Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 336
Filtrar
1.
Biol Open ; 13(5)2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38639409

RESUMO

Blood vessels serve as intermediate conduits for the extension of sympathetic axons towards target tissues, while also acting as crucial targets for their homeostatic processes encompassing the regulation of temperature, blood pressure, and oxygen availability. How sympathetic axons innervate not only blood vessels but also a wide array of target tissues is not clear. Here we show that in embryonic skin, after the establishment of co-branching between sensory nerves and blood vessels, sympathetic axons invade the skin alongside these sensory nerves and extend their branches towards these blood vessels covered by vascular smooth muscle cells (VSMCs). Our mosaic labeling technique for sympathetic axons shows that collateral branching predominantly mediates the innervation of VSMC-covered blood vessels by sympathetic axons. The expression of nerve growth factor (NGF), previously known to induce collateral axon branching in culture, can be detected in the vascular smooth muscle cell (VSMC)-covered blood vessels, as well as sensory nerves. Indeed, VSMC-specific Ngf knockout leads to a significant decrease of collateral branching of sympathetic axons innervating VSMC-covered blood vessels. These data suggest that VSMC-derived NGF serves as an inductive signal for collateral branching of sympathetic axons innervating blood vessels in the embryonic skin.


Assuntos
Músculo Liso Vascular , Fator de Crescimento Neural , Pele , Animais , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/inervação , Fator de Crescimento Neural/metabolismo , Camundongos , Pele/inervação , Pele/irrigação sanguínea , Pele/metabolismo , Miócitos de Músculo Liso/metabolismo , Axônios/metabolismo , Axônios/fisiologia , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/inervação , Vasos Sanguíneos/metabolismo , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/fisiologia , Sistema Nervoso Simpático/metabolismo , Camundongos Knockout
2.
Int J Mol Sci ; 22(22)2021 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-34830206

RESUMO

Nitric oxide (NO) has been shown to stimulate differentiation and increase the survival of ganglionic sympathetic neurons. The proportion of neuronal NOS-immunoreactive sympathetic preganglionic neurons is particularly high in newborn rats and decreases with maturation. However, the role of NO in the development of vascular sympathetic innervation has never been studied before. We tested the hypothesis that intrauterine NO deficiency weakened the development of vascular sympathetic innervation and thereby changed the contractility of peripheral arteries and blood pressure level in two-week-old offspring. Pregnant rats consumed NOS inhibitor L-NAME (250 mg/L in drinking water) from gestational day 10 until delivery. Pups in the L-NAME group had a reduced body weight and blood level of NO metabolites at 1-2 postnatal days. Saphenous arteries from two-week-old L-NAME offspring demonstrated a lower density of sympathetic innervation, a smaller inner diameter, reduced maximal active force and decreased α-actin/ß-actin mRNA expression ratio compared to the controls. Importantly, pups in the L-NAME group exhibited decreased blood pressure levels before, but not after, ganglionic blockade with chlorisondamine. In conclusion, intrauterine L-NAME exposure is followed by the impaired development of the sympathetic nervous system in early postnatal life, which is accompanied by the structural and functional remodeling of arterial blood vessels.


Assuntos
Artérias/inervação , Inibidores Enzimáticos/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/crescimento & desenvolvimento , Remodelação Vascular/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Artérias/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Feminino , Idade Gestacional , Masculino , Modelos Animais , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Gravidez , Ratos , Ratos Wistar , Sistema Nervoso Simpático/metabolismo
3.
J Anat ; 239(1): 32-45, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33641166

RESUMO

Although the development of the sympathetic trunks was first described >100 years ago, the topographic aspect of their development has received relatively little attention. We visualised the sympathetic trunks in human embryos of 4.5-10 weeks post-fertilisation, using Amira 3D-reconstruction and Cinema 4D-remodelling software. Scattered, intensely staining neural crest-derived ganglionic cells that soon formed longitudinal columns were first seen laterally to the dorsal aorta in the cervical and upper thoracic regions of Carnegie stage (CS)14 embryos. Nerve fibres extending from the communicating branches with the spinal cord reached the trunks at CS15-16 and became incorporated randomly between ganglionic cells. After CS18, ganglionic cells became organised as irregular agglomerates (ganglia) on a craniocaudally continuous cord of nerve fibres, with dorsally more ganglionic cells and ventrally more fibres. Accordingly, the trunks assumed a "pearls-on-a-string" appearance, but size and distribution of the pearls were markedly heterogeneous. The change in position of the sympathetic trunks from lateral (para-aortic) to dorsolateral (prevertebral or paravertebral) is a criterion to distinguish the "primary" and "secondary" sympathetic trunks. We investigated the position of the trunks at vertebral levels T2, T7, L1 and S1. During CS14, the trunks occupied a para-aortic position, which changed into a prevertebral position in the cervical and upper thoracic regions during CS15, and in the lower thoracic and lumbar regions during CS18 and CS20, respectively. The thoracic sympathetic trunks continued to move further dorsally and attained a paravertebral position at CS23. The sacral trunks retained their para-aortic and prevertebral position, and converged into a single column in front of the coccyx. Based on our present and earlier morphometric measurements and literature data, we argue that differential growth accounts for the regional differences in position of the sympathetic trunks.


Assuntos
Embrião de Mamíferos/anatomia & histologia , Desenvolvimento Embrionário , Sistema Nervoso Simpático/embriologia , Humanos
4.
J Anat ; 237(4): 672-688, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32592418

RESUMO

Realistic models to understand the developmental appearance of the pelvic nervous system in mammals are scarce. We visualized the development of the inferior hypogastric plexus and its preganglionic connections in human embryos at 4-8 weeks post-fertilization, using Amira 3D reconstruction and Cinema 4D-remodelling software. We defined the embryonic lesser pelvis as the pelvic area caudal to both umbilical arteries and containing the hindgut. Neural crest cells (NCCs) appeared dorsolateral to the median sacral artery near vertebra S1 at ~5 weeks and had extended to vertebra S5 1 day later. Once para-arterial, NCCs either formed sympathetic ganglia or continued to migrate ventrally to the pre-arterial region, where they formed large bilateral inferior hypogastric ganglionic cell clusters (IHGCs). Unlike more cranial pre-aortic plexuses, both IHGCs did not merge because the 'pelvic pouch', a temporary caudal extension of the peritoneal cavity, interposed. Although NCCs in the sacral area started to migrate later, they reached their pre-arterial position simultaneously with the NCCs in the thoracolumbar regions. Accordingly, the superior hypogastric nerve, a caudal extension of the lumbar splanchnic nerves along the superior rectal artery, contacted the IHGCs only 1 day later than the lumbar splanchnic nerves contacted the inferior mesenteric ganglion. The superior hypogastric nerve subsequently splits to become the superior hypogastric plexus. The IHGCs had two additional sources of preganglionic innervation, of which the pelvic splanchnic nerves arrived at ~6.5 weeks and the sacral splanchnic nerves only at ~8 weeks. After all preganglionic connections had formed, separate parts of the inferior hypogastric plexus formed at the bladder neck and distal hindgut.


Assuntos
Desenvolvimento Embrionário/fisiologia , Plexo Hipogástrico/embriologia , Pelve Menor/inervação , Crista Neural/citologia , Sistema Nervoso Simpático/embriologia , Humanos , Pelve Menor/embriologia
5.
Med Sci (Paris) ; 35(8-9): 643-650, 2019.
Artigo em Francês | MEDLINE | ID: mdl-31532376

RESUMO

Arterial sympathetic innervation (ASI) is a complex biological process requiring a fine axonal guidance by arteries. Its physiological impact has remained unknown for decades but recently started to be better understood and recognized. ASI is a key element of the adaptive response of the cardiovascular system to challenging situations (exposure to cold, exercise…) as ASI controls the diameter of resistance arteries, thus blood supply to organs and systemic arterial blood pressure via arterial tone modulation. Defaults in ASI can lead to diseases, acting as a main cause or as an aggravating factor. Its impact is actively studied in cardiovascular diseases representing major public health issues, like hypertension, but ASI could also play a role in aging and many more pathological processes including cancer.


TITLE: Les fonctions de l'innervation sympathique artérielle - Du développement à la pathologie. ABSTRACT: L'innervation sympathique artérielle (ISA) est un processus biologique complexe nécessitant un guidage fin des axones des neurones sympathiques par les artères. L'ISA est un élément clé de l'adaptation du système cardiovasculaire aux différentes contraintes (exposition au froid, exercice, etc.) : elle contrôle le diamètre des artères de résistance, donc le flux sanguin parvenant aux organes et la pression artérielle systémique via la modulation du tonus artériel. Son importance lors du vieillissement et dans de nombreux contextes pathologiques est de mieux en mieux reconnue et comprise. Son intégration à la prise en charge de nombreuses maladies (hypertension, cancer, etc.) permettrait d'en améliorer traitements et pronostic.


Assuntos
Artérias/inervação , Doenças Cardiovasculares/fisiopatologia , Desenvolvimento Embrionário/fisiologia , Sistema Nervoso Simpático/fisiologia , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Artérias/embriologia , Artérias/crescimento & desenvolvimento , Artérias/patologia , Axônios/fisiologia , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/patologia , Fenômenos Fisiológicos Cardiovasculares , Sistema Cardiovascular/inervação , Sistema Cardiovascular/fisiopatologia , Humanos , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/crescimento & desenvolvimento , Sistema Nervoso Simpático/patologia , Sinapses/fisiologia
6.
Development ; 145(2)2018 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-29358215

RESUMO

Hematopoietic stem cells (HSCs) develop in discrete anatomical niches, migrating during embryogenesis from the aorta-gonad-mesonephros (AGM) region to the fetal liver, and finally to the bone marrow, where most HSCs reside throughout adult life. These niches provide supportive microenvironments that specify, expand and maintain HSCs. Understanding the constituents and molecular regulation of HSC niches is of considerable importance as it could shed new light on the mechanistic principles of HSC emergence and maintenance, and provide novel strategies for regenerative medicine. However, controversy exists concerning the cellular complexity of the bone marrow niche, and our understanding of the different HSC niches during development remains limited. In this Review, we summarize and discuss what is known about the heterogeneity of the HSC niches at distinct stages of their ontogeny, from the embryo to the adult bone marrow, drawing predominantly on data from mouse studies.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Nicho de Células-Tronco/fisiologia , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Aorta/embriologia , Linhagem da Célula , Feminino , Gônadas/embriologia , Neoplasias Hematológicas/patologia , Sistema Hematopoético/embriologia , Humanos , Masculino , Mesonefro/embriologia , Camundongos , Placenta/citologia , Placenta/fisiologia , Gravidez , Células Estromais/citologia , Células Estromais/fisiologia , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/fisiologia
7.
Anat Rec (Hoboken) ; 300(12): 2250-2262, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28865185

RESUMO

The objective of this study was to clarify the typical architecture and morphological variations of cervical sympathetic trunk (CST) in sheep during fetal period. Components of CST were examined on both sides of 40 male and female sheep fetuses aged from 60 to 140 days under a stereomicroscope. Skeletotopy and frequency of presence of cranial cervical ganglion (CCG), syntopy of cervical ganglia, and composition and topography of vagosympathetic trunk were consistent among specimens whereas the shape of cervical ganglia, the skeletotopy and number of three middle cervical ganglia (MG), and the frequency of communicating branches of CCG to the first cervical spinal nerve exhibited differences during fetal period. A reduction in the number of MG and the caudal movement of main MG were noted by increasing fetal age. Based on these detailed findings, comparative and developmental anatomy and evolutionary changes are discussed and compared with previous studies. The number of MG, skeletotopy of CCG and main MG, the number and range of communicating branches of CCG to spinal nerves, and the association of vagus and sympathetic nerves in fetal sheep were fundamentally different from those of mostly reported species. These results suggest that data obtained from CST of fetal sheep are significantly different from those obtained from humans, and it is problematic to apply them to humans because of the more cranial position of CCG, very narrow contribution of CCG to spinal nerve, absence of the vertebral ganglion, existence of multiple MG, and no communicating branches from MG to spinal nerves. Anat Rec, 300:2250-2262, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Ovinos/anatomia & histologia , Ovinos/embriologia , Gânglio Cervical Superior/anatomia & histologia , Gânglio Cervical Superior/embriologia , Animais , Feminino , Desenvolvimento Fetal/fisiologia , Masculino , Sistema Nervoso Simpático/anatomia & histologia , Sistema Nervoso Simpático/embriologia
8.
Science ; 354(6314): 893-897, 2016 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-27856909

RESUMO

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.


Assuntos
Gânglios Simpáticos/fisiologia , Neurônios/fisiologia , Sacro/inervação , Sistema Nervoso Simpático/fisiologia , Animais , Gânglios Simpáticos/citologia , Gânglios Simpáticos/embriologia , Camundongos , Neurônios/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Sistema Nervoso Parassimpático/anatomia & histologia , Sistema Nervoso Parassimpático/embriologia , Sistema Nervoso Parassimpático/fisiologia , Pelve/anatomia & histologia , Pelve/embriologia , Pelve/inervação , Sacro/anatomia & histologia , Sacro/embriologia , Nervos Espinhais/fisiologia , Sistema Nervoso Simpático/anatomia & histologia , Sistema Nervoso Simpático/embriologia , Tórax/inervação , Transcrição Gênica , Proteínas Vesiculares de Transporte de Acetilcolina/metabolismo
10.
Development ; 143(9): 1560-70, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-27143756

RESUMO

During development of the peripheral nervous system, excess neurons are generated, most of which will be lost by programmed cell death due to a limited supply of neurotrophic factors from their targets. Other environmental factors, such as 'competition factors' produced by neurons themselves, and axon guidance molecules have also been implicated in developmental cell death. Semaphorin 3A (Sema3A), in addition to its function as a chemorepulsive guidance cue, can also induce death of sensory neurons in vitro The extent to which Sema3A regulates developmental cell death in vivo, however, is debated. We show that in compartmentalized cultures of rat sympathetic neurons, a Sema3A-initiated apoptosis signal is retrogradely transported from axon terminals to cell bodies to induce cell death. Sema3A-mediated apoptosis utilizes the extrinsic pathway and requires both neuropilin 1 and plexin A3. Sema3A is not retrogradely transported in older, survival factor-independent sympathetic neurons, and is much less effective at inducing apoptosis in these neurons. Importantly, deletion of either neuropilin 1 or plexin A3 significantly reduces developmental cell death in the superior cervical ganglia. Taken together, a Sema3A-initiated apoptotic signaling complex regulates the apoptosis of sympathetic neurons during the period of naturally occurring cell death.


Assuntos
Apoptose/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neuropilina-1/metabolismo , Receptores de Superfície Celular/metabolismo , Semaforina-3A/metabolismo , Gânglio Cervical Superior/embriologia , Sistema Nervoso Simpático/embriologia , Animais , Axônios/metabolismo , Caspase 3/metabolismo , Células Cultivadas , Camundongos , Camundongos Knockout , Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Neuropilina-1/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/genética , Transdução de Sinais , Gânglio Cervical Superior/citologia , Gânglio Cervical Superior/fisiologia
11.
Clin Cancer Res ; 22(13): 3398-409, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27076624

RESUMO

PURPOSE: Neuroblastoma is a childhood malignancy originating from the sympathetic nervous system with a complex biology, prone to metastasize and relapse. High-risk, metastatic cases are explained in part by amplification or mutation of oncogenes, such as MYCN and ALK, and loss of tumor suppressor genes in chromosome band 1p. However, it is fundamental to identify other pathways responsible for the large portion of neuroblastomas with no obvious molecular alterations. EXPERIMENTAL DESIGN: Neuroblastoma cell lines were used for the assessment of tumor growth in vivo and in vitro Protein expression in tissues and cells was assessed using immunofluorescence and IHC. The association of promyelocytic leukemia (PML) expression with neuroblastoma outcome and relapse was calculated using log-rank and Mann-Whitney tests, respectively. Gene expression was assessed using chip microarrays. RESULTS: PML is detected in the developing and adult sympathetic nervous system, whereas it is not expressed or is low in metastatic neuroblastoma tumors. Reduced PML expression in patients with low-risk cancers, that is, localized and negative for the MYCN proto-oncogene, is strongly associated with tumor recurrence. PML-I, but not PML-IV, isoform suppresses angiogenesis via upregulation of thrombospondin-2 (TSP2), a key inhibitor of angiogenesis. Finally, PML-I and TSP2 expression inversely correlates with tumor angiogenesis and recurrence in localized neuroblastomas. CONCLUSIONS: Our work reveals a novel PML-I-TSP2 axis for the regulation of angiogenesis and cancer relapse, which could be used to identify patients with low-risk, localized tumors that might benefit from chemotherapy. Clin Cancer Res; 22(13); 3398-409. ©2016 AACR.


Assuntos
Recidiva Local de Neoplasia/patologia , Neovascularização Patológica/patologia , Neuroblastoma/patologia , Proteína da Leucemia Promielocítica/metabolismo , Trombospondinas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/genética , Crista Neural/embriologia , Neuroblastoma/genética , Proteína da Leucemia Promielocítica/genética , Isoformas de Proteínas/genética , Proto-Oncogene Mas , Fatores de Risco , Células-Tronco/citologia , Sistema Nervoso Simpático/embriologia , Trombospondinas/genética , Proteínas Supressoras de Tumor/genética
12.
Dev Biol ; 413(1): 70-85, 2016 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-26988118

RESUMO

During amniote embryogenesis the nervous and vascular systems interact in a process that significantly affects the respective morphogenesis of each network by forming a "neurovascular" link. The importance of neurovascular cross-talk in the central nervous system has recently come into focus with the growing awareness that these two systems interact extensively both during development, in the stem-cell niche, and in neurodegenerative conditions such as Alzheimer's Disease and Amyotrophic Lateral Sclerosis. With respect to the peripheral nervous system, however, there have been no live, real-time investigations of the potential relationship between these two developing systems. To address this deficit, we used multispectral 4D time-lapse imaging in a transgenic quail model in which endothelial cells (ECs) express a yellow fluorescent marker, while neural crest cells (NCCs) express an electroporated red fluorescent marker. We monitored EC and NCC migration in real-time during formation of the peripheral nervous system. Our time-lapse recordings indicate that NCCs and ECs are physically juxtaposed and dynamically interact at multiple locations along their trajectories. These interactions are stereotypical and occur at precise anatomical locations along the NCC migratory pathway. NCCs migrate alongside the posterior surface of developing intersomitic vessels, but fail to cross these continuous streams of motile ECs. NCCs change their morphology and migration trajectory when they encounter gaps in the developing vasculature. Within the nascent dorsal root ganglion, proximity to ECs causes filopodial retraction which curtails forward persistence of NCC motility. Overall, our time-lapse recordings support the conclusion that primary vascular networks substantially influence the distribution and migratory behavior of NCCs and the patterned formation of dorsal root and sympathetic ganglia.


Assuntos
Células Endoteliais/citologia , Gânglios Espinais/embriologia , Microscopia/métodos , Crista Neural/embriologia , Sistema Nervoso Periférico/embriologia , Sistema Nervoso Simpático/embriologia , Imagem com Lapso de Tempo/métodos , Doença de Alzheimer/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Bactérias/metabolismo , Padronização Corporal , Comunicação Celular , Movimento Celular , Coturnix , Gânglios Espinais/citologia , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Proteínas Luminescentes/metabolismo , Crista Neural/citologia , Células-Tronco/citologia
13.
Biochim Biophys Acta ; 1863(7 Pt B): 1904-15, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26778332

RESUMO

Starting from the late embryonic development, the sympathetic nervous system extensively innervates the heart and modulates its activity during the entire lifespan. The distribution of myocardial sympathetic processes is finely regulated by the secretion of limiting amounts of pro-survival neurotrophic factors by cardiac cells. Norepinephrine release by the neurons rapidly modulates myocardial electrophysiology, and increases the rate and force of cardiomyocyte contractions. Sympathetic processes establish direct interaction with cardiomyocytes, characterized by the presence of neurotransmitter vesicles and reduced cell-cell distance. Whether such contacts have a functional role in both neurotrophin- and catecholamine-dependent communication between the two cell types, is poorly understood. In this review we will address the effects of the sympathetic neuron activity on the myocardium and the hypothesis that the direct neuro-cardiac contact might have a key role both in norepinephrine and neurotrophin mediated signaling. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Assuntos
Neurônios Adrenérgicos/fisiologia , Coração/inervação , Miócitos Cardíacos/fisiologia , Junção Neuromuscular/fisiologia , Sistema Nervoso Simpático/fisiologia , Potenciais de Ação , Neurônios Adrenérgicos/metabolismo , Fatores Etários , Envelhecimento , Animais , Cardiopatias/metabolismo , Cardiopatias/fisiopatologia , Humanos , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Fator de Crescimento Neural/metabolismo , Junção Neuromuscular/metabolismo , Norepinefrina/metabolismo , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/metabolismo
14.
Acta Histochem ; 117(4-5): 415-24, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25765113

RESUMO

The protein cyclin D1 (CD1), which belongs to a family of proteins functioning as regulators of CDKs (cyclin-dependent kinases) throughout the cell cycle, has been immunohistochemically detected in a wide variety of human malignant tumors. The aim of the present study was to investigate immunohistochemically the expression and distribution of CD1 in the developing human peripheral sympathetic nervous system (PSNS) and in childhood peripheral neuroblastic tumors (neuroblastomas, ganglioneuroblastomas, and ganglioneuromas). The above mentioned fetal and neoplastic tissues represent an in vivo model in which undifferentiated neuroblastic cells undergo ganglion cell differentiation. During development, a strong nuclear expression of CD1 was restricted to neuroblasts, disappearing progressively from the maturing ganglion cells with increasing gestational age. In neoplastic tissues, CD1 immunoreactivity was restricted to neuroblastic cell component of all neuroblastomas and ganglioneuroblastomas, whereas it was absent or only focally detectable in maturing/mature ganglion cell component of differentiating neuroblastomas, ganglioneuroblastomas, and ganglioneuromas. We conclude that CD1 is a reliable marker, which can be used routinely to stain neuroblastic cells in both developing and neoplastic tissues. Furthermore, our results indicate that CD1 expression in childhood peripheral neuroblastic tumors recapitulates the changes during normal development of PSNS, as previously reported for Bcl-2 oncoprotein, c-ErbB2, insulin-like growth factor 2, ß-2-microglobulin, and cathepsin D. This is consistent with the current view that childhood peripheral neuroblastic tumors exhibit gene expression profiles mirroring those occurring during PSNS development.


Assuntos
Biomarcadores Tumorais/biossíntese , Ciclina D1/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Neuroblastoma , Sistema Nervoso Simpático , Adolescente , Adulto , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Criança , Pré-Escolar , Feminino , Humanos , Imuno-Histoquímica/métodos , Lactente , Masculino , Neuroblastoma/embriologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/patologia
15.
Dev Dyn ; 244(1): 56-68, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25138596

RESUMO

BACKGROUND: In mice, the intestinal tube develops from the splanchopleure before embryonic day 9.5. Subsequent patterning of nerves and blood vessels is critical for normal digestive function. A hierarchical branching vascular network allows for efficient nutrient absorption, while the complex enteric nervous system regulates intestinal motility as well as secretion, absorption, and blood flow. Despite the well-recognized significance of these systems, the precise mechanisms by which they develop have not been clearly established in mammals. RESULTS: Using a novel whole-mount immunohistochemical protocol, we visualize the pattern of intestinal neurovascular development in mice between embryonic day 10.5 and birth. In particular, we focus on the development and remodeling of the enteric vascular plexus, the migration and organization of enteric neural crest-derived cells, and the integration of peripheral sympathetic nerves with the enteric nervous system. These correlative data lead us to hypothesize a functional interaction between migrating neural crest-derived cells and endothelial cells of the primary capillary plexus, as well as a subsequent interaction between developing peripheral autonomic nerves and differentiated neural crest-derived cells. CONCLUSIONS: These studies provide useful anatomical data for continuing investigations on the functional mechanisms underlying intestinal organogenesis.


Assuntos
Intestinos , Neovascularização Fisiológica/fisiologia , Crista Neural/embriologia , Nervos Periféricos/embriologia , Sistema Nervoso Simpático/embriologia , Animais , Intestinos/irrigação sanguínea , Intestinos/embriologia , Intestinos/inervação , Camundongos , Crista Neural/citologia , Nervos Periféricos/citologia , Sistema Nervoso Simpático/citologia
16.
Horm Mol Biol Clin Investig ; 18(2): 63-77, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-25390003

RESUMO

BACKGROUND: A substantial body of epidemiological and experimental evidence suggests that a poor fetal and neonatal environment may "program" susceptibility in the offspring to later development of cardiovascular, renal and metabolic diseases. MATERIALS AND METHODS: This review focuses on current knowledge from the available literature regarding the mechanisms linking an adverse developmental environment with an increased risk for cardiovascular, renal and metabolic diseases in adult life. Moreover, this review highlights important sex-dependent differences in the adaptation to developmental insults. RESULTS: Developmental programming of several diseases is secondary to changes in different mechanisms inducing important alterations in the normal development of several organs that lead to significant changes in birth weight. The different diseases occurring as a consequence of an adverse environment during development are secondary to morphological and functional cardiovascular and renal changes, to epigenetic changes and to an activation of several hormonal and regulatory systems, such as angiotensin II, sympathetic activity, nitric oxide, COX2-derived metabolites, oxidative stress and inflammation. The important sex-dependent differences in the developmental programming of diseases seem to be partly secondary to the effects of sex hormones. Recent studies have shown that the progression of these diseases is accelerated during aging in both sexes. CONCLUSIONS: The cardiovascular, renal and metabolic diseases during adult life that occur as a consequence of several insults during fetal and postnatal periods are secondary to multiple structural and functional changes. Future studies are needed in order to prevent the origin and reduce the incidence and consequences of developmental programmed diseases.


Assuntos
Hipertensão/fisiopatologia , Nefropatias/fisiopatologia , Animais , Peso ao Nascer , Epigênese Genética , Feminino , Hormônios Esteroides Gonadais/metabolismo , Humanos , Hipertensão/embriologia , Hipertensão/metabolismo , Nefropatias/embriologia , Nefropatias/metabolismo , Masculino , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , Estresse Oxidativo , Gravidez , Sistema Renina-Angiotensina/fisiologia , Fatores Sexuais , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/crescimento & desenvolvimento , Sistema Nervoso Simpático/fisiopatologia
17.
Nat Commun ; 5: 3918, 2014 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-24875861

RESUMO

The mechanisms responsible for establishing correct target innervation during organ development are largely unknown. Sympathetic nerves follow blood vessels--typically arteries--to reach their endorgans, suggesting the existence of vascular guidance cues that direct axonal extension. The sinoatrial node and the ventricle of the heart receive sympathetic innervation from the stellate ganglia (STG). Here we show that STG axons follow veins, specifically the superior vena cavae and sinus venosus, to reach these targets. We find that election of these routes is determined by venous endothelium-derived endothelin-1, acting through its specific receptor Ednra expressed within a subpopulation of STG neurons. Furthermore, we demonstrate that Edn1-Ednra signalling is essential for functional regulation of the heart by sympathetic nerves. Our findings present venous Edn1 as a sympathetic guidance cue, and show how axon guidance mechanisms are coordinated with endorgan morphogenesis.


Assuntos
Axônios/metabolismo , Endotelina-1/metabolismo , Coração/embriologia , Receptor de Endotelina A/metabolismo , Gânglio Estrelado/embriologia , Veias/embriologia , Animais , Coração/inervação , Camundongos , Transdução de Sinais , Gânglio Estrelado/metabolismo , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/metabolismo , Veias/metabolismo
18.
Cell Death Differ ; 21(7): 1025-35, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24769728

RESUMO

Developing sympathetic neurons of the superior cervical ganglion are one of the best studied models of neuronal apoptosis. These cells require nerve growth factor (NGF) for survival at the time that they innervate their final target tissues during late embryonic and early postnatal development. In the absence of NGF, developing sympathetic neurons die by apoptosis in a transcription-dependent manner. Molecular studies of sympathetic neuron apoptosis began in the 1980s. We now know that NGF withdrawal activates the mitochondrial (intrinsic) pathway of apoptosis in sympathetic neurons cultured in vitro, and the roles of caspases, Bcl-2 (B-cell CLL/lymphoma 2) family proteins and XIAP (X-linked inhibitor of apoptosis protein) have been extensively studied. Importantly, a considerable amount has also been learned about the intracellular signalling pathways and transcription factors that regulate programmed cell death in sympathetic neurons. In this article, we review the key papers published in the past few years, covering all aspects of apoptosis regulation in sympathetic neurons and focusing, in particular, on how signalling pathways and transcription factors regulate the cell death programme. We make some comparisons with other models of neuronal apoptosis and describe possible future directions for the field.


Assuntos
Apoptose , Neurônios/fisiologia , Sistema Nervoso Simpático/citologia , Animais , Sobrevivência Celular , Expressão Gênica , Humanos , Fator de Crescimento Neural/fisiologia , Transdução de Sinais , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/crescimento & desenvolvimento
19.
Differentiation ; 88(4-5): 124-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25582843

RESUMO

During development, the growth of neural processes is regulated by an array of cellular and molecular mechanisms which influence growth rate, direction and branching. Recently, many members of the TNF superfamily have been shown to be key regulators of neurite growth during development. The founder member of this family, TNFα can both promote and inhibit neurite growth depending on the cellular context. Specifically, transmembrane TNFα promotes neurite growth, while soluble TNFα inhibits it. While the growth promoting effects of TNFα are restricted to a defined developmental window of early postnatal development, whether the growth inhibitory effects of soluble TNFα occur throughout development is unknown. In this study we used the extensively studied, well characterised neurons of the superior cervical ganglion to show that the growth inhibitory effects of soluble TNFα are restricted to a specific period of late embryonic and early postnatal development. Furthermore, we show that this growth inhibitory effect of soluble TNFα requires NF-κB signalling at all developmental stages at which soluble TNFα inhibits neurite growth. These findings raise the possibility that increases in the amount of soluble TNFα in vivo, for example as a result of maternal inflammation, could negatively affect neurite growth in developing neurons at specific stages of development.


Assuntos
Neuritos/efeitos dos fármacos , Neurogênese , Sistema Nervoso Simpático/citologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Neuritos/metabolismo , Neuritos/fisiologia , Ratos , Ratos Sprague-Dawley , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/crescimento & desenvolvimento
20.
Nat Neurosci ; 16(7): 865-73, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23749144

RESUMO

Reverse signaling via members of the tumor necrosis factor (TNF) superfamily controls multiple aspects of immune function. Here we document TNFα reverse signaling in the nervous system to our knowledge for the first time and show that it has a crucial role in establishing sympathetic innervation. During postnatal development, sympathetic axons express TNFα as they grow and branch in their target tissues, which in turn express TNF receptor 1 (TNFR1). In culture, soluble forms of TNFR1 act directly on postnatal sympathetic axons to promote growth and branching by a mechanism that depends on membrane-integrated TNFα and on downstream activation of ERK. Sympathetic innervation density is substantially lower in several tissues in postnatal and adult mice lacking either TNFα or TNFR1. These findings reveal that target-derived TNFR1 acts as a reverse-signaling ligand for membrane-integrated TNFα to promote growth and branching of sympathetic axons.


Assuntos
Axônios/fisiologia , Fibras Nervosas/fisiologia , Neurônios/citologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas ADAM/farmacologia , Proteína ADAM17 , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Células Cultivadas , Quelantes/farmacologia , Relação Dose-Resposta a Droga , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos , Camundongos Transgênicos , Fator de Crescimento Neural/farmacologia , RNA Mensageiro/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Gânglio Cervical Superior/citologia , Sistema Nervoso Simpático/citologia , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/crescimento & desenvolvimento , Fator de Necrose Tumoral alfa/genética , Tirosina 3-Mono-Oxigenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...