Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
J Clin Invest ; 134(9)2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38690732

ABSTRACT

Epigenetic regulatory mechanisms are underappreciated, yet are critical for enteric nervous system (ENS) development and maintenance. We discovered that fetal loss of the epigenetic regulator Bap1 in the ENS lineage caused severe postnatal bowel dysfunction and early death in Tyrosinase-Cre Bap1fl/fl mice. Bap1-depleted ENS appeared normal in neonates; however, by P15, Bap1-deficient enteric neurons were largely absent from the small and large intestine of Tyrosinase-Cre Bap1fl/fl mice. Bowel motility became markedly abnormal with disproportionate loss of cholinergic neurons. Single-cell RNA sequencing at P5 showed that fetal Bap1 loss in Tyrosinase-Cre Bap1fl/fl mice markedly altered the composition and relative proportions of enteric neuron subtypes. In contrast, postnatal deletion of Bap1 did not cause enteric neuron loss or impaired bowel motility. These findings suggest that BAP1 is critical for postnatal enteric neuron differentiation and for early enteric neuron survival, a finding that may be relevant to the recently described human BAP1-associated neurodevelopmental disorder.


Subject(s)
Cell Differentiation , Enteric Nervous System , Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Animals , Enteric Nervous System/metabolism , Enteric Nervous System/pathology , Mice , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Mice, Knockout , Female , Gastrointestinal Motility/genetics , Humans
2.
Expert Rev Gastroenterol Hepatol ; 17(12): 1301-1312, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38117595

ABSTRACT

INTRODUCTION: Gastrointestinal (GI) motility disorders comprise a wide range of different diseases affecting the structural or functional integrity of the GI neuromusculature. Their clinical presentation and burden of disease depends on the predominant location and extent of gut involvement as well as the component of the gut neuromusculature affected. AREAS COVERED: A comprehensive literature review was conducted using the PubMed and Medline databases to identify articles related to GI motility and functional disorders, published between 2016 and 2023. In this article, we highlight the current knowledge of molecular and genetic mechanisms underlying GI dysmotility, including disorders of gut-brain interaction, which involve both GI motor and sensory disturbance. EXPERT OPINION: Although the pathophysiology and molecular mechanisms underlying many such disorders remain unclear, recent advances in the assessment of intestinal tissue samples, genetic testing with the application of 'omics' technologies and the use of animal models will provide better insights into disease pathogenesis as well as opportunities to improve therapy.


Subject(s)
Gastrointestinal Diseases , Animals , Humans , Gastrointestinal Diseases/diagnosis , Gastrointestinal Diseases/genetics , Gastrointestinal Motility/genetics , Brain , Head
3.
Neuron ; 111(14): 2184-2200.e7, 2023 07 19.
Article in English | MEDLINE | ID: mdl-37192624

ABSTRACT

Vagal sensory neurons monitor mechanical and chemical stimuli in the gastrointestinal tract. Major efforts are underway to assign physiological functions to the many distinct subtypes of vagal sensory neurons. Here, we use genetically guided anatomical tracing, optogenetics, and electrophysiology to identify and characterize vagal sensory neuron subtypes expressing Prox2 and Runx3 in mice. We show that three of these neuronal subtypes innervate the esophagus and stomach in regionalized patterns, where they form intraganglionic laminar endings. Electrophysiological analysis revealed that they are low-threshold mechanoreceptors but possess different adaptation properties. Lastly, genetic ablation of Prox2 and Runx3 neurons demonstrated their essential roles for esophageal peristalsis in freely behaving mice. Our work defines the identity and function of the vagal neurons that provide mechanosensory feedback from the esophagus to the brain and could lead to better understanding and treatment of esophageal motility disorders.


Subject(s)
Core Binding Factor Alpha 3 Subunit , Esophagus , Gastrointestinal Motility , Homeodomain Proteins , Sensory Receptor Cells , Vagus Nerve , Animals , Mice , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Esophagus/innervation , Esophagus/metabolism , Esophagus/physiology , Gastrointestinal Motility/genetics , Gastrointestinal Motility/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mechanoreceptors/physiology , Neurons, Afferent/physiology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Stomach/innervation , Stomach/metabolism , Stomach/physiology , Vagus Nerve/physiology
4.
Dig Dis Sci ; 68(2): 439-450, 2023 02.
Article in English | MEDLINE | ID: mdl-35947306

ABSTRACT

BACKGROUND: The specific role of the M3 muscarinic acetylcholine receptor in gastrointestinal motility under physiological conditions is unclear, due to a lack of subtype-selective compounds. AIMS: The objective of this study was to determine the region-specific role of the M3 receptor in gastrointestinal motility. METHODS: We developed a novel positive allosteric modulator (PAM) for the M3 receptor, PAM-369. The effects of PAM-369 on the carbachol-induced contractile response of porcine esophageal smooth muscle and mouse colonic smooth muscle (ex vivo) and on the transit in mouse small intestine and rat colon (in vivo) were examined. RESULTS: PAM-369 selectively potentiated the M3 receptor under the stimulation of its orthosteric ligands without agonistic or antagonistic activity. Half-maximal effective concentrations of PAM activity for human, mouse, and rat M3 receptors were 0.253, 0.345, and 0.127 µM, respectively. PAM-369 enhanced carbachol-induced contraction in porcine esophageal smooth muscle and mouse colonic smooth muscle without causing any contractile responses by itself. The oral administration of 30 mg/kg PAM-369 increased the small intestinal transit in both normal motility and loperamide-induced intestinal dysmotility mice but had no effects on the colonic transit, although the M3 receptor mRNA expression is higher in the colon than in the small intestine. CONCLUSIONS: This study provided the first direct evidence that the M3 receptor has different region-specific roles in the motility function between the small intestine and colon in physiological and pathophysiological contexts. Selective PAMs designed for targeted subtypes of muscarinic receptors are useful for elucidating the subtype-specific function.


Subject(s)
Gastrointestinal Motility , Receptor, Muscarinic M3 , Animals , Humans , Mice , Rats , Carbachol/pharmacology , Gastrointestinal Motility/genetics , Gastrointestinal Motility/physiology , Muscle Contraction , Receptor, Muscarinic M2/genetics , Receptor, Muscarinic M2/metabolism , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/metabolism , Receptors, Muscarinic/physiology , Swine
5.
Adv Exp Med Biol ; 1383: 9-17, 2022.
Article in English | MEDLINE | ID: mdl-36587142

ABSTRACT

Severe gut motility disorders are characterized by ineffective propulsion of intestinal contents. As a result, patients often develop extremely uncomfortable symptoms, ranging from nausea and vomiting along with alterations of bowel habits, up to radiologically confirmed subobstructive episodes. Chronic intestinal pseudo-obstruction (CIPO) is a typical clinical phenotype of severe gut dysmotility due to morphological and functional alterations of the intrinsic (enteric) innervation and extrinsic nerve supply (hence neuropathy), interstitial cells of Cajal (ICCs) (mesenchymopathy), and smooth muscle cells (myopathy). In this chapter, we highlight some molecular mechanisms of CIPO and review the clinical phenotypes and the genetics of the different types of CIPO. Specifically, we will detail the role of some of the most representative genetic mutations involving RAD21, LIG3, and ACTG2 to provide a better understanding of CIPO and related underlying neuropathic or myopathic histopathological abnormalities. This knowledge may unveil targeted strategies to better manage patients with such severe disease.


Subject(s)
Intestinal Pseudo-Obstruction , Humans , Intestinal Pseudo-Obstruction/genetics , Intestinal Pseudo-Obstruction/diagnosis , Intestine, Small , Mutation , Chronic Disease , Gastrointestinal Motility/genetics
6.
Am J Pathol ; 192(11): 1592-1603, 2022 11.
Article in English | MEDLINE | ID: mdl-35985479

ABSTRACT

Appropriate coordination of smooth muscle contraction and relaxation is essential for normal colonic motility. The impact of perturbed motility ranges from moderate, in conditions such as colitis, to potentially fatal in the case of pseudo-obstruction. The mechanisms underlying aberrant motility and the extent to which they can be targeted pharmacologically are incompletely understood. This study identified colonic smooth muscle as a major site of expression of neuropilin 2 (Nrp2) in mice and humans. Mice with inducible smooth muscle-specific knockout of Nrp2 had an increase in evoked contraction of colonic rings in response to carbachol at 1 and 4 weeks following initiation of deletion. KCl-induced contractions were also increased at 4 weeks. Colonic motility was similarly enhanced, as evidenced by faster bead expulsion in Nrp2-deleted mice versus Nrp2-intact controls. In length-tension analysis of the distal colon, passive tension was similar in Nrp2-deficient and Nrp2-intact mice, but at low strains, active stiffness was greater in Nrp2-deficient animals. Consistent with the findings in conditional Nrp2 mice, Nrp2-null mice showed increased contractility in response to carbachol and KCl. Evaluation of selected proteins implicated in smooth muscle contraction revealed no significant differences in the level of α-smooth muscle actin, myosin light chain, calponin, or RhoA. Together, these findings identify Nrp2 as a novel regulator of colonic contractility that may be targetable in conditions characterized by dysmotility.


Subject(s)
Colon , Gastrointestinal Motility , Muscle Contraction , Muscle, Smooth , Neuropilin-2 , Animals , Humans , Mice , Carbachol/pharmacology , Colon/metabolism , Colon/physiology , Mice, Knockout , Muscle Contraction/drug effects , Muscle Contraction/genetics , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Neuropilin-2/genetics , Neuropilin-2/metabolism , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/genetics
7.
Zhen Ci Yan Jiu ; 47(2): 141-7, 2022 Feb 25.
Article in Chinese | MEDLINE | ID: mdl-35218624

ABSTRACT

OBJECTIVE: To observe the effect of electroacupuncture (EA) of "Tianshu"(ST25) and "Dachangshu"(BL25) on the intestinal transit function, expression level of glial cell-derived neurotrophic factor (GDNF) and methylation level of GDNF gene promoter region in colon tissue of rats with slow transit constipation (STC), so as to explore its mechanisms underlying improvement of STC. METHODS: Male Sprague-Dawley (SD) rats were randomized into control, saline, model and EA groups (n=16 in each group). The STC model was replicated by gavage of compound diphenoxylate suspension (10 mL· kg-1· d-1) for 28 days. Rats of the saline group received the same dose of normal saline via gavage. EA (2 Hz/15 Hz, 0.1-1 mA) was applied to bila-teral ST25 and BL25 for 15 min, once daily for 14 days. The intestinal transmission function (the intestinal propulsion rate) was assessed by recording the first black grain stool discharge time and the number and weight of the discharged stool grains in 30 min after gavage of the activated carbon suspension (1 mL/100 g, 150 g/L). The score of fecal trait and the weight of stool within 24 h were recorded. The ultrastructural changes of Cajal interstitial cells in the colon tissue were observed by transmission electron microscope. The expression levels of GDNF protein and mRNA in the colon tissue were detected by using Western blot and real-time fluorescent quantitative PCR, separately, and changes of methylation level in the promoter region of GDNF gene detected by using Bisulfite sequencing method. RESULTS: Compared with the control group, the time of the 1st black stool grain discharging was obviously prolonged, and the number and weight of the discharged black stool grains were significantly decreased in the mo-del group (P<0.05), suggesting a success of STC. The weight and trait score of stool in 24 h, intestinal propulsive rate, and the expression levels of GDNF protein and mRNA were significantly lower in the model group than in the control group (P<0.01, P<0.05). After EA, the weight and trait score of stool within 24 h, intestinal propulsive rate,and the expression levels of GDNF protein and mRNA were significantly increased in the EA group in contrast to the model group (P<0.01,P<0.05). The total CpGs methylation level of GDNF gene in colon tissue was considerably higher in the model group than in the control group (P<0.05), and markedly lower in the EA group than in the model group (P<0.05). No significant differences were found between the control and saline groups in all the above-mentioned indexes (P>0.05). CONCLUSION: EA of back-shu and front-mu acupoints can effectively improve symptoms of constipation and intestinal transport function in STC rats, which may be related to its function in up-regulating the expression of GDNF and down-regulating the methylation level in the promoter region of GDNF gene in colon tissue.


Subject(s)
Electroacupuncture , Acupuncture Points , Animals , Constipation/genetics , Constipation/therapy , Gastrointestinal Motility/genetics , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Male , Methylation , Rats , Rats, Sprague-Dawley
8.
J Clin Invest ; 132(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35166239

ABSTRACT

Gastrointestinal motility disorders involve alterations to the structure and/or function of the enteric nervous system (ENS) but the causal mechanisms remain unresolved in most cases. Homeostasis and disease in the ENS are processes that are regulated by enteric glia. Signaling mediated through type I lysophosphatidic acid receptors (LPAR1) has recently emerged as an important mechanism that contributes to disease, in part, through effects on peripheral glial survival and function. Enteric glia express LPAR1 but its role in ENS function and motility disorders is unknown. We used a combination of genetic, immunohistochemical, calcium imaging, and in vivo pharmacological approaches to investigate the role of LPAR1 in enteric glia. LPAR1 was enriched in enteric glia in mice and humans and LPA stimulated intracellular calcium responses in enteric glia, subsequently recruiting activity in a subpopulation of myenteric neurons. Blocking LPAR1 in vivo with AM966 attenuated gastrointestinal motility in mice and produced marked enteric neuro- and gliopathy. Samples from humans with chronic intestinal pseudo-obstruction (CIPO), a severe motility disorder, showed reduced glial LPAR1 expression in the colon and ileum. These data suggest that enteric glial LPAR1 signaling regulates gastrointestinal motility through enteric glia and could contribute to severe motility disorders in humans such as CIPO.


Subject(s)
Enteric Nervous System/metabolism , Intestinal Pseudo-Obstruction/metabolism , Neuroglia/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction , Adult , Aged , Animals , Chronic Disease , Female , Gastrointestinal Motility/genetics , Humans , Intestinal Pseudo-Obstruction/genetics , Intestinal Pseudo-Obstruction/physiopathology , Male , Mice , Mice, Transgenic , Middle Aged , Receptors, Lysophosphatidic Acid/genetics
9.
Med Sci Monit ; 27: e932885, 2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34845181

ABSTRACT

BACKGROUND Moxibustion therapy has been found to ameliorate clinical symptoms of functional dyspepsia (FD). We aimed to examine the regulatory effect of moxibustion on the gastrointestinal (GI) motility in FD and explore the underlying mechanism based on the hyperpolarization-activated cyclic nucleotide-gated cation channel 1 (HCN1). MATERIAL AND METHODS Moxibustion therapy was used in FD rats induced by using classic tail-pinch and irregular feeding. Weight gain and food intake were recorded weekly, followed by detecting gastric residual rate (GRR) and small intestine propulsion rate (IPR). Next, western blotting was performed to determine the expression levels of HCN1 in the gastric antrum. qRT-PCR was used to detect HCN1 in the small intestine and hypothalamic satiety center. Double immunolabeling was used for HCN1 and ICCs in gastric antrum and small intestine. RESULTS The obtained results suggested that moxibustion treatment could increase weight gain and food intake in FD rats. The GRR and IPR were compared among the groups, which showed that moxibustion treatment could decrease GRR and increase IPR. Moxibustion increased the expression of HCN1 in the gastric antrum, small intestine, and hypothalamic satiety center. Histologically, the co-expressions of HCN1 and ICCs tended to increase in gastric antrum and small intestine. Meanwhile, HCN channel inhibitor ZD7288 prevented the above-mentioned therapeutic effects of moxibustion. CONCLUSIONS The results of the present study suggest that moxibustion can effectively improve the GI motility of FD rats, which may be related to the upregulation of HCN1 expression in gastric antrum, small intestine, and satiety center.


Subject(s)
Dyspepsia/genetics , Dyspepsia/therapy , Gastrointestinal Motility/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Moxibustion/methods , Potassium Channels/genetics , Animals , Disease Models, Animal , Rats
10.
Eur J Histochem ; 65(s1)2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34818877

ABSTRACT

The enteric nervous system (ENS) is the third division of the autonomic autonomic nervous system and the largest collection of neurons outside the central nervous system (CNS). The ENS has been referred to as "the brain in the gut" or "the second brain of the human body" because of its highly integrated neural circuits controlling a vast repertoire of gut functions, including absorption/secretion, splanchnic blood vessels, some immunological aspects, intestinal epithelial barrier, and gastrointestinal (GI) motility. The latter function is the result of the ENS fine-tuning over smooth musculature, along with the contribution of other key cells, such as enteric glia (astrocyte like cells supporting and contributing to neuronal activity), interstitial cells of Cajal (the pacemaker cells of the GI tract involved in neuromuscular transmission), and enteroendocrine cells (releasing bioactive substances, which affect gut physiology). Any noxa insult perturbing the ENS complexity may determine a neuropathy with variable degree of neuro-muscular dysfunction. In this review, we aim to cover the most recent update on genetic mechanisms leading to enteric neuropathies ranging from Hirschsprung's disease (characterized by lack of any enteric neurons in the gut wall) up to more generalized form of dysmotility such as chronic intestinal pseudo-obstruction (CIPO) with a significant reduction of enteric neurons. In this line, we will discuss the role of the RAD21 mutation, which we have demonstrated in a family whose affected members exhibited severe gut dysmotility. Other genes contributing to gut motility abnormalities will also be presented. In conclusion, the knowledge on the molecular mechanisms involved in enteric neuropathy may unveil strategies to better manage patients with neurogenic gut dysmotility and pave the way to targeted therapies.


Subject(s)
Gastrointestinal Motility/genetics , Intestinal Diseases/genetics , Intestinal Pseudo-Obstruction/genetics , Animals , Gastrointestinal Motility/physiology , Humans , Intestinal Diseases/physiopathology , Intestinal Pseudo-Obstruction/physiopathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/physiopathology , Mutation , Neurons/physiology
11.
Neurogastroenterol Motil ; 33(12): e14190, 2021 12.
Article in English | MEDLINE | ID: mdl-34190380

ABSTRACT

BACKGROUND: The breakpoint cluster region (BCR) is a protein that originally forms a fusion protein with c-Abl tyrosine kinase and induces leukemia. Researchers have shown that BCR is enriched in the central nervous system and may contribute to neurological disorders. We aimed to investigate the physiological function of BCR in neural development in the gastrointestinal (GI) tract and brain. METHODS: Whole-exome sequencing was used to screen for mutations in the BCR. Bcr knockout mice (Bcr-/- , ΔExon 2-22) were generated using the CRISPR/Cas9 system. Transit of carmine red dye and glass bead expulsion assays were used to record total and proximal GI transit and distal colonic transit. KEY RESULTS: In an infant with pediatric intestinal pseudo-obstruction, we found a heterozygous de novo mutation (NM_004327.3:c.3072+1G>A) in BCR. Bcr deficiency mice (Bcr-/- ) exhibited growth retardation and impaired gastrointestinal motility. Bcr-/- mice had a prolonged average total GI transit time with increased distal colonic transit and proximal GI transit in isolation. Morphology analysis indicated that Bcr-/- mice had a less number of neurons in the submucosal plexus and myenteric plexus. Bcr-/- mice exhibited apparent structural defects in the brain, particularly in the cortex. Additionally, Bcr- depletion in the mouse cortex altered the expression of Ras homologous (Rho) family small GTPases. CONCLUSIONS AND INFERENCES: BCR mutations are associated with intestinal obstruction in children. Loss of Bcr can cause intestinal dysmotility and brain developmental defects may via regulation of Rho GTPases.


Subject(s)
Brain/metabolism , Gastrointestinal Diseases/genetics , Gastrointestinal Motility/genetics , Intestinal Pseudo-Obstruction/genetics , Proto-Oncogene Proteins c-bcr/genetics , Animals , Female , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/physiopathology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/physiopathology , Gastrointestinal Transit/genetics , Humans , Intestinal Pseudo-Obstruction/metabolism , Intestinal Pseudo-Obstruction/pathology , Mice , Mice, Knockout , Neurons/metabolism , Proto-Oncogene Proteins c-bcr/metabolism
12.
J Virol ; 95(15): e0075121, 2021 07 12.
Article in English | MEDLINE | ID: mdl-33980599

ABSTRACT

Rotavirus infection is highly prevalent in children, and the most severe effects are diarrhea and vomiting. It is well accepted that the enteric nervous system (ENS) is activated and plays an important role, but knowledge of how rotavirus activates nerves within ENS and to the vomiting center is lacking. Serotonin is released during rotavirus infection, and antagonists to the serotonin receptor subtype 3 (5-HT3 receptor) can attenuate rotavirus-induced diarrhea. In this study, we used a 5-HT3 receptor knockout (KO) mouse model to investigate the role of this receptor in rotavirus-induced diarrhea, motility, electrolyte secretion, inflammatory response, and vomiting reflex. The number of diarrhea days (P = 0.03) and the number of mice with diarrhea were lower in infected 5-HT3 receptor KO than wild-type pups. In vivo investigation of fluorescein isothiocyanate (FITC)-dextran transit time showed that intestinal motility was lower in the infected 5-HT3 receptor KO compared to wild-type mice (P = 0.0023). Ex vivo Ussing chamber measurements of potential difference across the intestinal epithelia showed no significant difference in electrolyte secretion between the two groups. Immediate early gene cFos expression level showed no difference in activation of the vomiting center in the brain. Cytokine analysis of the intestine indicated a low effect of inflammatory response in rotavirus-infected mice lacking the 5-HT3 receptor. Our findings indicate that the 5-HT3 receptor is involved in rotavirus-induced diarrhea via its effect on intestinal motility and that the vagus nerve signaling to the vomiting center occurs also in the absence of the 5-HT3 receptor. IMPORTANCE The mechanisms underlying rotavirus-induced diarrhea and vomiting are not yet fully understood. To better understand rotavirus pathophysiology, characterization of nerve signaling within the ENS and through vagal efferent nerves to the brain, which have been shown to be of great importance to the disease, is necessary. Serotonin (5-HT), a mediator of both diarrhea and vomiting, has been shown to be released from enterochromaffin cells in response to rotavirus infection and the rotavirus enterotoxin NSP4. Here, we investigated the role of the serotonin receptor 5-HT3, which is known to be involved in the nerve signals that regulate gut motility, intestinal secretion, and signal transduction through the vagus nerve to the brain. We show that the 5-HT3 receptor is involved in rotavirus-induced diarrhea by promoting intestinal motility. The findings shed light on new treatment possibilities for rotavirus diarrhea.


Subject(s)
Diarrhea/physiopathology , Enteric Nervous System/physiopathology , Gastrointestinal Motility/physiology , Receptors, Serotonin, 5-HT3/metabolism , Rotavirus Infections/pathology , Vomiting/physiopathology , Animals , Enterochromaffin Cells/metabolism , Gastrointestinal Motility/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Serotonin, 5-HT3/genetics , Rotavirus/physiology , Serotonin/metabolism , Serotonin 5-HT3 Receptor Antagonists/pharmacology
13.
Brain ; 144(5): 1451-1466, 2021 06 22.
Article in English | MEDLINE | ID: mdl-33855352

ABSTRACT

Abnormal gut motility is a feature of several mitochondrial encephalomyopathies, and mutations in genes such as TYMP and POLG, have been linked to these rare diseases. The human genome encodes three DNA ligases, of which only one, ligase III (LIG3), has a mitochondrial splice variant and is crucial for mitochondrial health. We investigated the effect of reduced LIG3 activity and resulting mitochondrial dysfunction in seven patients from three independent families, who showed the common occurrence of gut dysmotility and neurological manifestations reminiscent of mitochondrial neurogastrointestinal encephalomyopathy. DNA from these patients was subjected to whole exome sequencing. In all patients, compound heterozygous variants in a new disease gene, LIG3, were identified. All variants were predicted to have a damaging effect on the protein. The LIG3 gene encodes the only mitochondrial DNA (mtDNA) ligase and therefore plays a pivotal role in mtDNA repair and replication. In vitro assays in patient-derived cells showed a decrease in LIG3 protein levels and ligase activity. We demonstrated that the LIG3 gene defects affect mtDNA maintenance, leading to mtDNA depletion without the accumulation of multiple deletions as observed in other mitochondrial disorders. This mitochondrial dysfunction is likely to cause the phenotypes observed in these patients. The most prominent and consistent clinical signs were severe gut dysmotility and neurological abnormalities, including leukoencephalopathy, epilepsy, migraine, stroke-like episodes, and neurogenic bladder. A decrease in the number of myenteric neurons, and increased fibrosis and elastin levels were the most prominent changes in the gut. Cytochrome c oxidase (COX) deficient fibres in skeletal muscle were also observed. Disruption of lig3 in zebrafish reproduced the brain alterations and impaired gut transit in vivo. In conclusion, we identified variants in the LIG3 gene that result in a mitochondrial disease characterized by predominant gut dysmotility, encephalopathy, and neuromuscular abnormalities.


Subject(s)
DNA Ligase ATP/genetics , Gastrointestinal Diseases/genetics , Gastrointestinal Motility/genetics , Mitochondrial Encephalomyopathies/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Animals , Female , Gastrointestinal Diseases/pathology , Humans , Male , Mitochondrial Encephalomyopathies/pathology , Mutation , Pedigree , Zebrafish
14.
Front Endocrinol (Lausanne) ; 12: 560055, 2021.
Article in English | MEDLINE | ID: mdl-33833735

ABSTRACT

Serotonin (5-HT) is pivotal in the complex regulation of gut motility and consequent digestion of nutrients via multiple receptors. We investigated the serotonergic system in an agastric fish species, the ballan wrasse (Labrus bergylta) as it represents a unique model for intestinal function. Here we present evidence of the presence of enterochromaffin cells (EC cells) in the gut of ballan wrasse comprising transcriptomic data on EC markers like adra2a, trpa1, adgrg4, lmxa1, spack1, serpina10, as well as the localization of 5-HT and mRNA of the rate limiting enzyme; tryptophan hydroxylase (tph1) in the gut epithelium. Second, we examined the effects of dietary marine lipids on the enteric serotonergic system in this stomach-less teleost by administrating a hydrolyzed lipid bolus in ex vivo guts in an organ bath system. Modulation of the mRNA expression from the tryptophan hydroxylase tph1 (EC cells isoform), tph2 (neural isoform), and other genes involved in the serotonergic machinery were tracked. Our results showed no evidence to confirm that the dietary lipid meal did boost the production of 5-HT within the EC cells as mRNA tph1 was weakly regulated postprandially. However, dietary lipid seemed to upregulate the post-prandial expression of tph2 found in the serotonergic neurons. 5-HT in the intestinal tissue increased 3 hours after "exposure" of lipids, as was observed in the mRNA expression of tph2. This suggest that serotonergic neurons and not EC cells are responsible for the substantial increment of 5-HT after a lipid-reach "meal" in ballan wrasse. Cells expressing tph1 were identified in the gut epithelium, characteristic for EC cells. However, Tph1 positive cells were also present in the lamina propria. Characterization of these cells together with their implications in the serotonergic system will contribute to broad the scarce knowledge of the serotonergic system across teleosts.


Subject(s)
Dietary Fats/pharmacology , Intestines/drug effects , Perciformes , Serotonin/metabolism , Animals , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/genetics , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Gene Expression Regulation/drug effects , In Vitro Techniques , Intestines/metabolism , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Perciformes/genetics , Perciformes/metabolism , Serotonin/pharmacology
15.
Mol Med Rep ; 23(5)2021 05.
Article in English | MEDLINE | ID: mdl-33760185

ABSTRACT

Intestinal inflammation frequently occurs alongside dysmotility, which is characterized by altered myosin light chain phosphorylation levels. Curcumin, an active component from the ginger family, is reported to confer anti­inflammatory effects. However, the effects of curcumin on both diarrhea and constipation associated inflammation remains to be elucidated. The present study was designed to investigate the effects of curcumin on diarrhea and constipation and to determine the related mechanisms. Sprague­Dawley rats were used to establish diarrhea and constipation models via intracolonic acetic acid (4%) instillation or cold water gavage for 2 weeks, respectively. Blood samples were collected to measure the serum levels of the cytokines TNF­α and IL­1ß using ELISA kits. Western blotting was performed to measure NF­κB, RhoA, Rho­related kinase 2, phosphorylated MLC20, phosphorylated myosin phosphorylated target subunit 1, 130k Da­MLC kinase (MLCK), c­kit tyrosine kinase protein expression, and reverse transcription­quantitative PCR was conducted to measure MLCK expression levels. The results indicated that curcumin reversed the elevations in the pro­inflammatory cytokines IL­1ß and TNF­α by inhibiting the NF­κB pathway in rats with diarrhea and constipation. The results also indicated that myosin light chain (MLC) phosphorylation in intestinal smooth muscle was positively and negatively associated with the motility of inflammation­related diarrhea and constipation in rats, respectively. Curcumin significantly reversed the increased MLC phosphorylation in the jejunum of the rats with diarrhea, significantly enhanced the reductions in inflammatory mediators, including TNF­α and IL­1ß, of rats with constipation and significantly ameliorated the related hyper­motility and hypo­motility in rats with both diarrhea and constipation. In conclusion, the potential roles of the MLC kinase, c­kit tyrosine and Rho A/Rho­associated kinase 2 pathways, which are involved in curcumin­induced amelioration of inflammation­related diarrhea and constipation, were explored in the present study. Results from the present study suggested that curcumin has potential therapeutic value for treating intestinal inflammation and inflammation­related motility disorders.


Subject(s)
Curcumin/pharmacology , Gastrointestinal Motility/drug effects , Inflammation/drug therapy , Interleukin-1beta/genetics , Animals , Gastrointestinal Motility/genetics , Humans , Inflammation/genetics , Inflammation/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Myosin-Light-Chain Kinase/genetics , NF-kappa B/genetics , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin B/genetics , Tumor Necrosis Factor-alpha/genetics , rhoA GTP-Binding Protein/genetics
16.
J Clin Invest ; 131(6)2021 03 15.
Article in English | MEDLINE | ID: mdl-33497358

ABSTRACT

Hirschsprung disease (HSCR) is the most frequent developmental anomaly of the enteric nervous system, with an incidence of 1 in 5000 live births. Chronic intestinal pseudo-obstruction (CIPO) is less frequent and classified as neurogenic or myogenic. Isolated HSCR has an oligogenic inheritance with RET as the major disease-causing gene, while CIPO is genetically heterogeneous, caused by mutations in smooth muscle-specific genes. Here, we describe a series of patients with developmental disorders including gastrointestinal dysmotility, and investigate the underlying molecular bases. Trio-exome sequencing led to the identification of biallelic variants in ERBB3 and ERBB2 in 8 individuals variably associating HSCR, CIPO, peripheral neuropathy, and arthrogryposis. Thorough gut histology revealed aganglionosis, hypoganglionosis, and intestinal smooth muscle abnormalities. The cell type-specific ErbB3 and ErbB2 function was further analyzed in mouse single-cell RNA sequencing data and in a conditional ErbB3-deficient mouse model, revealing a primary role for ERBB3 in enteric progenitors. The consequences of the identified variants were evaluated using quantitative real-time PCR (RT-qPCR) on patient-derived fibroblasts or immunoblot assays on Neuro-2a cells overexpressing WT or mutant proteins, revealing either decreased expression or altered phosphorylation of the mutant receptors. Our results demonstrate that dysregulation of ERBB3 or ERBB2 leads to a broad spectrum of developmental anomalies, including intestinal dysmotility.


Subject(s)
Developmental Disabilities/genetics , Intestinal Pseudo-Obstruction/genetics , Mutation , Neuregulin-1/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics , Adolescent , Animals , Child, Preschool , Developmental Disabilities/pathology , Disease Models, Animal , Female , Gastrointestinal Motility/genetics , Hirschsprung Disease/genetics , Hirschsprung Disease/pathology , Humans , Infant, Newborn , Intestinal Pseudo-Obstruction/pathology , Male , Mice , Models, Molecular , Pedigree , Phenotype , Pregnancy , Receptor, ErbB-2/chemistry , Receptor, ErbB-3/chemistry , Receptor, ErbB-3/deficiency
17.
Cell Mol Gastroenterol Hepatol ; 11(2): 623-637, 2021.
Article in English | MEDLINE | ID: mdl-32992050

ABSTRACT

BACKGROUND & AIMS: YAP (Yap1) and TAZ (Wwtr1) are transcriptional co-activators and downstream effectors of the Hippo pathway, which play crucial roles in organ size control and cancer pathogenesis. Genetic deletion of YAP/TAZ has shown their critical importance for embryonic development of the heart, vasculature, and gastrointestinal mesenchyme. The aim of this study was to determine the functional role of YAP/TAZ in adult smooth muscle cells in vivo. METHODS: Because YAP and TAZ are mutually redundant, we used YAP/TAZ double-floxed mice crossed with mice that express tamoxifen-inducible CreERT2 recombinase driven by the smooth muscle-specific myosin heavy chain promoter. RESULTS: Double-knockout of YAP/TAZ in adult smooth muscle causes lethality within 2 weeks, mainly owing to colonic pseudo-obstruction, characterized by severe distension and fecal impaction. RNA sequencing in colon and urinary bladder showed that smooth muscle markers and muscarinic receptors were down-regulated in the YAP/TAZ knockout. The same transcripts also correlated with YAP/TAZ in the human colon. Myograph experiments showed reduced contractility to depolarization by potassium chloride and a nearly abolished muscarinic contraction and spontaneous activity in colon rings of YAP/TAZ knockout. CONCLUSIONS: YAP and TAZ in smooth muscle are guardians of colonic contractility and control expression of contractile proteins and muscarinic receptors. The knockout model has features of human chronic intestinal pseudo-obstruction and may be useful for studying this disease.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Colon/physiopathology , Colonic Pseudo-Obstruction/genetics , Muscle, Smooth/physiopathology , YAP-Signaling Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Colonic Pseudo-Obstruction/physiopathology , Disease Models, Animal , Female , Gastrointestinal Motility/genetics , Humans , Male , Mice , Mice, Knockout , Muscle Contraction/genetics , YAP-Signaling Proteins/metabolism
18.
Biochem Genet ; 59(1): 335-345, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33136283

ABSTRACT

Uncontrolled diabetes mellitus may affect any part of the gastrointestinal tract (GIT) and impact negatively the quality of life. Angiotensin-converting enzyme (ACE) gene polymorphism can have direct effect on circulating level of ACE which further modifies the degradation of substance P and thus may influence the gut motility. Hence, it could be hypothesised that ACE gene polymorphism would influence the gut motility. An observational analytical study was conducted at PGIMER, Chandigarh. 300 Type2 diabetes mellitus (T2DM) and 200 age and sex matched healthy individuals were enrolled. After taking written consent, 5 ml blood sample was collected for measurement of substance P by ELISA method and for ACE gene polymorphism (insertion[I]/deletion[D]) by polymerase chain reaction. Orocecal transit time (OCTT) was measured using non-invasive lactulose breath test. Out of 300 diabetic patients, 32.7%, 44% and 23.3% belonged to II, ID and DD genotypes, respectively. The frequency of D allele (OR = 1.39) and DD genotype (OR = 2.17) was significantly higher in patients than in controls and was associated with increased risk. Moreover, more number of diabetes patients with constipation (90%) belonged to DD genotype and their OCTT was significantly delayed (166.7 ± 7.3 min) as compared to ID (143.5 ± 4.2 min) or II (121.8 ± 4.9 min) genotype. From this study, it could be concluded that ACE gene polymorphism could be an important contributing factor to influence the gut motility and thus giving rise to the GI symptoms for T2DM patients.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Gastrointestinal Motility/genetics , Peptidyl-Dipeptidase A/genetics , Polymorphism, Genetic , Substance P/metabolism , Alleles , Angiotensins/genetics , Female , Gene Frequency , Genotype , Humans , Male , Middle Aged , Polymerase Chain Reaction
19.
Int J Mol Sci ; 21(24)2020 Dec 12.
Article in English | MEDLINE | ID: mdl-33322729

ABSTRACT

(1) Background: We characterized a novel animal model with obesity-induced constipation because constipation is rarely known in genetically engineered mice (GEM); (2) Methods: The changes in the constipation parameters and mechanisms were analyzed in CRISPR-Cas9-mediated leptin (Lep) knockout (KO) mice from eight to 24 weeks; (3) Results: Significant constipation phenotypes were observed in the Lep KO mice since 16 weeks old. These mice showed a significant decrease in the gastrointestinal motility, mucosal layer thickness and ability for mucin secretion as well as the abnormal ultrastructure of Lieberkühn crypts in the transverse colon. The density or function of the enteric neurons, intestinal Cajal cells (ICC), smooth muscle cells, and the concentration of gastrointestinal (GI) hormones for the GI motility were remarkably changed in Lep KO mice. The downstream signaling pathway of muscarinic acetylcholine receptors (mAChRs) were activated in Lep KO mice, while the expression of adipogenesis-regulating genes were alternatively reduced in the transverse colon of the same mice; (4) Conclusions: These results provide the first strong evidence that Lep KO mice can represent constipation successfully through dysregulation of the GI motility mediated by myenteric neurons, ICC, and smooth muscle cells in the transverse colon during an abnormal function of the lipid metabolism.


Subject(s)
Colon/metabolism , Constipation/metabolism , Gastrointestinal Motility , Leptin/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Muscarinic/metabolism , Adipogenesis/genetics , Animals , Aquaporin 3/metabolism , Aquaporins/metabolism , CRISPR-Cas Systems , Colon/cytology , Colon/pathology , Colon/ultrastructure , Constipation/complications , Constipation/genetics , Constipation/pathology , Disease Models, Animal , Female , Gastrointestinal Hormones/metabolism , Gastrointestinal Motility/genetics , Gastrointestinal Motility/physiology , Interstitial Cells of Cajal/metabolism , Leptin/genetics , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mucins/metabolism , Neurons/metabolism , Obesity/complications , Obesity/genetics , Signal Transduction/genetics
20.
Sci Rep ; 10(1): 11521, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32661417

ABSTRACT

Intestinal ischemia/reperfusion (I/R) injury has severe consequences on myenteric neurons, which can be irreversibly compromised resulting in slowing of transit and hindered food digestion. Myenteric neurons synthesize hyaluronan (HA) to form a well-structured perineuronal net, which undergoes derangement when myenteric ganglia homeostasis is perturbed, i.e. during inflammation. In this study we evaluated HA involvement in rat small intestine myenteric plexus after in vivo I/R injury induced by clamping a branch of the superior mesenteric artery for 60 min, followed by 24 h of reperfusion. In some experiments, 4-methylumbelliferone (4-MU, 25 mg/kg), a HA synthesis inhibitor, was intraperitoneally administered to normal (CTR), sham-operated (SH) and I/R animals for 24 h. In longitudinal muscle myenteric plexus (LMMP) whole-mount preparations, HA binding protein staining as well as HA levels were significantly higher in the I/R group, and were reduced after 4-MU treatment. HA synthase 1 and 2 (HAS1 and HAS2) labelled myenteric neurons and mRNA levels in LMMPs increased in the I/R group with respect to CTR, and were reduced by 4-MU. The efficiency of the gastrointestinal transit was significantly reduced in I/R and 4-MU-treated I/R groups with respect to CTR and SH groups. In the 4-MU-treated I/R group gastric emptying was reduced with respect to the CTR, SH and I/R groups. Carbachol (CCh) and electrical field (EFS, 0.1-40 Hz) stimulated contractions and EFS-induced (10 Hz) NANC relaxations were reduced in the I/R group with respect to both CTR and SH groups. After I/R, 4-MU treatment increased EFS contractions towards control values, but did not affect CCh-induced contractions. NANC on-relaxations after I/R were not influenced by 4-MU treatment. Main alterations in the neurochemical coding of both excitatory (tachykinergic) and inhibitory pathways (iNOS, VIPergic) were also observed after I/R, and were influenced by 4-MU administration. Overall, our data suggest that, after an intestinal I/R damage, changes of HA homeostasis in specific myenteric neuron populations may influence the efficiency of the gastrointestinal transit. We cannot exclude that modulation of HA synthesis in these conditions may ameliorate derangement of the enteric motor function preventing, at least in part, the development of dysmotility.


Subject(s)
Gastrointestinal Transit/physiology , Hyaluronic Acid/metabolism , Intestine, Small/metabolism , Reperfusion Injury/metabolism , Animals , Disease Models, Animal , Ganglia/metabolism , Gastrointestinal Motility/genetics , Gastrointestinal Motility/physiology , Gastrointestinal Transit/genetics , Humans , Hyaluronan Synthases/genetics , Ileum/metabolism , Ileum/physiology , Intestine, Small/pathology , Myenteric Plexus/metabolism , Nervous System Physiological Phenomena , Neurons/metabolism , Neurons/pathology , Rats , Reperfusion Injury/genetics , Reperfusion Injury/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...