Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.280
Filter
1.
Science ; 382(6670): 527-528, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37917691

ABSTRACT

Glial cells in the gut are specialized to fine-tune intestinal function.


Subject(s)
Intestines , Neuroglia , Neuroglia/physiology , Intestines/innervation , Intestines/physiology , Intestines/ultrastructure , Animals , Mice , Myenteric Plexus/physiology , Myenteric Plexus/ultrastructure
2.
Med Sci Monit ; 29: e939695, 2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37876155

ABSTRACT

BACKGROUND Neurological bowel dysfunction (NBD) due to spinal cord injuries (SCIs) is common and significantly impacts patients' quality of life. This study evaluated the efficacy of quantitative assessment-based nursing interventions on bowel function recovery, quality of life, and caregivers' satisfaction with SCI patients with NBD. MATERIAL AND METHODS The study included 418 SCI patients with NBD. Patients were categorized into 3 cohorts: quantitative assessment-based nursing intervention (QN, n=114), conventional nursing intervention (CN, n=125), or no nursing intervention (DN, n=189). The 3 cohorts were followed over a 6-month period. RESULTS At 6 months post-intervention, patients in the QN and CN cohorts showed significant reductions in symptoms of fecal incontinence, constipation, and abdominal distension compared to the DN cohort. Additionally, defecation time decreased significantly in the QN and CN cohorts compared to both initial measures and the DN cohort. Notably, patients in the QN cohort demonstrated substantial improvement in overall quality of life scores compared to baseline, CN, and DN cohorts. The QN cohort also reported marked improvement in caregivers' satisfaction, surpassing that of caregivers in the CN and DN cohorts. CONCLUSIONS Six months of quantitative assessment-based nursing interventions significantly improved bowel function, quality of life, and caregiver satisfaction in SCI patients with NBD. This intervention appears beneficial for managing NBD in SCI patients and improving their quality of life and caregiver satisfaction.


Subject(s)
Intestinal Diseases , Quality of Life , Spinal Cord Injuries , Humans , East Asian People , Intestines/innervation , Intestines/physiopathology , Recovery of Function , Spinal Cord Injuries/complications , Intestinal Diseases/etiology , Intestinal Diseases/therapy
3.
Nature ; 618(7966): 818-826, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37316669

ABSTRACT

Correct development and maturation of the enteric nervous system (ENS) is critical for survival1. At birth, the ENS is immature and requires considerable refinement to exert its functions in adulthood2. Here we demonstrate that resident macrophages of the muscularis externa (MMϕ) refine the ENS early in life by pruning synapses and phagocytosing enteric neurons. Depletion of MMϕ before weaning disrupts this process and results in abnormal intestinal transit. After weaning, MMϕ continue to interact closely with the ENS and acquire a neurosupportive phenotype. The latter is instructed by transforming growth factor-ß produced by the ENS; depletion of the ENS and disruption of transforming growth factor-ß signalling result in a decrease in neuron-associated MMϕ associated with loss of enteric neurons and altered intestinal transit. These findings introduce a new reciprocal cell-cell communication responsible for maintenance of the ENS and indicate that the ENS, similarly to the brain, is shaped and maintained by a dedicated population of resident macrophages that adapts its phenotype and transcriptome to the timely needs of the ENS niche.


Subject(s)
Enteric Nervous System , Intestines , Macrophages , Enteric Nervous System/cytology , Enteric Nervous System/growth & development , Enteric Nervous System/physiology , Intestines/innervation , Lymphotoxin-alpha/metabolism , Macrophages/metabolism , Macrophages/physiology , Neurons/physiology , Weaning , Cell Communication , Transcriptome , Phenotype , Phagocytosis , Synapses , Neuronal Plasticity , Gastrointestinal Transit
5.
Biomolecules ; 12(12)2022 11 27.
Article in English | MEDLINE | ID: mdl-36551197

ABSTRACT

Galanin (GAL) is an important neurotransmitter released by the enteric nervous system (ENS) neurons located in the muscularis externa and submucosa enteric plexuses that acts by binding to GAL receptors 1, 2 and 3 (GALR1, 2 and 3). In our previous studies, the GAL immunoexpression was compared in colorectal cancer (CRC) tissue and the adjacent parts of the large intestine wall including myenteric and submucosal plexuses. Recently we have also found that expression levels of GALR1 and GALR3 proteins are elevated in CRC tissue as compared with their expression in epithelial cells of unchanged mucosa. Moreover, higher GALR3 immunoreactivity in CRC cells correlated with better prognosis of CRC patients. To understand the distribution of GALRs in enteric plexuses distal and close to CRC invasion, in the present study we decided to evaluate GALRs expression within the myenteric and submucosal plexuses located proximally and distally to the cancer invasion and correlated the GALRs expression levels with the clinico-pathological data of CRC patients. The immunohistochemical and immunofluorescent methods showed only slightly decreased immunoexpression of GALR1 and GALR3 in myenteric plexuses close to cancer but did not reveal any correlation in the immunoexpression of all three GAL receptors in myenteric plexuses and tumour progression. No significant changes were found between the expression levels of GALRs in submucosal plexuses distal and close to the tumour. However, elevated GALR1 expression in submucosal plexuses in vicinity of CRC correlated with poor prognosis, higher tumour grading and shorter overall survival. When myenteric plexuses undergo morphological and functional alterations characteristic for atrophy, GALRs maintain or only slightly decrease their expression status. In contrast, the correlation between high expression of GALR1 in the submucosal plexuses and overall survival of CRC patients suggest that GAL and GALRs can act as a components of local neuro-paracrine pro-proliferative pathways accelerating the invasion and metastasis of cancer cell. The obtained results suggest an important role of GALR1 in submucosal plexuses function during the progression of CRC and imply that GALR1 expression in submucosal plexuses of ENS could be an important predictive factor for CRC progression.


Subject(s)
Colorectal Neoplasms , Myenteric Plexus , Receptor, Galanin, Type 1 , Receptor, Galanin, Type 2 , Receptor, Galanin, Type 3 , Humans , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Intestines/innervation , Myenteric Plexus/metabolism , Receptor, Galanin, Type 1/metabolism , Receptor, Galanin, Type 2/metabolism , Receptor, Galanin, Type 3/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis
6.
Nature ; 610(7933): 722-730, 2022 10.
Article in English | MEDLINE | ID: mdl-36070796

ABSTRACT

The perception of fat evokes strong appetitive and consummatory responses1. Here we show that fat stimuli can induce behavioural attraction even in the absence of a functional taste system2,3. We demonstrate that fat acts after ingestion via the gut-brain axis to drive preference for fat. Using single-cell data, we identified the vagal neurons responding to intestinal delivery of fat, and showed that genetic silencing of this gut-to-brain circuit abolished the development of fat preference. Next, we compared the gut-to-brain pathways driving preference for fat versus sugar4, and uncovered two parallel systems, one functioning as a general sensor of essential nutrients, responding to intestinal stimulation with sugar, fat and amino acids, whereas the other is activated only by fat stimuli. Finally, we engineered mice lacking candidate receptors to detect the presence of intestinal fat, and validated their role as the mediators of gut-to-brain fat-evoked responses. Together, these findings reveal distinct cells and receptors that use the gut-brain axis as a fundamental conduit for the development of fat preference.


Subject(s)
Brain-Gut Axis , Brain , Food Preferences , Intestines , Neurons , Animals , Mice , Amino Acids/metabolism , Brain/cytology , Brain/physiology , Neurons/metabolism , Sugars/metabolism , Vagus Nerve/cytology , Vagus Nerve/physiology , Food Preferences/physiology , Single-Cell Analysis , Brain-Gut Axis/genetics , Brain-Gut Axis/physiology , Intestines/innervation , Intestines/metabolism
7.
Sci Rep ; 12(1): 2018, 2022 02 07.
Article in English | MEDLINE | ID: mdl-35132112

ABSTRACT

Zebrafish larval gut could be considered as an excellent model to study functions of vertebrate digestive organs, by virtue of its simplicity and transparency as well as the availability of mutants. However, there has been scant investigation of the detailed behavior of muscular and enteric nervous systems to convey bolus, an aggregate of digested food. Here we visualized peristalsis using transgenic lines expressing a genetically encoded Ca2+ sensor in the circular smooth muscles. An intermittent Ca2+ signal cycle was observed at the oral side of the bolus, with Ca2+ waves descending and ascending from there. We also identified a regular cycle of weaker movement that occurs regardless of the presence or absence of bolus, corresponding likely to slow waves. Direct photo-stimulation of circular smooth muscles expressing ChR2 could cause local constriction of the gut, while the stimulation of a single or a few neurons could cause the local induction or arrest of gut movements. These results indicate that the larval gut of zebrafish has basic features found in adult mammals despite the small number of enteric neurons, providing a foundation for the study, at the single-cell level in vivo, in controlling the gut behaviors in vertebrates.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Intestines/physiology , Larva , Muscle, Smooth/physiology , Peristalsis/physiology , Zebrafish , Animals , Animals, Genetically Modified , Intestines/innervation , Models, Animal , Muscle Contraction , Muscle, Smooth/metabolism , Neurons/physiology , Photic Stimulation
8.
Nature ; 602(7897): 468-474, 2022 02.
Article in English | MEDLINE | ID: mdl-35082448

ABSTRACT

Ingested food and water stimulate sensory systems in the oropharyngeal and gastrointestinal areas before absorption1,2. These sensory signals modulate brain appetite circuits in a feed-forward manner3-5. Emerging evidence suggests that osmolality sensing in the gut rapidly inhibits thirst neurons upon water intake. Nevertheless, it remains unclear how peripheral sensory neurons detect visceral osmolality changes, and how they modulate thirst. Here we use optical and electrical recording combined with genetic approaches to visualize osmolality responses from sensory ganglion neurons. Gut hypotonic stimuli activate a dedicated vagal population distinct from mechanical-, hypertonic- or nutrient-sensitive neurons. We demonstrate that hypotonic responses are mediated by vagal afferents innervating the hepatic portal area (HPA), through which most water and nutrients are absorbed. Eliminating sensory inputs from this area selectively abolished hypotonic but not mechanical responses in vagal neurons. Recording from forebrain thirst neurons and behavioural analyses show that HPA-derived osmolality signals are required for feed-forward thirst satiation and drinking termination. Notably, HPA-innervating vagal afferents do not sense osmolality itself. Instead, these responses are mediated partly by vasoactive intestinal peptide secreted after water ingestion. Together, our results reveal visceral hypoosmolality as an important vagal sensory modality, and that intestinal osmolality change is translated into hormonal signals to regulate thirst circuit activity through the HPA pathway.


Subject(s)
Intestines , Satiation , Sensory Receptor Cells , Thirst , Ganglia, Sensory/cytology , Intestines/cytology , Intestines/innervation , Osmolar Concentration , Osmotic Pressure , Satiation/physiology , Sensory Receptor Cells/cytology , Thirst/physiology , Vagus Nerve/cytology , Vagus Nerve/physiology , Water/metabolism
9.
Sci Rep ; 11(1): 21788, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34750477

ABSTRACT

Alternative nutrient sources to fishmeal for fish feed, such as insect meals, represent a promising sustainable supply. However, the consequences for fish digestive function have not been exhaustively investigated. In the present study we evaluated the effect of partial fishmeal substitution with 10% Hermetia illucens (Hi10) larvae meal on the neuromuscular function of proximal and distal intestine in gilthead sea bream. In animals fed with insect meal, weight and growth parameters were similar to controls fed with conventional fishmeal. In addition, no anomalies in intestinal gross morphology and no overt signs of inflammation were observed. The gastrointestinal transit was significantly reduced in Hi10 fed animals. In the proximal and distal intestine longitudinal muscle, Hi10 feeding downregulated the excitatory cholinergic and serotoninergic transmission. Sodium nitroprusside-induced inhibitory relaxations increased in the proximal intestine and decreased in the distal intestine after Hi10 meal. Changes in the excitatory and inhibitory components of peristalsis were associated with adaptive changes in the chemical coding of both proximal and distal intestine myenteric plexus. However, these neuromuscular function alterations were not associated with considerable variations in morphometric growth parameters, suggesting that 10% Hi meal may represent a tolerable alternative protein source for gilthead sea bream diets.


Subject(s)
Intestines/physiology , Neuromuscular Junction/physiology , Sea Bream/physiology , Animal Feed , Animals , Diet/veterinary , Diptera , Gastrointestinal Transit/physiology , Intestines/anatomy & histology , Intestines/innervation , Muscle, Smooth/anatomy & histology , Muscle, Smooth/physiology , Sea Bream/anatomy & histology
10.
Development ; 148(22)2021 11 15.
Article in English | MEDLINE | ID: mdl-34792104

ABSTRACT

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


Subject(s)
Enteric Nervous System/metabolism , Neural Crest/metabolism , Neurons/metabolism , Wnt Proteins/genetics , Animals , Cell Differentiation/genetics , Cell Movement/genetics , Chick Embryo , Chickens/genetics , Chickens/growth & development , Digestive System/growth & development , Digestive System/metabolism , Enteric Nervous System/growth & development , Hirschsprung Disease/genetics , Hirschsprung Disease/pathology , Humans , Intestines/innervation , Neural Crest/cytology , RNA/genetics , RNA-Seq , Transcriptome/genetics , Wnt Signaling Pathway/genetics
11.
Nature ; 597(7875): 250-255, 2021 09.
Article in English | MEDLINE | ID: mdl-34497389

ABSTRACT

The cellular landscape of the human intestinal tract is dynamic throughout life, developing in utero and changing in response to functional requirements and environmental exposures. Here, to comprehensively map cell lineages, we use single-cell RNA sequencing and antigen receptor analysis of almost half a million cells from up to 5 anatomical regions in the developing and up to 11 distinct anatomical regions in the healthy paediatric and adult human gut. This reveals the existence of transcriptionally distinct BEST4 epithelial cells throughout the human intestinal tract. Furthermore, we implicate IgG sensing as a function of intestinal tuft cells. We describe neural cell populations in the developing enteric nervous system, and predict cell-type-specific expression of genes associated with Hirschsprung's disease. Finally, using a systems approach, we identify key cell players that drive the formation of secondary lymphoid tissue in early human development. We show that these programs are adopted in inflammatory bowel disease to recruit and retain immune cells at the site of inflammation. This catalogue of intestinal cells will provide new insights into cellular programs in development, homeostasis and disease.


Subject(s)
Aging , Enteric Nervous System/cytology , Fetus/cytology , Health , Intestines/cytology , Intestines/growth & development , Lymph Nodes/cytology , Lymph Nodes/growth & development , Adult , Animals , Child , Crohn Disease/pathology , Datasets as Topic , Enteric Nervous System/anatomy & histology , Enteric Nervous System/embryology , Enteric Nervous System/growth & development , Epithelial Cells/cytology , Female , Fetus/anatomy & histology , Fetus/embryology , Humans , Intestines/embryology , Intestines/innervation , Lymph Nodes/embryology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Organogenesis , Receptors, IgG/metabolism , Signal Transduction , Spatio-Temporal Analysis , Time Factors
12.
Pol J Vet Sci ; 24(2): 293-301, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34250785

ABSTRACT

The development of the enteric nervous system (ENS) is still a valid and intensely studied issue. However, literature in the field has no data on this topic in the dog. The present investigations were performed in three groups of fetuses from mongrel dogs - from the third, sixth- -seventh, and ninth week of pregnancy - and in 3-5-day-old puppies (3 specimens for each age group). The tissues (the medial parts of the duodenum, jejunum, and ileum with the cecum and a small portion of the adjacent ascending colon) were cut using a cryostat and the sections were processed for single- and double-labeling immunohistochemistry using antisera against acetylated tubulin (AcTub), vesicular acetylcholine transporter (VAChT), nitric oxide synthase (NOS), vasoactive intestinal polypeptide (VIP), galanin (GAL), neuropeptide Y (NPY), substance P (SP), and calcitonin gene-related peptide (CGRP). In the 3-week-old fetuses, some oval cells invading the gut wall were found. From the seventh week of pregnancy onwards, two different enteric ganglia were present: submucosal and myenteric. The estimated number of nerve elements in the 9-week-old fetuses was much higher than that observed in the 6-7-week-old individuals. There was no significant difference in the estimated number of nerve structures between the 9-week-old fetuses and the 3-5-day-old puppies. The colonization pattern and the development of the ENS in the canine small intestine are very similar to those observed in other mam- mals. However, a few exceptions have been confirmed, regarding the time of appearance of the VIP-, GAL-, and CGRP-immunoreactive neurons, and their distribution in different portions of the canine bowel during development.


Subject(s)
Dogs/growth & development , Fetal Development , Fetus/innervation , Immunohistochemistry/veterinary , Intestines/innervation , Animals , Female , Intestines/growth & development , Pregnancy
13.
Int J Mol Sci ; 22(7)2021 Mar 28.
Article in English | MEDLINE | ID: mdl-33800707

ABSTRACT

Over unimaginable expanses of evolutionary time, our gut microbiota have co-evolved with us, creating a symbiotic relationship in which each is utterly dependent upon the other. Far from confined to the recesses of the alimentary tract, our gut microbiota engage in complex and bi-directional communication with their host, which have far-reaching implications for overall health, wellbeing and normal physiological functioning. Amongst such communication streams, the microbiota-gut-brain axis predominates. Numerous complex mechanisms involve direct effects of the microbiota, or indirect effects through the release and absorption of the metabolic by-products of the gut microbiota. Proposed mechanisms implicate mitochondrial function, the hypothalamus-pituitary-adrenal axis, and autonomic, neuro-humeral, entero-endocrine and immunomodulatory pathways. Furthermore, dietary composition influences the relative abundance of gut microbiota species. Recent human-based data reveal that dietary effects on the gut microbiota can occur rapidly, and that our gut microbiota reflect our diet at any given time, although much inter-individual variation pertains. Although most studies on the effects of dietary macronutrients on the gut microbiota report on associations with relative changes in the abundance of particular species of bacteria, in broad terms, our modern-day animal-based Westernized diets are relatively high in fats and proteins and impoverished in fibres. This creates a perfect storm within the gut in which dysbiosis promotes localized inflammation, enhanced gut wall permeability, increased production of lipopolysaccharides, chronic endotoxemia and a resultant low-grade systemic inflammatory milieu, a harbinger of metabolic dysfunction and many modern-day chronic illnesses. Research should further focus on the colony effects of the gut microbiota on health and wellbeing, and dysbiotic effects on pathogenic pathways. Finally, we should revise our view of the gut microbiota from that of a seething mass of microbes to one of organ-status, on which our health and wellbeing utterly depends. Future guidelines on lifestyle strategies for wellbeing should integrate advice on the optimal establishment and maintenance of a healthy gut microbiota through dietary and other means. Although we are what we eat, perhaps more importantly, we are what our gut microbiota thrive on and they thrive on what we eat.


Subject(s)
Brain/physiology , Diet , Gastrointestinal Microbiome , Intestines/innervation , Intestines/physiology , Animals , Appetite , Autonomic Nervous System/embryology , Brain/metabolism , Diet, High-Fat , Dietary Fats , Dysbiosis/microbiology , Endotoxemia/microbiology , Humans , Incretins/metabolism , Inflammation , Lipopolysaccharides , Mice , Mitochondria/metabolism , Oligosaccharides/chemistry , Permeability
14.
Gastroenterology ; 160(5): 1486-1501, 2021 04.
Article in English | MEDLINE | ID: mdl-33493503

ABSTRACT

The gut-brain axis plays an important role in maintaining homeostasis. Many intrinsic and extrinsic factors influence signaling along this axis, modulating the function of both the enteric and central nervous systems. More recently the role of the microbiome as an important factor in modulating gut-brain signaling has emerged and the concept of a microbiota-gut-brain axis has been established. In this review, we highlight the role of this axis in modulating enteric and central nervous system function and how this may impact disorders such as irritable bowel syndrome and disorders of mood and affect. We examine the overlapping biological constructs that underpin these disorders with a special emphasis on the neurotransmitter serotonin, which plays a key role in both the gastrointestinal tract and in the brain. Overall, it is clear that although animal studies have shown much promise, more progress is necessary before these findings can be translated for diagnostic and therapeutic benefit in patient populations.


Subject(s)
Affect , Bacteria/growth & development , Brain/physiopathology , Central Nervous System Diseases/microbiology , Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome , Gastrointestinal Motility , Intestines/innervation , Intestines/microbiology , Animals , Bacteria/metabolism , Brain/metabolism , Central Nervous System Diseases/physiopathology , Central Nervous System Diseases/psychology , Central Nervous System Diseases/therapy , Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Diseases/physiopathology , Gastrointestinal Diseases/psychology , Gastrointestinal Diseases/therapy , Humans
15.
Hum Genet ; 140(5): 813-825, 2021 May.
Article in English | MEDLINE | ID: mdl-33433679

ABSTRACT

Hirschsprung disease (HSCR) is a neurocristopathy characterized by the absence of enteric ganglia along variable lengths of the intestine. Genetic defects play a major role in HSCR pathogenesis with nearly 50% of patients having a structural or regulatory deficiency in the major susceptibility gene RET. However, complete molecular defects remain poorly characterized in most patients. Here, we performed detailed genetic, molecular, and populational investigations of rare null mutations and modifiers at the RET locus. We first verified the pathogenicity of three RET splice site mutants (c.1879 + 1G > A, c.2607 + 5G > A and c.2608-3C > G) at the RNA level. We also identified significantly higher risk allele (genotype) frequencies, and their over-transmission, from unaffected parents to affected offspring of three functionally independent enhancer variants (rs2506030, rs7069590 and rs2435357, with odd ratios (OR) of 2.09, 2.71 and 7.59, respectively, P < 0.001). These three common variants are in significant (P < 4.64 × 10-186) linkage disequilibrium in the Han Chinese population with ~ 60% of them carrying at least one copy and > 10% with two copies. We show that RET compound inheritance of rare and common variants prevails in 64% (seven out of 11) of Chinese HSCR families. This study supports the idea that common RET variants can modify the penetrance of rare null RET mutations in HSCR, and the combined high susceptibility allele dosage may constitute the unique raised "risk baseline" among the Chinese population.


Subject(s)
Hirschsprung Disease/genetics , Intestines/innervation , Penetrance , Protein Isoforms/genetics , Proto-Oncogene Proteins c-ret/genetics , China , Female , Genetic Predisposition to Disease/genetics , Humans , Infant , Infant, Newborn , Linkage Disequilibrium/genetics , Male , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA
16.
Development ; 148(2)2021 01 26.
Article in English | MEDLINE | ID: mdl-33376126

ABSTRACT

The enteric nervous system (ENS), which is derived from neural crest, is essential for gut function, and its deficiency causes severe congenital diseases. Since the capacity for ENS regeneration in mammals is limited, additional complementary models would be useful. Here, we show that the ENS in zebrafish larvae at 10-15 days postfertilization is highly regenerative. After laser ablation, the number of enteric neurons recovered to ∼50% of the control by 10 days post-ablation (dpa). Using transgenic lines in which enteric neural crest-derived cells (ENCDCs) and enteric neurons are labeled with fluorescent proteins, we live imaged the regeneration process and found covering by neurites that extended from the unablated area and entry of ENCDCs into the ablated areas by 1-3 dpa. BrdU assays suggested that ∼80% of the enteric neurons and ∼90% of the Sox10-positive ENCDCs therein at 7 dpa were generated through proliferation. Thus, ENS regeneration involves proliferation, entrance and neurogenesis of ENCDCs. This is the first report regarding the regeneration process of the zebrafish ENS. Our findings provide a basis for further in vivo research at single-cell resolution in this vertebrate model.


Subject(s)
Cell Movement , Enteric Nervous System/cytology , Enteric Nervous System/physiology , Nerve Regeneration , Neural Crest/cytology , Animals , Animals, Genetically Modified , Axons/metabolism , Cell Proliferation , Green Fluorescent Proteins/metabolism , Intestines/innervation , Larva , Neurites/metabolism , Neurogenesis , Time Factors
17.
Int J Surg Pathol ; 29(3): 246-256, 2021 May.
Article in English | MEDLINE | ID: mdl-32964744

ABSTRACT

Gastrointestinal inflammatory neuropathy, namely, eosinophilic myenteric ganglioneuronitis (EMG) and lymphocytic ganglioneuronitis (LG), is a form of chronic intestinal pseudo-obstruction and results from the infiltration of the myenteric plexus by eosinophils and lymphocytes, respectively. The literature related to the clinicopathological features of adult inflammatory neuropathy is scarce. We aim to elucidate the clinical and histological details of 7 cases of inflammatory neuropathy (EMG, n = 4, and LG, n = 3) and compare the features of EMG and LG retrospectively. There was no difference between these two entities in terms of clinical, hematological, or biochemical parameters. Histologically, almost all cases (n = 6/7) showed accompanying elements of ganglion cell vacuolization, mesenchymopathy, and partial/complete desmosis in addition to the disease-defining pathology. Besides, all cases of EMG showed infiltration of the inner circular muscle of muscularis propria by eosinophils. Two cases of LG showed additional muscular pathology pertaining to the muscularis propria. Inflammatory infiltration of the myenteric plexus is pathognomonic for the diagnosis of gastrointestinal inflammatory neuropathy although additional features in the form of ganglion cell vacuolization, reduction in the number of ganglia, desmosis, mesenchymopathy, and inflammation of the muscularis propria (eosinophils in EMG) can be seen. The pathologists need proper awareness along with judicious use of special and immunostains for clinching the diagnosis.


Subject(s)
Eosinophilia/diagnosis , Intestinal Pseudo-Obstruction/diagnosis , Lymphocytes/immunology , Myenteric Plexus/pathology , Peripheral Nervous System Diseases/diagnosis , Adult , Aged , Aged, 80 and over , Chronic Disease , Eosinophilia/immunology , Eosinophilia/pathology , Female , Humans , Inflammation/diagnosis , Inflammation/immunology , Inflammation/pathology , Inflammation/surgery , Intestinal Pseudo-Obstruction/immunology , Intestinal Pseudo-Obstruction/pathology , Intestinal Pseudo-Obstruction/surgery , Intestines/immunology , Intestines/innervation , Intestines/pathology , Intestines/surgery , Male , Middle Aged , Myenteric Plexus/immunology , Peripheral Nervous System Diseases/immunology , Peripheral Nervous System Diseases/pathology , Peripheral Nervous System Diseases/surgery , Retrospective Studies
19.
Front Immunol ; 11: 604179, 2020.
Article in English | MEDLINE | ID: mdl-33362788

ABSTRACT

The human microbiota has a fundamental role in host physiology and pathology. Gut microbial alteration, also known as dysbiosis, is a condition associated not only with gastrointestinal disorders but also with diseases affecting other distal organs. Recently it became evident that the intestinal bacteria can affect the central nervous system (CNS) physiology and inflammation. The nervous system and the gastrointestinal tract are communicating through a bidirectional network of signaling pathways called the gut-brain axis, which consists of multiple connections, including the vagus nerve, the immune system, and bacterial metabolites and products. During dysbiosis, these pathways are dysregulated and associated with altered permeability of the blood-brain barrier (BBB) and neuroinflammation. However, numerous mechanisms behind the impact of the gut microbiota in neuro-development and -pathogenesis remain poorly understood. There are several immune pathways involved in CNS homeostasis and inflammation. Among those, the inflammasome pathway has been linked to neuroinflammatory conditions such as multiple sclerosis, Alzheimer's and Parkinson's diseases, but also anxiety and depressive-like disorders. The inflammasome complex assembles upon cell activation due to exposure to microbes, danger signals, or stress and lead to the production of pro-inflammatory cytokines (interleukin-1ß and interleukin-18) and to pyroptosis. Evidences suggest that there is a reciprocal influence of microbiota and inflammasome activation in the brain. However, how this influence is precisely working is yet to be discovered. Herein, we discuss the status of the knowledge and the open questions in the field focusing on the function of intestinal microbial metabolites or products on CNS cells during healthy and inflammatory conditions, such as multiple sclerosis, Alzheimer's and Parkinson's diseases, and also neuropsychiatric disorders. In particular, we focus on the innate inflammasome pathway as immune mechanism that can be involved in several of these conditions, upon exposure to certain microbes.


Subject(s)
Bacteria/metabolism , Brain Diseases/microbiology , Brain/metabolism , Gastrointestinal Microbiome , Inflammasomes/metabolism , Intestines/microbiology , Mental Disorders/microbiology , Animals , Bacteria/immunology , Brain/immunology , Brain/pathology , Brain/physiopathology , Brain Diseases/immunology , Brain Diseases/metabolism , Brain Diseases/physiopathology , Dysbiosis , Host-Pathogen Interactions , Humans , Inflammasomes/immunology , Intestines/immunology , Intestines/innervation , Mental Disorders/immunology , Mental Disorders/metabolism , Mental Disorders/physiopathology
20.
Eur J Histochem ; 64(3)2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32875777

ABSTRACT

Inflammatory bowel disease (IBD) includes inflammation of the gastrointestinal (GI) tract and is characterized by periods of acute inflammation and remission. Therapeutic management of IBD is still problematic, because of incomplete understanding its pathogenesis. This study focuses on the effect of 2,4,6-trinitrobenzene sulphonic acid (TNBS)-induced colitis on changes in enteric neuronal subpopulations in adult zebrafish. These changes are suggested to be related to the altered neuro-immune interactions and GI motility, and in IBD pathogenesis. New insights into neuroplasticity will be instrumental in finding appropriate therapeutic treatments. TNBS was intraluminally administered in the distal intestine (DI) of anesthetized adult zebrafish. A histological time course of the intestinal inflammatory response was created to establish optimal TNBS concentration and acute inflammation phase. Using double immunolabelling on whole mounts, the effect of inflammation on neuronal populations was analyzed. Based on intestinal wall thickening, epithelial fold disruption, reduced goblet cell number, and eosinophil infiltration, our analysis indicated that the optimal TNBS concentration (320 mM in 25% ethanol) inducing non-lethal inflammation reached a peak at 6 hours post-induction. The inflammatory response returned to baseline values at 3 days post-induction. At the acute inflammation phase, no influence on the distribution or proportion of nitrergic neurons was observed, while only the proportion of cholinergic neurons was significantly reduced in the DI. The proportion of serotonergic neurons was significantly increased in the entire intestine during inflammation. This study describes a method of TNBS-induced colitis in the adult zebrafish. Given that the acute inflammation phase is accompanied by neuroplasticity comparable to changes observed in IBD patients, and the unique and versatile characteristics of the zebrafish, allows this model to be used alongside IBD animal models to unravel IBD pathology and to test new IBD therapies.


Subject(s)
Cholinergic Neurons/drug effects , Colitis/chemically induced , Disease Models, Animal , Trinitrobenzenesulfonic Acid/adverse effects , Zebrafish , Animals , Colitis/pathology , Dose-Response Relationship, Drug , Female , Inflammation/chemically induced , Inflammation/pathology , Intestines/innervation , Intestines/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...