Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Infect Dis ; 2024 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-39397525

RESUMEN

Clostridioides difficile infection (CDI) is a predominant cause of intestinal infections. The intrinsic enteric nervous system (ENS) occupies the intestinal tissue in large numbers and intricately regulates various aspects of intestinal function. Nonetheless, the specific effects of CDI on the intrinsic ENS remain underexplored. Herein, we employed the TcdB variant (TcdB2) derived from hypervirulent C. difficile to elucidate the impact of CDI on neurons located in colonic wall. We found that TcdB2 directly induced dose-dependent cytopathic effects on enteric neurons both in vitro and in adult mice colons. Notably, an increased expression of choline acetyltransferase (ChAT) and neural nitric oxide synthase (nNOS) in colonic neurons prior to the onset of cytopathic changes following treatment with TcdB2 were observed, both in vivo and in vitro. These findings suggest that during CDI, TcdB not only causes neuronal loss but also alters the composition of neurotransmitters in the ENS.

2.
Part Fibre Toxicol ; 21(1): 5, 2024 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-38321545

RESUMEN

BACKGROUND: Currently, society and industry generate huge amounts of plastics worldwide. The ubiquity of microplastics is obvious, but its impact on the animal and human organism remains not fully understood. The digestive tract is one of the first barriers between pathogens and xenobiotics and a living organism. Its proper functioning is extremely important in order to maintain homeostasis. The aim of this study was to determine the effect of microplastic on enteric nervous system and histological structure of swine duodenum. The experiment was carried out on 15 sexually immature gilts, approximately 8 weeks old. The animals were randomly divided into 3 study groups (n = 5/group). The control group received empty gelatin capsules once a day for 28 days, the first research group received daily gelatin capsules with polyethylene terephthalate (PET) particles as a mixture of particles of various sizes (maximum particle size 300 µm) at a dose of 0.1 g/animal/day. The second study group received a dose ten times higher-1 g/animal/day. RESULTS: A dose of 1 g/day/animal causes more changes in the enteric nervous system and in the histological structure of duodenum. Statistically significant differences in the expression of cocaine and amphetamine regulated transcript, galanin, neuronal nitric oxide synthase, substance P, vesicular acetylcholine transporter and vasoactive intestinal peptide between control and high dose group was noted. The histopathological changes were more frequently observed in the pigs receiving higher dose of PET. CONCLUSION: Based on this study it may be assumed, that oral intake of microplastic might have potential negative influence on digestive tract, but it is dose-dependent.


Asunto(s)
Microplásticos , Plásticos , Humanos , Porcinos , Animales , Femenino , Tereftalatos Polietilenos/metabolismo , Tereftalatos Polietilenos/farmacología , Gelatina/metabolismo , Gelatina/farmacología , Duodeno/metabolismo , Neuronas
3.
Cell Commun Signal ; 21(1): 273, 2023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37798789

RESUMEN

BACKGROUND: Diabetes can lead to extensive damage to the enteric nervous system (ENS), causing gastrointestinal motility disorders. However, there is currently a lack of effective treatments for diabetes-induced ENS damage. Enteric neural precursor cells (ENPCs) closely regulate the structural and functional integrity of the ENS. L-Fucose, is a dietary sugar that has been showed to effectively ameliorate central nervous system injuries, but its potential for ameliorating ENS damage and the involvement of ENPCs in this process remains uncertain. METHODS: Genetically engineered mice were generated for lineage tracing of ENPCs in vivo. Using diabetic mice in vivo and high glucose-treated primary ENPCs in vitro, the effects of L-Fucose on the injured ENS and ENPCs was evaluated by assessing gastrointestinal motility, ENS structure, and the differentiation of ENPCs. The key signaling pathways in regulating neurogenesis and neural precursor cells properties, transforming growth factor-ß (TGF-ß) and its downstream signaling pathways were further examined to clarify the potential mechanism of L-Fucose on the injured ENS and ENPCs. RESULTS: L-Fucose improved gastrointestinal motility in diabetic mice, including increased defecation frequency (p < 0.05), reduced total gastrointestinal transmission time (p < 0.001) and bead expulsion time (p < 0.05), as well as enhanced spontaneous contractility and electric field stimulation-induced contraction response in isolated colonic muscle strips (p < 0.001). The decrease in the number of neurons and glial cells in the ENS of diabetic mice were reversed by L-Fucose treatment. More importantly, L-Fucose treatment significantly promoted the proportion of ENPCs differentiated into neurons and glial cells both in vitro and in vivo, accompanied by inhibiting SMAD2 phosphorylation. CONCLUSIONS: L-Fucose could promote neurogenesis and gliogenesis derived from ENPCs by inhibiting the SMAD2 signaling, thus facilitating ENS regeneration and gastrointestinal motility recovery in type 1 diabetic mice. Video Abstract.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Sistema Nervioso Entérico , Células-Madre Neurales , Ratones , Animales , Fucosa/farmacología , Fucosa/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Neuronas/metabolismo , Sistema Nervioso Entérico/metabolismo , Transducción de Señal
4.
Int J Mol Sci ; 24(12)2023 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-37373470

RESUMEN

Several studies have shown that the gut microbiota influences behavior and, in turn, changes in the immune system associated with symptoms of depression or anxiety disorder may be mirrored by corresponding changes in the gut microbiota. Although the composition/function of the intestinal microbiota appears to affect the central nervous system (CNS) activities through multiple mechanisms, accurate epidemiological evidence that clearly explains the connection between the CNS pathology and the intestinal dysbiosis is not yet available. The enteric nervous system (ENS) is a separate branch of the autonomic nervous system (ANS) and the largest part of the peripheral nervous system (PNS). It is composed of a vast and complex network of neurons which communicate via several neuromodulators and neurotransmitters, like those found in the CNS. Interestingly, despite its tight connections to both the PNS and ANS, the ENS is also capable of some independent activities. This concept, together with the suggested role played by intestinal microorganisms and the metabolome in the onset and progression of CNS neurological (neurodegenerative, autoimmune) and psychopathological (depression, anxiety disorders, autism) diseases, explains the large number of investigations exploring the functional role and the physiopathological implications of the gut microbiota/brain axis.


Asunto(s)
Microbioma Gastrointestinal , Sistema Nervioso , Humanos , Animales , Sistema Nervioso Entérico , Sistema Nervioso/metabolismo , Eje Cerebro-Intestino , Intestinos/metabolismo , Intestinos/microbiología , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/microbiología , Enfermedades del Sistema Nervioso/patología , Disbiosis/metabolismo , Disbiosis/microbiología , Disbiosis/patología
5.
Adv Exp Med Biol ; 1383: 307-318, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36587168

RESUMEN

Historically and quantitatively, the enteric site of serotonin (5-HT) storage has primacy over those of any other organ. 5-HT, by the name of "enteramine", was first discovered in the bowel, and the gut produces most of the body's 5-HT. Not only does the bowel secrete 5-HT prodigiously but it also expresses a kaleidoscopic abundance of 5-HT receptors. The larger of two enteric 5-HT stores is mucosal, biosynthetically dependent upon tryptophan hydroxylase1 (TPH1), and located in EC cells. Mechanical stimuli, nutrients, luminal bacteria, and neurotransmitters such as acetylcholine and norepinephrine are all able to stimulate EC cells. Paracrine actions of 5-HT allow the mucosa to signal to neurons to initiate peristaltic and secretory reflexes as well as to inflammatory cells to promote intestinal inflammation. Endocrine effects of 5-HT allow EC cells to influence distant organs, including bone, liver, and endocrine pancreas. The smaller enteric 5-HT store is biosynthetically dependent upon TPH2 and is located within a small subset of myenteric neurons. 5-HT is responsible for slow excitatory neurotransmission manifested primarily in type II/AH neurons. Importantly, neuronal 5-HT also promotes enteric nervous system (ENS) neurogenesis, both pre- and postnatally, through 5-HT2B and especially 5-HT4 receptors. The early birth of serotonergic neurons allows these cells to function as sculptors of the mature ENS. The inactivation of secreted 5-HT depends on transmembrane transport mediated by a serotonin transporter (SERT; SLC6A4). The importance of SERT in control of 5-HT's function means that pharmacological inhibition of SERT, as well as gain- or loss-of-function mutations in SLC6A4, can exert profound effects on development and function of the ENS. Extra-enteric, TPH1-derived 5-HT from yolk sac and placenta promotes neurogenesis before enteric neurons synthesize 5-HT and contribute to ENS patterning. The impressive multi-functional nature of enteric 5-HT has made the precise identification of individual physiological roles difficult and sometimes controversial.


Asunto(s)
Sistema Nervioso Entérico , Serotonina , Sistema Nervioso Entérico/fisiología , Motilidad Gastrointestinal/fisiología , Intestino Delgado , Neuronas , Serotonina/farmacología , Humanos
6.
Int J Mol Sci ; 22(24)2021 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-34948356

RESUMEN

Neurodegenerative diseases such as Alzheimer's disease (AD) have long been acknowledged as mere disorders of the central nervous system (CNS). However, in recent years the gut with its autonomous nervous system and the multitude of microbial commensals has come into focus. Changes in gut properties have been described in patients and animal disease models such as altered enzyme secretion or architecture of the enteric nervous system. The underlying cellular mechanisms have so far only been poorly investigated. An important organelle for integrating potentially toxic signals such as the AD characteristic A-beta peptide is the primary cilium. This microtubule-based signaling organelle regulates numerous cellular processes. Even though the role of primary cilia in a variety of developmental and disease processes has recently been recognized, the contribution of defective ciliary signaling to neurodegenerative diseases such as AD, however, has not been investigated in detail so far. The AD mouse model 5xFAD was used to analyze possible changes in gut functionality by organ bath measurement of peristalsis movement. Subsequently, we cultured primary enteric neurons from mutant mice and wild type littermate controls and assessed for cellular pathomechanisms. Neurite mass was quantified within transwell culturing experiments. Using a combination of different markers for the primary cilium, cilia number and length were determined using fluorescence microscopy. 5xFAD mice showed altered gut anatomy, motility, and neurite mass of enteric neurons. Moreover, primary cilia could be demonstrated on the surface of enteric neurons and exhibited an elongated phenotype in 5xFAD mice. In parallel, we observed reduced ß-Catenin expression, a key signaling molecule that regulates Wnt signaling, which is regulated in part via ciliary associated mechanisms. Both results could be recapitulated via in vitro treatments of enteric neurons from wild type mice with A-beta. So far, only a few reports on the probable role of primary cilia in AD can be found. Here, we reveal for the first time an architectural altered phenotype of primary cilia in the enteric nervous system of AD model mice, elicited potentially by neurotoxic A-beta. Potential changes on the sub-organelle level-also in CNS-derived neurons-require further investigations.


Asunto(s)
Enfermedad de Alzheimer/patología , Cilios/patología , Neuronas/patología , Enfermedad de Alzheimer/genética , Animales , Cilios/genética , Modelos Animales de Enfermedad , Sistema Nervioso Entérico/metabolismo , Sistema Nervioso Entérico/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Neuronas/metabolismo
7.
BMC Vet Res ; 14(1): 169, 2018 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-29793486

RESUMEN

BACKGROUND: The enteric nervous system (ENS), located in the intestinal wall and characterized by considerable independence from the central nervous system, consists of millions of cells. Enteric neurons control the majority of functions of the gastrointestinal tract using a wide range of substances, which are neuromediators and/or neuromodulators. One of them is leucine-enkephalin (leuENK), which belongs to the endogenous opioid family. It is known that opioids in the gastrointestinal tract have various functions, including visceral pain conduction, intestinal motility and secretion and immune processes, but many aspects of distribution and function of leuENK in the ENS, especially during pathological states, remain unknown. RESULTS: During this experiment, the distribution of leuENK - like immunoreactive (leuENK-LI) nervous structures using the immunofluorescence technique were studied in the porcine colon in physiological conditions, during chemically-induced inflammation and after axotomy. The study included the circular muscle layer, myenteric (MP), outer submucous (OSP) and inner submucous plexus (ISP) and the mucosal layer. In control animals, the number of leuENK-LI neurons amounted to 4.86 ± 0.17%, 2.86 ± 0.28% and 1.07 ± 0.08% in the MP, OSP and ISP, respectively. Generally, both pathological stimuli caused an increase in the number of detected leuENK-LI cells, but the intensity of the observed changes depended on the factor studied and part of the ENS. The percentage of leuENK-LI perikarya amounted to 11.48 ± 0.96%, 8.71 ± 0.13% and 9.40 ± 0.76% during colitis, and 6.90 ± 0.52% 8.46 ± 12% and 4.48 ± 0.44% after axotomy in MP, OSP and ISP, respectively. Both processes also resulted in an increase in the number of leuENK-LI nerves in the circular muscle layer, whereas changes were less visible in the mucosa during inflammation and axotomy did not change the number of leuENK-LI mucosal fibers. CONCLUSIONS: LeuENK in the ENS takes part in intestinal regulatory processes not only in physiological conditions, but also under pathological factors. The observed changes are probably connected with the participation of leuENK in sensory and motor innervation and the neuroprotective effects of this substance. Differences in the number of leuENK-LI neurons during inflammation and after axotomy may suggest that the exact functions of leuENK probably depend on the type of pathological factor acting on the intestine.


Asunto(s)
Colitis/veterinaria , Colon Descendente/metabolismo , Encefalina Leucina/metabolismo , Enfermedades de los Porcinos/metabolismo , Animales , Axotomía/veterinaria , Colitis/metabolismo , Colitis/fisiopatología , Colon Descendente/inervación , Colon Descendente/fisiología , Encefalina Leucina/fisiología , Femenino , Técnica del Anticuerpo Fluorescente/veterinaria , Porcinos , Enfermedades de los Porcinos/fisiopatología
8.
Gastroenterology ; 151(5): 836-844, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27521479

RESUMEN

The gastrointestinal (GI) tract is essential for the absorption of nutrients, induction of mucosal and systemic immune responses, and maintenance of a healthy gut microbiota. Key aspects of gastrointestinal physiology are controlled by the enteric nervous system (ENS), which is composed of neurons and glial cells. The ENS is exposed to and interacts with the outer (microbiota, metabolites, and nutrients) and inner (immune cells and stromal cells) microenvironment of the gut. Although the cellular blueprint of the ENS is mostly in place by birth, the functional maturation of intestinal neural networks is completed within the microenvironment of the postnatal gut, under the influence of gut microbiota and the mucosal immune system. Recent studies have shown the importance of molecular interactions among microbiota, enteric neurons, and immune cells for GI homeostasis. In addition to its role in GI physiology, the ENS has been associated with the pathogenesis of neurodegenerative disorders, such as Parkinson's disease, raising the possibility that microbiota-ENS interactions could offer a viable strategy for influencing the course of brain diseases. Here, we discuss recent advances on the role of microbiota and the immune system on the development and homeostasis of the ENS, a key relay station along the gut-brain axis.


Asunto(s)
Sistema Nervioso Entérico/inmunología , Sistema Nervioso Entérico/microbiología , Microbioma Gastrointestinal , Sistema Nervioso Entérico/embriología , Sistema Nervioso Entérico/fisiología , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Homeostasis/fisiología , Humanos , Mucosa Intestinal/embriología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/fisiología , Enfermedad de Parkinson/etiología
9.
Handb Exp Pharmacol ; 239: 1-16, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28176047

RESUMEN

The gastrointestinal (GI) system is responsible for the digestion and absorption of ingested food and liquids. Due to the complexity of the GI tract and the substantial volume of material that could be covered under the scope of GI physiology, this chapter briefly reviews the overall function of the GI tract, and discusses the major factors affecting GI physiology and function, including the intestinal microbiota, chronic stress, inflammation, and aging with a focus on the neural regulation of the GI tract and an emphasis on basic brain-gut interactions that serve to modulate the GI tract. GI diseases refer to diseases of the esophagus, stomach, small intestine, colon, and rectum. The major symptoms of common GI disorders include recurrent abdominal pain and bloating, heartburn, indigestion/dyspepsia, nausea and vomiting, diarrhea, and constipation. GI disorders rank among the most prevalent disorders, with the most common including esophageal and swallowing disorders, gastric and peptic ulcer disease, gastroparesis or delayed gastric emptying, irritable bowel syndrome (IBS), and inflammatory bowel disease (IBD). Many GI disorders are difficult to diagnose and their symptoms are not effectively managed. Thus, basic research is required to drive the development of novel therapeutics which are urgently needed. One approach is to enhance our understanding of gut physiology and pathophysiology especially as it relates to gut-brain communications since they have clinical relevance to a number of GI complaints and represent a therapeutic target for the treatment of conditions including inflammatory diseases of the GI tract such as IBD and functional gut disorders such as IBS.


Asunto(s)
Sistema Nervioso Entérico/fisiopatología , Enfermedades Gastrointestinales/fisiopatología , Animales , Jugo Gástrico/metabolismo , Absorción Gastrointestinal , Enfermedades Gastrointestinales/inmunología , Motilidad Gastrointestinal , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/fisiopatología , Humanos , Secreciones Intestinales/metabolismo
10.
Int J Mol Sci ; 18(2)2017 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-28178198

RESUMEN

Zinc transporter 3 (ZnT3) is a member of the solute-linked carrier 30 (SLC 30) zinc transporter family. It is closely linked to the nervous system, where it takes part in the transport of zinc ions from the cytoplasm to the synaptic vesicles. ZnT3 has also been observed in the enteric nervous system (ENS), but its reactions in response to pathological factors remain unknown. This study, based on the triple immunofluorescence technique, describes changes in ZnT3-like immunoreactive (ZnT3-LI) enteric neurons in the porcine ileum, caused by chemically-induced inflammation. The inflammatory process led to a clear increase in the percentage of neurons immunoreactive to ZnT3 in all "kinds" of intramural enteric plexuses, i.e., myenteric (MP), outer submucous (OSP) and inner submucous (ISP) plexuses. Moreover, a wide range of other active substances was noted in ZnT3-LI neurons under physiological and pathological conditions, and changes in neurochemical characterisation of ZnT3⁺ cells in response to inflammation depended on the "kind" of enteric plexus. The obtained results show that ZnT3 is present in the ENS in a relatively numerous and diversified neuronal population, not only in physiological conditions, but also during inflammation. The reasons for the observed changes are not clear; they may be connected with the functions of zinc ions and their homeostasis disturbances in pathological processes. On the other hand, they may be due to adaptive and/or neuroprotective processes within the pathologically altered gastrointestinal tract.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Sistema Nervioso Entérico/fisiología , Ileítis/genética , Ileítis/metabolismo , Íleon/fisiología , Animales , Proteínas de Transporte de Catión/genética , Neuronas Colinérgicas/metabolismo , Modelos Animales de Enfermedad , Ileítis/patología , Plexo Submucoso/fisiología , Porcinos , Transmisión Sináptica
11.
BMC Complement Altern Med ; 16(1): 392, 2016 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-27756367

RESUMEN

BACKGROUND: Electroacupuncture (EA) at acupoint ST 37 (Shangjuxu) has been used to alleviate gastrointestinal symptoms and improve gastrointestinal motility. However, the mechanisms by which EA affects the enteric nervous system (ENS) have scarcely been investigated. In this study, we investigated whether EA could improve ENS function. METHODS: A constipation model was established by gastric instillation of ice-cold saline daily for 14 days. The constipated mice were divided into two groups: the model group, which was not treated, and the EA group, which received EA at ST 37 at a frequency of 2-15 HZ and an amplitude of 1 mA for 15 min a day for 3 days. A further six mice were included as a non-constipated control group. After EA treatment, intestinal propulsion and defecation time were measured. Additionally, in jejunum, ileum and proximal colon myenteric plexus, the expressions of PGP9.5 and nNOS were measured by immunohistochemistry. RESULTS: The EA group demonstrated significant improvements in carbon propulsion rates and defecation time compared to model group (P < 0.05). In addition, after EA, the PGP9.5 and nNOS expression in jejunum, ileum and proximal colonic myenteric plexus was back to normal levels. CONCLUSION: This study suggests that EA stimulation at ST 37 is capable of ameliorating intestinal motility dysfunction, and can partly restore enteric neuron function. The ENS can participate in changes in intestinal motility by affecting inhibitory neurons.


Asunto(s)
Puntos de Acupuntura , Estreñimiento/terapia , Electroacupuntura/métodos , Motilidad Gastrointestinal/fisiología , Animales , Modelos Animales de Enfermedad , Mucosa Intestinal/metabolismo , Intestinos/química , Intestinos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo I/metabolismo
12.
Biomolecules ; 14(8)2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-39199372

RESUMEN

Pregnancy is a particularly vulnerable period for the growing fetus, when exposure to toxic agents, especially in the early phases, can decisively harm embryo development and compromise the future health of the newborn. The inclusion of various chemical substances in personal care products (PCPs) and cosmetic formulations can be associated with disruption and damage to the nervous system. Microplastics, benzophenones, parabens, phthalates and metals are among the most common chemical substances found in cosmetics that have been shown to induce neurotoxic mechanisms. Although cosmetic neurotoxin exposure is believed to be minimal, different exposure scenarios of cosmetics suggest that these neurotoxins remain a threat. Special attention should be paid to early exposure in the first weeks of gestation, when critical processes, like the migration and proliferation of the neural crest derived cells, start to form the ENS. Importantly, cosmetic neurotoxins can cross the placental barrier and affect the future embryo, but they are also secreted in breast milk, so babies remain exposed for longer periods, even after birth. In this review, we explore how neurotoxins contained in cosmetics and PCPs may have a role in the pathogenesis of various neurodevelopmental disorders and neurodegenerative diseases and, therefore, also in congenital enteric aganglionosis as well as in postnatal motility disorders. Understanding the mechanisms of these chemicals used in cosmetic formulations and their role in neurotoxicity is crucial to determining the safety of use for cosmetic products during pregnancy.


Asunto(s)
Cosméticos , Humanos , Femenino , Embarazo , Cosméticos/efectos adversos , Neurotoxinas/toxicidad , Síndromes de Neurotoxicidad/etiología , Ácidos Ftálicos/toxicidad , Animales
13.
Biomolecules ; 14(8)2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39199380

RESUMEN

Hirschsprung's disease (HSCR, incidence 1/5000 live births) is caused by the failure of neural crest-derived precursors to migrate, survive, proliferate, or differentiate during the embryonic development of the Enteric Nervous System (ENS), which could be disrupted by many factors, including inflammatory processes. The NF-κB family controls several biological processes, including inflammation, neurogenesis, and cell migration. With the aim of studying the potential role of NF-κB in HSCR, we have analyzed the expression of the NF-κB main subunits and other NF-κB-related genes by RT-qPCR in HSCR tissue samples (sub-divided into ganglionic and aganglionic segments). We found decreased gene expression of the NF-κB main subunit RELA but also of NFKBIA, TNFA, TFGBR2, and ERBB3 in the pathologic distal aganglionic segments compared to the proximal ganglionic segments. Moreover, we could also confirm the lower protein expression of RelA/p65 in the aganglionic distal segments by immunofluorescence staining. Further, we show that the expression of RelA/p65 protein in the proximal segments concurs with lymphocyte infiltration in the bowel tissue, indicating a pro-inflammatory activation of p65 in the proximal ganglionic HSCR tissue in the patients analyzed. All in all, our findings suggest that the modulation of NF-κB signaling in the neuro-enteric system does obviously contribute to the pathological effects of HSCR.


Asunto(s)
Enfermedad de Hirschsprung , Inflamación , FN-kappa B , Transducción de Señal , Factor de Transcripción ReIA , Enfermedad de Hirschsprung/metabolismo , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/genética , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , FN-kappa B/metabolismo , Femenino , Masculino , Lactante
14.
J Neurosci Methods ; 407: 110144, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38670535

RESUMEN

BACKGROUND: The enteric nervous system (ENS) is comprised of neurons, glia, and neural progenitor cells that regulate essential gastrointestinal functions. Advances in high-efficiency enteric neuron culture would facilitate discoveries surrounding ENS regulatory processes, pathophysiology, and therapeutics. NEW METHOD: Development of a simple, robust, one-step method to culture murine enteric neurospheres in a 3D matrix that supports neural growth and differentiation. RESULTS: Myenteric plexus cells isolated from the entire length of adult murine small intestine formed ≥3000 neurospheres within 7 days. Matrigel-embedded neurospheres exhibited abundant neural stem and progenitor cells expressing Sox2, Sox10 and Msi1 by day 4. By day 5, neural progenitor cell marker Nestin appeared in the periphery of neurospheres prior to differentiation. Neurospheres produced extensive neurons and neurites, confirmed by Tubulin beta III, PGP9.5, HuD/C, and NeuN immunofluorescence, including neural subtypes Calretinin, ChAT, and nNOS following 8 days of differentiation. Individual neurons within and external to neurospheres generated depolarization induced action potentials which were inhibited in the presence of sodium channel blocker, Tetrodotoxin. Differentiated neurospheres also contained a limited number of glia and endothelial cells. COMPARISON WITH EXISTING METHODS: This novel one-step neurosphere growth and differentiation culture system, in 3D format (in the presence of GDNF, EGF, and FGF2), allows for ∼2-fold increase in neurosphere count in the derivation of enteric neurons with measurable action potentials. CONCLUSION: Our method describes a novel, robust 3D culture of electrophysiologically active enteric neurons from adult myenteric neural stem and progenitor cells.


Asunto(s)
Plexo Mientérico , Neuronas , Animales , Plexo Mientérico/citología , Plexo Mientérico/fisiología , Neuronas/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Célula/métodos , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Células-Madre Neurales/efectos de los fármacos , Diferenciación Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Células Cultivadas , Potenciales de Acción/fisiología , Potenciales de Acción/efectos de los fármacos , Laminina/farmacología , Combinación de Medicamentos , Proteoglicanos/farmacología , Masculino , Neurogénesis/fisiología , Neurogénesis/efectos de los fármacos , Colágeno
15.
Cells ; 13(10)2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38786037

RESUMEN

Intestinal homeostasis results from the proper interplay among epithelial cells, the enteric nervous system (ENS), interstitial cells of Cajal (ICCs), smooth muscle cells, the immune system, and the microbiota. The disruption of this balance underpins the onset of gastrointestinal-related diseases. The scarcity of models replicating the intricate interplay between the ENS and the intestinal epithelium highlights the imperative for developing novel methods. We have pioneered a sophisticated tridimensional in vitro technique, coculturing small intestinal organoids with myenteric and submucosal neurons. Notably, we have made significant advances in (1) refining the isolation technique for culturing the myenteric plexus, (2) enhancing the isolation of the submucosal plexus-both yielding mixed cultures of enteric neurons and glial cells from both plexuses, and (3) subsequently co-culturing myenteric and submucosal neurons with small intestinal organoids. This co-culture system establishes neural innervations with intestinal organoids, allowing for the investigation of regulatory interactions in the context of gastrointestinal diseases. Furthermore, we have developed a method for microinjecting the luminal space of small intestinal organoids with fluorescently labeled compounds. This technique possesses broad applicability such as the assessment of intestinal permeability, transcytosis, and immunocytochemical and immunofluorescence applications. This microinjection method could be extended to alternative experimental setups, incorporating bacterial species, or applying treatments to study ENS-small intestinal epithelium interactions. Therefore, this technique serves as a valuable tool for evaluating the intricate interplay between neuronal and intestinal epithelial cells (IECs) and shows great potential for drug screening, gene editing, the development of novel therapies, the modeling of infectious diseases, and significant advances in regenerative medicine. The co-culture establishment process spans twelve days, making it a powerful asset for comprehensive research in this critical field.


Asunto(s)
Técnicas de Cocultivo , Intestino Delgado , Plexo Mientérico , Organoides , Animales , Ratones , Técnicas de Cocultivo/métodos , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/citología , Intestino Delgado/citología , Plexo Mientérico/citología , Neuronas/citología , Neuronas/metabolismo , Organoides/citología , Plexo Submucoso/citología
16.
Cells ; 13(10)2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38786042

RESUMEN

This review addresses the need for innovative co-culture systems integrating the enteric nervous system (ENS) with intestinal organoids. The breakthroughs achieved through these techniques will pave the way for a transformative era in gastrointestinal (GI) disease modeling and treatment strategies. This review serves as an introduction to the companion protocol paper featured in this journal. The protocol outlines the isolation and co-culture of myenteric and submucosal neurons with small intestinal organoids. This review provides an overview of the intestinal organoid culture field to establish a solid foundation for effective protocol application. Remarkably, the ENS surpasses the number of neurons in the spinal cord. Referred to as the "second brain", the ENS orchestrates pivotal roles in GI functions, including motility, blood flow, and secretion. The ENS is organized into myenteric and submucosal plexuses. These plexuses house diverse subtypes of neurons. Due to its proximity to the gut musculature and its cell type complexity, there are methodological intricacies in studying the ENS. Diverse approaches such as primary cell cultures, three-dimensional (3D) neurospheres, and induced ENS cells offer diverse insights into the multifaceted functionality of the ENS. The ENS exhibits dynamic interactions with the intestinal epithelium, the muscle layer, and the immune system, influencing epithelial physiology, motility, immune responses, and the microbiome. Neurotransmitters, including acetylcholine (ACh), serotonin (5-HT), and vasoactive intestinal peptide (VIP), play pivotal roles in these intricate interactions. Understanding these dynamics is imperative, as the ENS is implicated in various diseases, ranging from neuropathies to GI disorders and neurodegenerative diseases. The emergence of organoid technology presents an unprecedented opportunity to study ENS interactions within the complex milieu of the small and large intestines. This manuscript underscores the urgent need for standardized protocols and advanced techniques to unravel the complexities of the ENS and its dynamic relationship with the gut ecosystem. The insights gleaned from such endeavors hold the potential to revolutionize GI disease modeling and treatment paradigms.


Asunto(s)
Técnicas de Cocultivo , Sistema Nervioso Entérico , Enfermedades Gastrointestinales , Organoides , Humanos , Técnicas de Cocultivo/métodos , Enfermedades Gastrointestinales/patología , Enfermedades Gastrointestinales/terapia , Animales , Modelos Biológicos , Neuronas/metabolismo , Intestinos
17.
J Clin Med ; 13(18)2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39337066

RESUMEN

The enteric nervous system (ENS) is a fundamental component of the gastrointestinal system, composed of a vast network of neurons and glial cells. It operates autonomously but is interconnected with the central nervous system (CNS) through the vagus nerve. This communication, known as the gut-brain axis, influences the bidirectional communication between the brain and the gut. Background/Objectives: This study aimed to review neurological pathologies related to the ENS. Methods: To this end, a comprehensive literature search was conducted in the "PubMed" database. Articles available in "free format" were selected, applying the filters "Humans" and limiting the search to publications from the last ten years. Results: The ENS has been linked to various neurological diseases, from autism spectrum disorder to Parkinson's disease including neurological infection with the varicella zoster virus (VZV), even sharing pathologies with the CNS. This finding suggests that the ENS could serve as an early diagnostic marker or therapeutic target for neurological diseases. Gastrointestinal symptoms often precede CNS symptoms, and the ENS's accessibility aids in diagnosis and treatment. Parkinson's patients may show intestinal lesions up to twenty years before CNS symptoms, underscoring the potential for early diagnosis. However, challenges include developing standardized diagnostic protocols and the uneven distribution of dopaminergic neurons in the ENS. Continued research is needed to explore the ENS's potential in improving disease prognosis. Conclusions: The ENS is a promising area for early diagnosis and therapeutic development. Nevertheless, it is essential to continue research in this area, especially to gain a deeper understanding of its organization, function, and regenerative capacity.

18.
Eur J Med Res ; 28(1): 485, 2023 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-37932838

RESUMEN

In recent years, the role of nobiletin in neuronal disorders has received extensive attention. However, the study of nobiletin in the peripheral nervous system is limited. Nobiletin, as a compound with high fat solubility, high bioavailability and low toxicity, has been extensively studied. Accumulating scientific evidence has shown that nobiletin has a variety of biological functions in the nervous system, such as inhibiting the expression of inflammatory factors, reducing the neurotoxic response, improving the antioxidant capacity, promoting the survival of nerve cells, promoting axon growth, reducing blood‒brain barrier permeability, reducing brain oedema, promoting cAMP response element binding protein expression, improving memory, and promoting mild depolarization of nerve cell mitochondria to improve antioxidative stress capacity. Accumulating studies have shown that nobiletin also protects enteric nervous system, spinal cord and sciatic nerve. To explore the new therapeutic potential of nobiletin in the nervous system, recent and relevant research progress is reviewed in this article. This will provide a new research idea for nobiletin in the nervous system.


Asunto(s)
Flavonas , Enfermedades del Sistema Nervioso Periférico , Humanos , Flavonas/química , Flavonas/farmacología , Antioxidantes , Estrés Oxidativo
19.
Front Cell Neurosci ; 17: 1175614, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37293625

RESUMEN

Recently, the myelin proteolipid protein gene (Plp1) was shown to be expressed in the glia of the enteric nervous system (ENS) in mouse. However, beyond this, not much is known about its expression in the intestine. To address this matter, we investigated Plp1 expression at the mRNA and protein levels in the intestine of mice at different ages (postnatal days 2, 9, 21, and 88). In this study, we show that Plp1 expression preferentially occurs during early postnatal development, primarily as the DM20 isoform. Western blot analysis indicated that DM20 migrated according to its formula weight when isolated from the intestine. However, mobilities of both PLP and DM20 were faster than expected when procured from the brain. The 6.2hPLP(+)Z/FL transgene, which uses the first half of the human PLP1 gene to drive expression of a lacZ reporter gene, recapitulated the developmental pattern observed with the native gene in the intestine, indicating that it can be used as a proxy for Plp1 gene expression. As such, the relative levels of ß-galactosidase (ß-gal) activity emanating from the 6.2hPLP(+)Z/FL transgene suggest that Plp1 expression is highest in the duodenum, and decreases successively along the segments, toward the colon. Moreover, removal of the wmN1 enhancer region from the transgene (located within Plp1 intron 1) resulted in a dramatic reduction in both transgene mRNA levels and ß-gal activity in the intestine, throughout development, suggesting that this region contains a regulatory element crucial for Plp1 expression. This is consistent with earlier studies in both the central and peripheral nervous systems, indicating that it may be a common (if not universal) means by which Plp1 gene expression is governed.

20.
Mol Neurodegener ; 18(1): 32, 2023 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-37173733

RESUMEN

BACKGROUND: Parkinson's disease (PD) is a progressive neurodegenerative disorder associated with a loss of dopaminergic (DA) neurons. Despite symptomatic therapies, there is currently no disease-modifying treatment to halt neuronal loss in PD. A major hurdle for developing and testing such curative therapies results from the fact that most DA neurons are already lost at the time of the clinical diagnosis, rendering them inaccessible to therapy. Understanding the early pathological changes that precede Lewy body pathology (LBP) and cell loss in PD will likely support the identification of novel diagnostic and therapeutic strategies and help to differentiate LBP-dependent and -independent alterations. Several previous studies identified such specific molecular and cellular changes that occur prior to the appearance of Lewy bodies (LBs) in DA neurons, but a concise map of such early disease events is currently missing. METHODS: Here, we conducted a literature review to identify and discuss the results of previous studies that investigated cases with incidental Lewy body disease (iLBD), a presumed pathological precursor of PD. RESULTS: Collectively, our review demonstrates numerous cellular and molecular neuropathological changes occurring prior to the appearance of LBs in DA neurons. CONCLUSIONS: Our review provides the reader with a summary of early pathological events in PD that may support the identification of novel therapeutic and diagnostic targets and aid to the development of disease-modifying strategies in PD.


Asunto(s)
Enfermedad por Cuerpos de Lewy , Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/patología , Cuerpos de Lewy/patología , Enfermedad por Cuerpos de Lewy/patología , Degeneración Nerviosa/patología , Neuropatología , alfa-Sinucleína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA