerves are normal, but the development of dorsal root and sympathetic ganglia are impaired. Furthermore, loss of Gsα in NCCs does not affect cranial NCCs migration or cell proliferation, but significantly accelerate osteochondrogenic differentiation. Taken together, our study suggests that Gsα is required for neural crest cells-derived craniofacial development.


Subject(s)
Facial Bones/embryology , GTP-Binding Protein alpha Subunits, Gs/metabolism , Morphogenesis , Skull/embryology , Animals , Cell Differentiation , Cleft Palate/embryology , Cleft Palate/genetics , Facial Bones/abnormalities , GTP-Binding Protein alpha Subunits, Gs/deficiency , GTP-Binding Protein alpha Subunits, Gs/genetics , Ganglia, Sympathetic/embryology , Gene Knockout Techniques , Mice , Mutation , Neural Crest/cytology , Neural Crest/metabolism , Skull/abnormalities , Spinal Nerve Roots/embryology
6.
Dev Neurobiol ; 75(12): 1352-67, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25788138

ABSTRACT

Neurogenesis in embryonic sympathetic ganglia involves neuroblasts that resume proliferation following neuronal differentiation. As cell cycle exit is not associated with neuronal differentiation, the identity of proliferating neuroblasts is incompletely understood. Here, we use sympathetic ganglia of chick embryos to define the timing of neurogenesis and neuroblast identity focusing on the expression and function of the transcription factor Prox1. We show that a large fraction of neuroblasts has initially withdrawn from the cell cycle at embryonic day 3 (E3), which is reflected by a high proportion of p27(+)/Islet1(+) neuroblasts (63%) and low numbers of EdU(+)/Islet1(+) cells (12%). The proportion of proliferating Islet1(+) neuroblasts, identified by EdU pulse labeling and by the absence of the postmitotic marker p27 increases to reach maximal levels at E5, when virtually all neuroblasts are in the cell cycle (95%). Subsequently, the proportion of EdU-labeled and p27(-) neuroblasts is reduced to reach low levels at E11. Interestingly, the expression of the transcription factor Prox1 is restricted to the neuronal lineage, that is, Sox10(+)/Phox2b(+) neuron progenitors, proliferating p27(-)/Islet1(+) neuroblasts and nascent neurons but is rapidly lost in postmitotic neurons. In vitro and in vivo knockdown and overexpression experiments demonstrate effects of Prox1 in the support of neuroblast proliferation and survival. Taken together, these results define the neurogenesis period in the chick paravertebral sympathetic ganglia including an initial cell cycle withdrawal and identify Prox1 as a marker and regulator of proliferating sympathetic neuroblasts.


Subject(s)
Avian Proteins/metabolism , Ganglia, Sympathetic/embryology , Homeodomain Proteins/metabolism , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Tumor Suppressor Proteins/metabolism , Animals , Avian Proteins/genetics , Cell Cycle/physiology , Cells, Cultured , Chick Embryo , Ganglia, Parasympathetic/embryology , Ganglia, Parasympathetic/physiology , Ganglia, Sympathetic/physiology , Gene Knockdown Techniques , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , SOXE Transcription Factors/metabolism , Transcription Factors/metabolism , Transfection , Tumor Suppressor Proteins/genetics
7.
Dev Biol ; 400(2): 210-23, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25661788

ABSTRACT

The development of sympathetic neurons and chromaffin cells is differentially controlled at distinct stages by various extrinsic and intrinsic signals. Here we use conditional deletion of Dicer1 in neural crest cells and noradrenergic neuroblasts to identify stage specific functions in sympathoadrenal lineages. Conditional Dicer1 knockout in neural crest cells of Dicer1(Wnt1Cre) mice results in a rapid reduction in the size of developing sympathetic ganglia and adrenal medulla. In contrast, Dicer1 elimination in noradrenergic neuroblasts of Dicer1(DbhiCre) animals affects sympathetic neuron survival starting at late embryonic stages and chromaffin cells persist at least until postnatal week 1. A differential function of Dicer1 signaling for the development of embryonic noradrenergic and cholinergic sympathetic neurons is demonstrated by the selective increase in the expression of Tlx3 and the cholinergic marker genes VAChT and ChAT at E16.5. The number of Dbh, Th and TrkA expressing noradrenergic neurons is strongly decreased in Dicer1-deficient sympathetic ganglia at birth, whereas Tlx3(+)/ Ret(+) cholinergic neurons cells are spared from cell death. The postnatal death of chromaffin cells is preceded by the loss of Ascl1, mir-375 and Pnmt and an increase in the markers Ret and NF-M, which suggests that Dicer1 is required for the maintenance of chromaffin cell differentiation and survival. Taken together, these findings demonstrate distinct stage and lineage specific functions of Dicer1 signaling in differentiation and survival of sympathetic neurons and adrenal chromaffin cells.


Subject(s)
Adrenal Medulla/cytology , Chromaffin Cells/cytology , DEAD-box RNA Helicases/metabolism , Ganglia, Sympathetic/cytology , Ribonuclease III/metabolism , Adrenal Medulla/embryology , Adrenal Medulla/innervation , Adrenal Medulla/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Survival , Chromaffin Cells/metabolism , Ganglia, Sympathetic/embryology , Ganglia, Sympathetic/metabolism , Mice , Neural Crest/metabolism , Stem Cells/metabolism
8.
Dev Growth Differ ; 57(2): 146-57, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25581786

ABSTRACT

Neural crest cells (NCCs) are highly migratory progenitor cells that give rise to a vast array of differentiated cell types. One of their key derivatives is the autonomic nervous system (ANS) that is comprised in part from chromaffin cells of the adrenal medulla and organ of Zuckerkandl, the sympathetic chain and additional prevertebral ganglia such as the celiac ganglia, suprarenal ganglia and mesenteric ganglia. In this review we discuss recent advances toward our understanding of how the NCC precursors of the ANS migrate to their target regions, how they are instructed to differentiate into the correct cell types, and the morphogenetic signals controlling their development. Many of these processes remain enigmatic to developmental biologists worldwide. Taking advantage of lineage tracing mouse models one of our own aims is to address the morphogenetic events underpinning the formation of the ANS and to identify the molecular mechanisms that help to segregate a mixed population of NCCs into pathways specific for the sympathetic ganglia, sensory ganglia or adrenal medulla.


Subject(s)
Cell Movement/physiology , Ganglia, Sympathetic/embryology , Neural Crest/embryology , Neural Stem Cells/metabolism , Para-Aortic Bodies/metabolism , Animals , Ganglia, Sympathetic/cytology , Humans , Mice , Neural Crest/cytology , Neural Stem Cells/cytology , Para-Aortic Bodies/cytology
9.
Int J Dev Neurosci ; 41: 8-16, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25490618

ABSTRACT

Neural crest cells emerge from the dorsal neural tube early in development and give rise to sensory and sympathetic ganglia, adrenal cells, teeth, melanocytes and especially enteric nervous system. Several inhibitory molecules have been shown to play important roles in neural crest migration, among them are the chemorepulsive Slit1-3. It was known that Slits chemorepellants are expressed at the entry to the gut, and thus could play a role in the differential ability of vagal but not trunk neural crest cells to invade the gut and form enteric ganglia. Especially since trunk neural crest cells express Robo receptor while vagal do not. Thus, although we know that Robo mediates migration along the dorsal pathway in neural crest cells, we do not know if it is responsible in preventing their entry into the gut. The goal of this study was to further corroborate a role for Slit molecules in keeping trunk neural crest cells away from the gut. We observed that when we silenced Robo receptor in trunk neural crest, the sympathoadrenal (somites 18-24) were capable of invading gut mesenchyme in larger proportion than more rostral counterparts. The more rostral trunk neural crest tended not to migrate beyond the ventral aorta, suggesting that there are other repulsive molecules keeping them away from the gut. Interestingly, we also found that when we silenced Robo in sacral neural crest they did not wait for the arrival of vagal crest but entered the gut and migrated rostrally, suggesting that Slit molecules are the ones responsible for keeping them waiting at the hindgut mesenchyme. These combined results confirm that Slit molecules are responsible for keeping the timeliness of colonization of the gut by neural crest cells.


Subject(s)
Enteric Nervous System/cytology , Enteric Nervous System/embryology , Gene Expression Regulation, Developmental/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Crest/physiology , Age Factors , Animals , Cell Differentiation/genetics , Cell Movement/genetics , Chick Embryo , Electroporation , Ganglia, Sympathetic/embryology , Ganglia, Sympathetic/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , SOXE Transcription Factors/metabolism , Tubulin/metabolism
10.
Neural Dev ; 8: 16, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23961995

ABSTRACT

BACKGROUND: Neurons in sympathetic ganglia and neuroendocrine cells in the adrenal medulla share not only their embryonic origin from sympathoadrenal precursors in the neural crest but also a range of functional features. These include the capacity for noradrenaline biosynthesis, vesicular storage and regulated release. Yet the regulation of neuronal properties in early neuroendocrine differentiation is a matter of debate and the developmental expression of the vesicle fusion machinery, which includes components found in both neurons and neuroendocrine cells, is not resolved. RESULTS: Analysis of synaptic protein and pan-neuronal marker mRNA expression during mouse development uncovers profound differences between sympathetic neurons and adrenal chromaffin cells, which result in qualitatively similar but quantitatively divergent transcript profiles. In sympathetic neurons embryonic upregulation of synaptic protein mRNA follows early and persistent induction of pan-neuronal marker transcripts. In adrenal chromaffin cells pan-neuronal marker expression occurs only transiently and synaptic protein messages remain at distinctly low levels throughout embryogenesis. Embryonic induction of synaptotagmin I (Syt1) in sympathetic ganglia and postnatal upregulation of synaptotagmin VII (Syt7) in adrenal medulla results in a cell type-specific difference in isoform prevalence. Dicer 1 inactivation in catecholaminergic cells reduces high neuronal synaptic protein mRNA levels but not their neuroendocrine low level expression. Pan-neuronal marker mRNAs are induced in chromaffin cells to yield a more neuron-like transcript pattern, while ultrastructure is not altered. CONCLUSIONS: Our study demonstrates that remarkably different gene regulatory programs govern the expression of synaptic proteins in the neuronal and neuroendocrine branch of the sympathoadrenal system. They result in overlapping but quantitatively divergent transcript profiles. Dicer 1-dependent regulation is required to establish high neuronal mRNA levels for synaptic proteins and to maintain repression of neurofilament messages in neuroendocrine cells.


Subject(s)
Chromaffin System/embryology , DEAD-box RNA Helicases/metabolism , Ganglia, Sympathetic/embryology , Gene Expression Regulation, Developmental , Neurons/metabolism , Ribonuclease III/metabolism , Vesicular Transport Proteins/metabolism , Animals , Chromaffin Cells/metabolism , Chromaffin Cells/ultrastructure , Chromaffin System/growth & development , Chromaffin System/metabolism , Ganglia, Sympathetic/growth & development , Ganglia, Sympathetic/metabolism , Mice , Mice, Mutant Strains , Neurofilament Proteins/metabolism , RNA, Messenger/metabolism , Synaptosomal-Associated Protein 25/metabolism , Synaptotagmins/metabolism , rab3A GTP-Binding Protein/metabolism
11.
Auton Neurosci ; 177(2): 154-62, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23639816

ABSTRACT

This study aims at clarifying the detailed morphological and topographical changes of the thoracic part of the sympathetic trunk of sheep during fetal development. Bilateral micro-dissection of the thoracic sympathetic trunk was performed on 40 sheep fetuses aged 6-20 weeks (18 males and 22 females) under a stereomicroscope. The cervicothoracic ganglion (CTG) was observed on 75/80 sides (93.7%) and was composed of the caudal cervical and the first thoracic ganglia on 45/80 sides (56.2%), and of the caudal cervical and the first two thoracic ganglia on 30/80 sides (37.5%). The presence of the two last (12th-13th) thoracic ganglia was not constant. The influence of the sex, the side of the body, and the ages of the fetus on the morphology and topography of the thoracic sympathetic trunk in sheep were identified. In spite of the differences in the morphology and topography of the thoracic sympathetic trunk between early and late fetal developments, the morphology and topography of the older fetal thoracic sympathetic trunk tended to be similar to that of the adult sheep. To comprehend the comparative morphology of the fetal thoracic sympathetic trunk more completely, our results were compared with previous studies. Consequently, differences and similarities in the composition and position of the CTG, presence of single caudal cervical ganglion without fusion to the thoracic ganglia, and absence of the thoracic ganglia, and presence of splitting of the interganglionic branch were found among sheep, pig, and human fetuses. Therefore, sheep might be the appropriate animal model to be applied in human sympathetic nervous system.


Subject(s)
Autonomic Pathways/embryology , Fetal Development , Ganglia, Sympathetic/embryology , Sympathetic Nervous System/embryology , Thorax/embryology , Thorax/innervation , Animals , Autonomic Pathways/physiology , Female , Fetal Development/physiology , Ganglia, Sympathetic/physiology , Male , Pregnancy , Sheep , Sympathetic Nervous System/physiology , Thorax/physiology
12.
Oncogene ; 32(21): 2640-8, 2013 May 23.
Article in English | MEDLINE | ID: mdl-22751132

ABSTRACT

AT-rich interaction domain molecule 3B (ARID3B) and MYCN are expressed in a portion of neuroblastoma, and form a combination that has strong oncogenic activity in mouse embryonic fibroblasts (MEFs). Here, we show that this combination can also convert neural stem cells to neuroblastoma-like tumor. To address whether there are common mechanisms regulating the expression of this combination of genes, we examined public repositories of gene expression data and found that although these genes are rarely expressed together, co-expression was observed in a proportion of germ cell tumors (GCTs), in embryonic stem (ES) cells and in testis. These cell types and tissues are related to pluripotency and we show here that in mouse ES cells, Arid3b and Mycn are indeed involved in cell proliferation; the former in avoiding cell death and the latter in driving cell cycle progression. Accordingly, the two genes are induced during somatic cell reprogramming to iPS, and this induction is accompanied by the switching of promoter histone marks from H3K27me3 to H3K4me3. Conversely, the switch from H3K4me3 to H3K27me3 in these genes occurs during the differentiation of neural crest to mature sympathetic ganglia cells. In many, if not most, neuroblastomas these genes carry H3K4me3 marks within their promoters. Thus, a failure of the epigenetic silencing of these genes during development may be an underlying factor responsible for neuroblastoma.


Subject(s)
DNA-Binding Proteins/biosynthesis , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Neuroblastoma/metabolism , Nuclear Proteins/biosynthesis , Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Ganglia, Sympathetic/embryology , Ganglia, Sympathetic/pathology , Histones/genetics , Histones/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Neural Crest/embryology , Neural Crest/pathology , Neuroblastoma/genetics , Neuroblastoma/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Proto-Oncogene Proteins/genetics
13.
Int. j. morphol ; 30(4): 1569-1576, dic. 2012. ilus
Article in English | LILACS | ID: lil-670181

ABSTRACT

The cardiac plexus is formed by sympathetic nerves originating from the superior, middle, inferior cervical or cervicothoracic ganglia as well as from the first to the fifth thoracic ganglia. Furthermore, the vagus nerve and its counterpart, the recurrent laryngeal nerve supply the cardiac plexus with parasympathetic cardiac nerves. This investigation aimed to review and record the medial contributions of the cervical ganglia, first to fifth thoracic ganglia and medial contributions of the vagus and recurrent laryngeal nerves to the cardiac plexus. The study involved bilateral micro-dissection of forty cadaveric fetal specimens (n=80). The origins of sympathetic contributions to the cardiac plexus were described as either ganglionic, inter-ganglionic or from both the ganglion and the inter-ganglionic sympathetic chain. The number of cervical sympathetic ganglia varied from two to five in this study; the superior cervical ganglion was constant while the middle cervical, vertebral, inferior cervical or cervicothoracic ganglia were variable. The prevalence of cardiac nerves were as follows: superior cervical cardiac nerve (95%); middle cervical cardiac nerve (73%); vertebral cardiac nerve (41%); inferior cervical cardiac nerve (21%) and cervicothoracic cardiac nerve (24%). This investigation records the thoracic caudal limit of the thoracic sympathetic contributions to the cardiac plexus as the T5 ganglion. The findings of this study highlight the importance of understanding the medial sympathetic contributions and their variations to the cardiac plexus as this may assist surgeons during minimal access surgical procedures, sympathectomies, pericardiectomies and in the management of diseases like Raynaud's Phenomenon and angina pectoris.


El plexo cardíaco está formado por los nervios simpáticos procedentes de los ganglios cervicales superior, medio e inferior o cervicotorácico, así como los ganglios torácicos desde el primero al quinto. Por otra parte, el nervio vago y su contraparte, el nervio laríngeo recurrente suministra al plexo cardíaco nervios cardíacos parasimpático. Esta investigación tuvo como objetivo revisar y registrar las contribuciones mediales de los ganglios cervicales, ganglios torácicos del primero al quinto ganglios y contribuciones mediales de los nervios laríngeos recurrentes y vagos en el plexo cardíaco. Se realizó la micro-disección bilateral de cuarenta especímenes cadavéricos fetales (n = 80). Los orígenes de las contribuciones simpáticas hacia el plexo cardíaco se describen de forma independiente como ganglionar o inter-ganglionar, o desde ambos ganglios y la cadena simpática interganglionar. El número de ganglios simpáticos cervicales varió de dos a cinco; el ganglio cervical superior fue constante, mientras que los ganglios medio-cervical, vertebral, cervical inferior o cervicotorácico fueron variables. La prevalencia de los nervios cardíacos fueron: nervio cardíaco cervical superior (95%); nervio cardíaco cervical medio (73%); nervio cardiaco vertebral (41%); nervio cardíaco cervical inferior (21%) y nervio cardíaco cervicotorácico (24% ). La investigación registró el límite torácico caudal de las contribuciones torácicas simpáticos al plexo cardíaco como el ganglio T5. Los resultados de este estudio muestran la importancia de comprender las contribuciones simpáticas mediales y sus variaciones en el plexo cardíaco, ya que podrían ayudar a los cirujanos durante los procedimientos quirúrgicos mínimanente invasivos, simpatectomías, pericardiectomías y en el manejo de enfermedades como el fenómeno de Raynaud y la angina de pecho.


Subject(s)
Humans , Fetal Heart/innervation , Ganglia, Parasympathetic/embryology , Ganglia, Sympathetic/embryology , Cadaver , Fetus , Ganglia, Parasympathetic/anatomy & histology , Ganglia, Sympathetic/anatomy & histology
14.
Rom J Morphol Embryol ; 53(3): 591-6, 2012.
Article in English | MEDLINE | ID: mdl-22990552

ABSTRACT

The vagus trunks, anterior and posterior, and their respective branches control the parasympathetic innervation of the stomach. After giving off a few thin branches, at the lower part of the esophagus and the cardiac region of the stomach, the anterior vagal trunk divides into its main branches: four or five consecutive direct branches which supply the upper part of the lesser curvature; these nerves do not form plexuses and thus, they may be individually dissected. One of the branches is stronger than the others and is called the principal anterior nerve of the lesser curvature (anterior nerve of Latarjet). The present study was conducted on eight fetuses of different gestational age (resulting from spontaneous abortions, without malformations), observing the Romanian laws of professional ethics, and 15 adult cadavers (male and female) whose celiac region was dissected macro- and mesoscopically to reveal both the celiac ganglia and their afferent and efferent vessels. For the microscopic study, we used the Bielschowsky silver staining method. The meso- and macroscopic dissections revealed the anterior and posterior vagal trunks in all the specimens (100%), as well as a rich gastric periarterial plexus. The microscopic samples focused on the ontogeny of the celiac ganglion in various gestational stages.


Subject(s)
Celiac Plexus/anatomy & histology , Celiac Plexus/embryology , Ganglia, Sympathetic/anatomy & histology , Ganglia, Sympathetic/embryology , Stomach/innervation , Vagus Nerve/anatomy & histology , Vagus Nerve/embryology , Female , Fetus/anatomy & histology , Gestational Age , Humans , Male , Stomach/anatomy & histology , Stomach/embryology
15.
J Clin Invest ; 122(9): 3145-58, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22922260

ABSTRACT

The most common forms of neurocristopathy in the autonomic nervous system are Hirschsprung disease (HSCR), resulting in congenital loss of enteric ganglia, and neuroblastoma (NB), childhood tumors originating from the sympathetic ganglia and adrenal medulla. The risk for these diseases dramatically increases in patients with congenital central hypoventilation syndrome (CCHS) harboring a nonpolyalanine repeat expansion mutation of the Paired-like homeobox 2b (PHOX2B) gene, but the molecular mechanism of pathogenesis remains unknown. We found that introducing nonpolyalanine repeat expansion mutation of the PHOX2B into the mouse Phox2b locus recapitulates the clinical features of the CCHS associated with HSCR and NB. In mutant embryos, enteric and sympathetic ganglion progenitors showed sustained sex-determining region Y (SRY) box10 (Sox10) expression, with impaired proliferation and biased differentiation toward the glial lineage. Nonpolyalanine repeat expansion mutation of PHOX2B reduced transactivation of wild-type PHOX2B on its known target, dopamine ß-hydroxylase (DBH), in a dominant-negative fashion. Moreover, the introduced mutation converted the transcriptional effect of PHOX2B on a Sox10 enhancer from repression to transactivation. Collectively, these data reveal that nonpolyalanine repeat expansion mutation of PHOX2B is both a dominant-negative and gain-of-function mutation. Our results also demonstrate that Sox10 regulation by PHOX2B is pivotal for the development and pathogenesis of the autonomic ganglia.


Subject(s)
Gene Expression Regulation , Homeodomain Proteins/genetics , SOXE Transcription Factors/genetics , Sequence Deletion , Transcription Factors/genetics , Animals , Cell Count , Cell Differentiation , Cell Proliferation , Cells, Cultured , Enhancer Elements, Genetic , Enteric Nervous System/embryology , Enteric Nervous System/metabolism , Enteric Nervous System/pathology , Ganglia, Sympathetic/embryology , Ganglia, Sympathetic/metabolism , Ganglia, Sympathetic/pathology , Gastrointestinal Tract/embryology , Gastrointestinal Tract/innervation , Gastrointestinal Tract/pathology , Gene Knock-In Techniques , Hirschsprung Disease/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Hypoventilation/congenital , Hypoventilation/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Neural Crest/metabolism , Neural Crest/pathology , Neural Stem Cells/physiology , Neuroblastoma/genetics , Neuroglia/physiology , Neurons/metabolism , Neurons/physiology , Peptides/genetics , Phenotype , Repetitive Sequences, Amino Acid/genetics , SOXE Transcription Factors/metabolism , Sleep Apnea, Central/genetics , Spheroids, Cellular/physiology , Transcription Factors/metabolism , Transcription Factors/physiology , Transcriptional Activation
16.
J Neurosci Res ; 90(7): 1335-46, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22431311

ABSTRACT

We report extraordinary perpendicular orientations of neurons dependent on the presence of an external direct current (DC) voltage gradient. We chose chick dorsal root and postganglionic sympathetic neurons to evaluate. These were cultured in observation chambers in which the cells were separated from electrode products or substrate effects and maintained at 35°C. Both types of neurons showed a rapid restructuring of their anatomy. Typically, neurites that were not perpendicular to the voltage gradient were quickly resorbed into the cell body within a few minutes. Over 3-6 hr, significant new neurite growth occurred and was patterned perpendicular to the DC electrical field (Ef). This preferred asymmetry was dependent on the Ef, as was the initial retrograde degeneration of fibers. At 400-500 mV/mm, over 90% of the cells in culture assumed this orientation. Removal of the DC Ef led to a loss of the preferred orientation, with further random growth within the chambers. This is the first report of such responses in dorsal root ganglion neurons. We also used sympathetic neurons as a meaningful comparison to analyze whether there were any qualitative or quantitative differences between these two cell types of neural crest origin. We discuss the means by which these orientations were achieved.


Subject(s)
Cell Polarity/radiation effects , Electromagnetic Fields , Ganglia, Spinal/embryology , Ganglia, Spinal/radiation effects , Ganglia, Sympathetic/embryology , Ganglia, Sympathetic/radiation effects , Animals , Cell Differentiation/physiology , Cell Differentiation/radiation effects , Cell Polarity/physiology , Chick Embryo , Electric Stimulation/methods , Electricity , Ganglia, Spinal/cytology , Ganglia, Sympathetic/cytology , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/radiation effects , Neurites/radiation effects , Neurites/ultrastructure , Patch-Clamp Techniques/methods , Primary Cell Culture
17.
Dev Biol ; 363(1): 219-33, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22236961

ABSTRACT

Different prespecification of mesencephalic and trunk neural crest cells determines their response to environmental differentiation signals and contributes to the generation of different autonomic neuron subtypes, parasympathetic ciliary neurons in the head and trunk noradrenergic sympathetic neurons. The differentiation of ciliary and sympathetic neurons shares many features, including the initial BMP-induced expression of noradrenergic characteristics that is, however, subsequently lost in ciliary but maintained in sympathetic neurons. The molecular basis of specific prespecification and differentiation patterns has remained unclear. We show here that HoxB gene expression in trunk neural crest is maintained in sympathetic neurons. Ectopic expression of a single HoxB gene, HoxB8, in mesencephalic neural crest results in a strongly increased expression of sympathetic neuron characteristics like the transcription factor Hand2, tyrosine hydroxylase (TH) and dopamine-beta-hydroxylase (DBH) in ciliary neurons. Other subtype-specific properties like RGS4 and RCad are not induced. HoxB8 has only minor effects in postmitotic ciliary neurons and is unable to induce TH and DBH in the enteric nervous system. Thus, we conclude that HoxB8 acts by maintaining noradrenergic properties transiently expressed in ciliary neuron progenitors during normal development. HoxC8, HoxB9, HoxB1 and HoxD10 elicit either small and transient or no effects on noradrenergic differentiation, suggesting a selective effect of HoxB8. These results implicate that Hox genes contribute to the differential development of autonomic neuron precursors by maintaining noradrenergic properties in the trunk sympathetic neuron lineage.


Subject(s)
Adrenergic Neurons/metabolism , Autonomic Nervous System/metabolism , Avian Proteins/genetics , Cell Differentiation/genetics , Homeodomain Proteins/genetics , Adrenergic Neurons/cytology , Animals , Autonomic Nervous System/cytology , Autonomic Nervous System/embryology , Avian Proteins/metabolism , Chick Embryo , Chickens , Dopamine beta-Hydroxylase/genetics , Dopamine beta-Hydroxylase/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Spinal/metabolism , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/embryology , Ganglia, Sympathetic/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/metabolism , Neural Tube/cytology , Neural Tube/embryology , Neural Tube/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
18.
Dev Biol ; 358(2): 309-17, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21864525

ABSTRACT

Examining calcium dynamics within the neural crest (NC) has the potential to shed light on mechanisms that regulate complex cell migration and patterning events during embryogenesis. Unfortunately, typical calcium indicators are added to culture media or have low signal to noise after microinjection into tissue that severely limit analyses to cultured cells or superficial events. Here, we studied in vivo calcium dynamics during NC cell migration and patterning, using a genetically encoded calcium sensor, GCaMP3. We discovered that trunk NC cells displayed significantly more spontaneous calcium transients than cranial NC cells, and during cell aggregation versus cell migration events. Spontaneous calcium transients were more prevalent during NC cell aggregation into discrete sympathetic ganglia (SG). Blocking of N-cadherin activity in trunk NC cells near the presumptive SG led to a dramatic decrease in the frequency of spontaneous calcium transients. Detailed analysis and mathematical modeling of cell behaviors during SG formation showed NC cells aggregated into clusters after displaying a spontaneous calcium transient. This approach highlights the novel application of a genetically encoded calcium indicator to study subsets of cells during ventral events in embryogenesis.


Subject(s)
Calcium Signaling , Neural Crest/cytology , Neural Crest/metabolism , Animals , Body Patterning/physiology , Cell Movement/physiology , Chick Embryo , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/embryology , Ganglia, Sympathetic/metabolism , Indicators and Reagents , Microscopy, Confocal , Models, Neurological , Neural Crest/embryology , Time-Lapse Imaging
19.
J Neurosci ; 30(44): 14896-906, 2010 Nov 03.
Article in English | MEDLINE | ID: mdl-21048148

ABSTRACT

Kinesin-12 (also called Kif15) is a mitotic motor protein that continues to be expressed in developing neurons. Depletion of kinesin-12 causes axons to grow faster, more than doubles the frequency of microtubule transport in both directions in the axon, prevents growth cones from turning properly, and enhances the invasion of microtubules into filopodia. These results are remarkably similar to those obtained in previous studies in which neurons were depleted of kinesin-5 (also called Eg5 or Kif11), another mitotic motor protein that continues to be expressed in developing neurons. However, there are also notable differences in the phenotypes obtained with depleting each of these motors. Depleting kinesin-12 decreases axonal branching and growth cone size, whereas inhibiting kinesin-5 increases these parameters. In addition, depleting kinesin-12 diminishes the appearance of growth-cone-like waves along the length of the axon, an effect not observed with depletion of kinesin-5. Finally, depletion of kinesin-12 abolishes the "waggling" behavior of microtubules that occurs as they assemble along actin bundles within filopodia, whereas inhibition of kinesin-5 does not. Interestingly, and perhaps relevant to these differences in phenotype, in biochemical studies, kinesin-12 coimmunoprecipitates with actin but kinesin-5 does not. Collectively, these findings support a scenario whereby kinesin-12 shares functions with kinesin-5 related to microtubule-microtubule interactions, but kinesin-12 has other functions not shared by kinesin-5 that are related to the ability of kinesin-12 to interact with actin.


Subject(s)
Cell Differentiation/physiology , Growth Cones/metabolism , Kinesins/physiology , Nervous System/embryology , Nervous System/growth & development , Neurogenesis/physiology , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/embryology , Ganglia, Sympathetic/growth & development , Growth Cones/ultrastructure , Microtubule-Associated Proteins/physiology , Microtubules/metabolism , Microtubules/ultrastructure , Nervous System/cytology , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Rats
20.
J Neurosci ; 30(32): 10833-43, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20702712

ABSTRACT

The transcription factor Gata3 is essential for the development of sympathetic neurons and adrenal chromaffin cells. As Gata3 expression is maintained up to the adult stage, we addressed its function in differentiated sympathoadrenal cells at embryonic and adult stages by conditional Gata3 elimination. Inactivation of Gata3 in embryonic DBH-expressing neurons elicits a strong reduction in neuron numbers due to apoptotic cell death and reduced proliferation. No selective effect on noradrenergic gene expression (TH and DBH) was observed. Interestingly, Gata3 elimination in DBH-expressing neurons of adult animals also results in a virtually complete loss of sympathetic neurons. In the Gata3-deficient population, the expression of anti-apoptotic genes (Bcl-2, Bcl-xL, and NFkappaB) is diminished, whereas the expression of pro-apoptotic genes (Bik, Bok, and Bmf) was increased. The expression of noradrenergic genes (TH and DBH) is not affected. These results demonstrate that Gata3 is continuously required for maintaining survival but not differentiation in the sympathetic neuron lineage up to mature neurons of adult animals.


Subject(s)
GATA3 Transcription Factor/metabolism , Ganglia, Sympathetic/cytology , Gene Expression Regulation, Developmental/physiology , Neurons/physiology , Age Factors , Animals , Calcium-Binding Proteins , Caspase 3/metabolism , Cell Proliferation , Cell Size , Cell Survival/genetics , Cells, Cultured , Chick Embryo , Chromaffin Cells/metabolism , Dopa Decarboxylase/genetics , Dopa Decarboxylase/metabolism , Embryo, Mammalian , GATA3 Transcription Factor/deficiency , Ganglia, Sympathetic/embryology , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , In Situ Nick-End Labeling/methods , Intracellular Signaling Peptides and Proteins/metabolism , Ki-67 Antigen/metabolism , Mice , Mice, Knockout , Mutation/genetics , RNA, Messenger/metabolism , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/genetics , Receptor, trkA/metabolism , Stathmin , Transcription Factors/metabolism , Transfection/methods , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...