Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Am J Physiol Gastrointest Liver Physiol ; 322(6): G583-G597, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35319286

ABSTRACT

Intestinal ganglionic cells in the adult enteric nervous system (ENS) are continually exposed to stimuli from the surrounding microenvironment and need at times to respond to disturbed homeostasis following acute intestinal injury. The kinase DCLK1 and intestinal Dclk1-positive cells have been reported to contribute to intestinal regeneration. Although Dclk1-positive cells are present in adult enteric ganglia, their cellular identity and response to acute injury have not been investigated in detail. Here, we reveal the presence of distinct Dclk1-tdTom+/CD49b+ glial-like and Dclk1-tdTom+/CD49b- neuronal cell types in adult myenteric ganglia. These ganglionic cells demonstrate distinct patterns of tracing over time yet show a similar expansion in response to elevated serotonergic signaling. Interestingly, Dclk1-tdTom+ glial-like and neuronal cell types appear resistant to acute irradiation injury-mediated cell death. Moreover, Dclk1-tdTom+/CD49b+ glial-like cells show prominent changes in gene expression profiles induced by injury, in contrast to Dclk1-tdTom+/CD49b- neuronal cell types. Finally, subsets of Dclk1-tdTom+/CD49b+ glial-like cells demonstrate prominent overlap with Nestin and p75NTR and strong responses to elevated serotonergic signaling or acute injury. These findings, together with their role in early development and their neural crest-like gene expression signature, suggest the presence of reserve progenitor cells in the adult Dclk1 glial cell lineage.NEW & NOTEWORTHY The kinase DCLK1 identifies glial-like and neuronal cell types in adult murine enteric ganglia, which resist acute injury-mediated cell death yet differ in their cellular response to injury. Interestingly, Dclk1-labeled glial-like cells show prominent transcriptional changes in response to injury and harbor features reminiscent of previously described enteric neural precursor cells. Our data thus add to recently emerging evidence of reserve cellular plasticity in the adult enteric nervous system.


Subject(s)
Enteric Nervous System , Neural Stem Cells , Animals , Enteric Nervous System/physiology , Integrin alpha2/metabolism , Mice , Mice, Transgenic , Neuroglia/metabolism , Neurons/metabolism
2.
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
3.
Liver Int ; 41(9): 2068-2075, 2021 09.
Article in English | MEDLINE | ID: mdl-33826804

ABSTRACT

BACKGROUND AND AIMS: Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) associated acute liver injury (ALI) has been linked to poor outcomes in adults. Here we compare characteristics in children with elevated ALT (E-ALT) in two distinct manifestations of the infection, multisystem inflammatory syndrome-children (MIS-C) and coronavirus disease 2019 (COVID-19). METHODS: This is a retrospective study of patients ≤21 years of age with positive for SARS-CoV-2 PCR. E-ALT was defined as alanine aminotransferase (ALT) > 40 U/L. Bivariate analysis and multivariable logistic regression were obtained to describe differences in children with and without E-ALT in COVID-19 and MIS-C. RESULTS: E-ALT was detected in 36% of the 291 patients; 31% with COVID-19, and 51% with MIS-C. E-ALT in COVID-19 was associated with obesity (P < .001), immunocompromised status (P = .04), and chronic liver disease (P = .01). In the regression models, E-ALT in COVID-19 was associated with higher c-reactive protein (OR 1.08, P = .01) after adjusting for common independent predictors. Children with E-ALT and MIS-C were more often boys (P = .001), Hispanic (P = .04), or Black (P < .001). In MIS-C, male gender (OR 5.3, P = .02) and Black race (OR 4.4, P = .04) were associated with increased odds of E-ALT. Children with E-ALT in both cohorts had significantly higher multiorgan dysfunction, longer hospitalization, and ICU stay. Children with MIS-C had 2.3-fold increased risk of E-ALT compared to COVID-19. No association was found between E-ALT and mortality. CONCLUSION: E-ALT with SARS-CoV-2 presents as elevated transaminases without hepatic synthetic dysfunction. Patients with either manifestation of SARS-CoV-2 infection and E-ALT experienced more severe disease.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Liver , Male , Phenotype , Retrospective Studies , Systemic Inflammatory Response Syndrome
5.
Gastroenterology ; 157(2): 507-521.e4, 2019 08.
Article in English | MEDLINE | ID: mdl-31071306

ABSTRACT

BACKGROUND & AIMS: Mood disorders and constipation are often comorbid, yet their shared etiologies have rarely been explored. The neurotransmitter serotonin (5-HT) regulates central nervous system and enteric nervous system (ENS) development and long-term functions, including gastrointestinal (GI) motility and mood. Therefore, defects in neuron production of 5-HT might result in brain and intestinal dysfunction. Tryptophan hydroxylase 2 (TPH2) is the rate-limiting enzyme in 5-HT biosynthesis. A variant of TPH2 that encodes the R441H substitution (TPH2-R441H) was identified in individuals with severe depression. We studied mice with an analogous mutation (TPH2-R439H), which results in a 60%-80% decrease in levels of 5-HT in the central nervous system and behaviors associated with depression in humans. Feeding chow that contains 5-HTP slow release (5-HTP SR) to TPH2-R439H mice restores levels of 5-HT in the central nervous system and reduces depressive-like behaviors. METHODS: We compared the effects of feeding chow, with or without 5-HTP SR, to mice with the TPH2-R439H mutation and without this mutation (control mice). Myenteric and submucosal plexuses were isolated from all 4 groups of mice, and immunocytochemistry was used to quantify total enteric neurons, serotonergic neurons, and 5-HT-dependent subsets of neurons. We performed calcium imaging experiments to evaluate responses of enteric neurons to tryptamine-evoked release of endogenous 5-HT. In live mice, we measured total GI transit, gastric emptying, small intestinal transit, and propulsive colorectal motility. To measure colonic migrating motor complexes (CMMCs), we isolated colons and constructed spatiotemporal maps along the proximodistal length to quantify the frequency, velocity, and length of CMMCs. We measured villus height, crypt perimeter, and relative densities of enterochromaffin and enteroendocrine cells in small intestinal tissue. RESULTS: Levels of 5-HT were significantly lower in enteric neurons from TPH2-R439H mice than from control mice. TPH2-R439H mice had abnormalities in ENS development and ENS-mediated GI functions, including reduced motility and intestinal epithelial growth. Total GI transit and propulsive colorectal motility were slower in TPH2-R439H mice than controls, and CMMCs were slower and less frequent. Villus height and crypt perimeter were significantly decreased in colon tissues from TPH2-R439H mice compared with controls. Administration of 5-HTP SR to adult TPH2-R439H mice restored 5-HT to enteric neurons and reversed these abnormalities. Adult TPH2-R439H mice given oral 5-HTP SR had normalized numbers of enteric neurons, total GI transit, and colonic motility. Intestinal tissue from these mice had normal measures of CMMCs and enteric epithelial growth CONCLUSIONS: In studies of TPH2-R439H mice, we found evidence for reduced release of 5-HT from enteric neurons that results in defects in ENS development and GI motility. Our findings indicate that neuron production of 5-HT links constipation with mood dysfunction. Administration of 5-HTP SR to mice restored 5-HT to the ENS and normalized GI motility and growth of the enteric epithelium. 5-HTP SR might be used to treat patients with intestinal dysfunction associated with low levels of 5-HT.


Subject(s)
5-Hydroxytryptophan/administration & dosage , Constipation/drug therapy , Depression/drug therapy , Gastrointestinal Tract/physiopathology , Serotonin/metabolism , Animals , Constipation/etiology , Constipation/physiopathology , Delayed-Action Preparations/administration & dosage , Depression/complications , Depression/genetics , Depression/physiopathology , Disease Models, Animal , Enteric Nervous System/drug effects , Enteric Nervous System/physiopathology , Female , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/innervation , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neurons/drug effects , Neurons/metabolism , Treatment Outcome , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
6.
Dig Dis Sci ; 65(3): 818-828, 2020 03.
Article in English | MEDLINE | ID: mdl-32056091

ABSTRACT

While there are numerous medical comorbidities associated with ASD, gastrointestinal (GI) issues have a significant impact on quality of life for these individuals. Recent findings continue to support the relationship between the gut microbiome and both GI symptoms and behavior, but the heterogeneity within the autism spectrum requires in-depth clinical characterization of these clinical cohorts. Large, diverse, well-controlled studies in this area of research are still needed. Although there is still much to discover about the brain-gut-microbiome axis in ASD, microbially mediated therapies, specifically probiotics and fecal microbiota transplantation have shown promise in the treatment of GI symptoms in ASD, with potential benefit to the core behavioral symptoms of ASD as well. Future research and clinical trials must increasingly consider complex phenotypes in ASD in stratification of large datasets as well as in design of inclusion criteria for individual therapeutic interventions.


Subject(s)
Autism Spectrum Disorder/microbiology , Brain/microbiology , Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Animals , Autism Spectrum Disorder/psychology , Autism Spectrum Disorder/therapy , Brain/physiology , Fecal Microbiota Transplantation/trends , Gastrointestinal Diseases/psychology , Gastrointestinal Diseases/therapy , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/physiology , Humans , Probiotics/administration & dosage
7.
Pediatr Dermatol ; 37(4): 695-697, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32342551

ABSTRACT

We report a case of a newly recognized primary immunodeficiency due to biallelic mutations in CARMIL2 manifesting as an actinic prurigo-like photodermatitis, allergic diathesis and recurrent infections in a child. We present this case to highlight a rare phenotype seen in this T-cell immunodeficiency and provide an overview of other dermatologic manifestations among published reports of this condition.


Subject(s)
Immunologic Deficiency Syndromes , Photosensitivity Disorders , Prurigo , Skin Diseases, Genetic , Child , Humans , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Photosensitivity Disorders/diagnosis , Photosensitivity Disorders/genetics
10.
Am J Physiol Gastrointest Liver Physiol ; 307(8): G848-62, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25147234

ABSTRACT

Enteric neurons express oxytocin (OT); moreover, enteric neurons and enterocytes express developmentally regulated OT receptors (OTRs). Although OT (with secretin) opposes intestinal inflammation, physiological roles played by enteric OT/OTR signaling have not previously been determined. We tested hypotheses that OT/OTR signaling contributes to enteric nervous system (ENS)-related gastrointestinal (GI) physiology. GI functions and OT effects were compared in OTR-knockout (OTRKO) and wild-type (WT) mice. Stool mass and water content were greater in OTRKO mice than in WT. GI transit time in OTRKO animals was faster than in WT; OT inhibited in vitro generation of ENS-dependent colonic migrating motor complexes in WT but not in OTRKO mice. Myenteric neurons were hyperplastic in OTRKO animals, and mucosal exposure to cholera toxin (CTX) in vitro activated Fos in more myenteric neurons in OTRKO than WT than in WT mice; OT inhibited the CTX response in WT but not in OTRKO mice. Villi and crypts were shorter in OTRKO than in WT mice, and transit-amplifying cell proliferation in OTRKO crypts was deficient. Macromolecular intestinal permeability in OTRKO was greater than WT mice, and experimental colitis was more severe in OTRKO mice; moreover, OT protected WT animals from colitis. Observations suggest that OT/OTR signaling acts as a brake on intestinal motility, decreases mucosal activation of enteric neurons, and promotes enteric neuronal development and/or survival. It also regulates proliferation of crypt cells and mucosal permeability; moreover OT/OTR signaling is protective against inflammation. Oxytocinergic signaling thus appears to play an important role in multiple GI functions that are subject to neuronal regulation.


Subject(s)
Gastrointestinal Motility , Intestinal Absorption , Intestinal Mucosa/metabolism , Oxytocin/metabolism , Receptors, Oxytocin/metabolism , Animals , Cell Proliferation , Gene Deletion , Inflammation/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myenteric Plexus/cytology , Myenteric Plexus/physiology , Neurons/metabolism , Neurons/physiology , Receptors, Oxytocin/genetics , Signal Transduction
11.
Gastroenterology ; 143(2): 408-17.e2, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22609381

ABSTRACT

BACKGROUND & AIMS: The enteric abundance of serotonin (5-HT), its ability to promote proliferation of neural precursors, and reports that 5-HT antagonists affect crypt epithelial proliferation led us to investigate whether 5-HT affects growth and maintenance of the intestinal mucosa in mice. METHODS: cMice that lack the serotonin re-uptake transporter (SERTKO mice) and wild-type mice were given injections of selective serotonin re-uptake inhibitors (gain-of-function models). We also analyzed mice that lack tryptophan hydroxylase-1 (TPH1KO mice, which lack mucosal but not neuronal 5-HT) and mice deficient in tryptophan hydroxylase-2 (TPH2KO mice, which lack neuronal but not mucosal 5-HT) (loss-of-function models). Wild-type and SERTKO mice were given ketanserin (an antagonist of the 5-HT receptor, 5-HT(2A)) or scopolamine (an antagonist of the muscarinic receptor). 5-HT(2A) receptors and choline acetyltransferase were localized by immunocytochemical analysis. RESULTS: Growth of the mucosa and proliferation of mucosal cells were significantly greater in SERTKO mice and in mice given selective serotonin re-uptake inhibitors than in wild-type mice, but were diminished in TPH2KO (but not in TPH1KO) mice. Ketanserin and scopolamine each prevented the ability of SERT knockout or inhibition to increase mucosal growth and proliferation. Cholinergic submucosal neurons reacted with antibodies against 5-HT(2A). CONCLUSIONS: 5-HT promotes growth and turnover of the intestinal mucosal epithelium. Surprisingly, these processes appear to be mediated by neuronal, rather than mucosal, 5-HT. The 5-HT(2A) receptor activates cholinergic neurons, which provide a muscarinic innervation to epithelial effectors.


Subject(s)
Cholinergic Neurons/metabolism , Enterochromaffin Cells/metabolism , Intestinal Mucosa/growth & development , Receptor, Serotonin, 5-HT2A/metabolism , Serotonergic Neurons/metabolism , Serotonin/physiology , Animals , Choline O-Acetyltransferase/metabolism , Intestinal Mucosa/metabolism , Ketanserin/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscarinic Antagonists/administration & dosage , Scopolamine/administration & dosage , Serotonin/metabolism , Serotonin Antagonists/administration & dosage , Serotonin Plasma Membrane Transport Proteins/deficiency , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/metabolism , Tryptophan Hydroxylase/deficiency
12.
Gastroenterology ; 141(2): 576-87, 587.e1-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21669203

ABSTRACT

BACKGROUND & AIMS: Hand2 is a basic helix-loop-helix transcription factor required for terminal differentiation of enteric neurons. We studied Hand2 haploinsufficient mice, to determine whether reduced expression of Hand2 allows sufficient enteric neurogenesis for survival, but not for development of a normal enteric nervous system (ENS). METHODS: Enteric transcripts that encode Hand2 and the neuron-specific embryonic lethal abnormal vision proteins HuB, HuC, and HuD were quantified. Immunocytochemistry was used to identify and quantify neurons. Apoptosis was analyzed with the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling procedure. Intracellular microelectrodes were used to record inhibitory junction potentials. Gastrointestinal transit and colonic motility were measured in vivo. RESULTS: Levels of of enteric Hand2 transcripts were associated with genotypes of mice, in the following order: Hand2(+/+) > Hand2(LoxP/+) > Hand2(+/-) > Hand2(LoxP/-). Parallel reductions were found in expression of HuD and in regional and phenotypic manners. Numbers of neurons, numbers of neuronal nitric oxide synthase(+) and calretinin(+), but not substance P(+) or vasoactive intestinal peptide(+) neurons, decreased. No effects were observed in stomach or cecum. Apoptosis was not detected, consistent with the concept that Hand2 inhibits neuronal differentiation, rather than regulates survival. The amplitude of inhibitory junction potentials in colonic circular muscle was similar in Hand2 wild-type and haploinsufficient mice, although in haploinsufficient mice, the purinergic component was reduced and a nitrergic component appeared. The abnormal ENS of haploinsufficient mice slowed gastrointestinal motility but protected mice against colitis. CONCLUSIONS: Reduced expression of factors required for development of the ENS can cause defects in the ENS that are subtle enough to escape detection yet cause significant abnormalities in bowel function.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Enteric Nervous System/cytology , Gastrointestinal Motility/physiology , Neurons/cytology , Neurons/metabolism , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , Calbindin 2 , Cell Count , Colitis/chemically induced , Colitis/prevention & control , Colon/innervation , Colon/metabolism , Colon/physiology , ELAV Proteins/metabolism , ELAV-Like Protein 2 , ELAV-Like Protein 3 , ELAV-Like Protein 4 , Enteric Nervous System/growth & development , Gastrointestinal Motility/genetics , Genotype , Intercellular Junctions/physiology , Mice , Muscle, Smooth/innervation , Muscle, Smooth/physiology , Neuroglia/cytology , Nitric Oxide Synthase/metabolism , S100 Calcium Binding Protein G/metabolism , Substance P/metabolism , Synaptic Transmission/physiology , Trinitrobenzenesulfonic Acid , Vasoactive Intestinal Peptide/metabolism
13.
Research (Wash D C) ; 2022: 9804014, 2022.
Article in English | MEDLINE | ID: mdl-35958108

ABSTRACT

Tremendous progress has been made in the past decade regarding our understanding of the gut microbiome's role in human health. Currently, however, a comprehensive and focused review marrying the two distinct fields of gut microbiome and material research is lacking. To bridge the gap, the current paper discusses critical aspects of the rapidly emerging research topic of "material engineering in the gut microbiome and human health." By engaging scientists with diverse backgrounds in biomaterials, gut-microbiome axis, neuroscience, synthetic biology, tissue engineering, and biosensing in a dialogue, our goal is to accelerate the development of research tools for gut microbiome research and the development of therapeutics that target the gut microbiome. For this purpose, state-of-the-art knowledge is presented here on biomaterial technologies that facilitate the study, analysis, and manipulation of the gut microbiome, including intestinal organoids, gut-on-chip models, hydrogels for spatial mapping of gut microbiome compositions, microbiome biosensors, and oral bacteria delivery systems. In addition, a discussion is provided regarding the microbiome-gut-brain axis and the critical roles that biomaterials can play to investigate and regulate the axis. Lastly, perspectives are provided regarding future directions on how to develop and use novel biomaterials in gut microbiome research, as well as essential regulatory rules in clinical translation. In this way, we hope to inspire research into future biomaterial technologies to advance gut microbiome research and gut microbiome-based theragnostics.

14.
J Neurosci ; 30(49): 16730-40, 2010 Dec 08.
Article in English | MEDLINE | ID: mdl-21148012

ABSTRACT

The norepinephrine transporter (NET), which is expressed on the plasma membranes of noradrenergic neurons, is important in terminating neurotransmission. The noradrenergic sympathetic neurons that innervate the bowel express NET, but they are extrinsic and their cell bodies are not components of the enteric nervous system (ENS). Subsets of neurons were nevertheless found in the murine ENS that express transcripts encoding NET, NET protein, and dopamine ß-hydroxylase; these neurons lack tyrosine hydroxylase (TH) and thus are not catecholaminergic. Enteric NET expression, moreover, preceded the ingrowth of sympathetic axons during development and did not disappear when the gut was extrinsically denervated. Transiently catecholaminergic (TC), neural crest-derived precursors of enteric neurons expressed NET at embryonic day 10 (E10) and NET expression in the fetal gut peaked coincidentally with early neurogenesis at E12. Serotonergic neurons, which are born early from TC progenitors, were found to express NET in the adult ENS, as did also other early-born neurons containing calretinin or neuronal nitric oxide synthase (nNOS) immunoreactivities. NET was not expressed in TH-immunoreactive dopaminergic neurons, which are born perinatally. Genetic deletion of NET almost eliminated tryptophan hydroxylase 2 expression and significantly reduced the numbers of total, 5-HT- and calretinin-immunoreactive enteric neurons, without affecting the immunoreactivities of nNOS or TH. These observations indicate that TC precursors of subsets of noncatecholaminergic enteric neurons express NET that persists in the successors of these cells despite their loss of TH. NET expression is essential for development and/or survival of some (5-HT- and calretinin-expressing), but not all (nNOS-expressing), of these neurons.


Subject(s)
Enteric Nervous System/cytology , Gene Expression Regulation, Developmental/physiology , Neurons/metabolism , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Norepinephrine/metabolism , Serotonin/metabolism , Animals , Animals, Newborn , Autonomic Denervation/methods , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Embryo, Mammalian , Enteric Nervous System/embryology , Enteric Nervous System/growth & development , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type I/metabolism , Norepinephrine Plasma Membrane Transport Proteins/deficiency , Norepinephrine Plasma Membrane Transport Proteins/genetics , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism , Tyrosine 3-Monooxygenase/metabolism
15.
Neurogastroenterol Motil ; 33(8): e14100, 2021 08.
Article in English | MEDLINE | ID: mdl-33655600

ABSTRACT

BACKGROUND: Intrinsic primary afferent neurons (IPANs) enable the gut to manifest reflexes in the absence of CNS input. PKG1α is selectively expressed in a subset of neurons in dorsal root ganglia (DRG) and has been linked to nociception and long-term hyperexcitability. METHODS: We used immunoblotting, immunocytochemistry, and in vitro assays of IPAN-dependent enteric functions to test hypotheses that subsets of primary neurons of the ENS and DRG share a reliance on PKG1α expression. KEY RESULTS: PKG1α immunoreactivity was demonstrated in immunoblots from isolated myenteric ganglia. PKG1α, but not PKG1ß, immunoreactivity, was coincident with that of neuronal markers (HuC/D; ß3-tubulin) in both enteric plexuses. PKG1α immunoreactivity also co-localized with the immunoreactivities of the IPAN markers, calbindin (100%; myenteric plexus) and cytoplasmic NeuN (98 ± 1% submucosal plexus). CGRP-immunoreactive DRG neurons, identified as visceral afferents by retrograde transport, were PKG1α-immunoreactive. We used intraluminal cholera toxin to determine whether PKG1α was necessary to enable stimulation of the mucosa to activate Fos in enteric neurons. Tetrodotoxin (1.0 µM), low Ca2+ /high Mg2+ media, and the PKG inhibitor, N46 (100 µM), all inhibited Fos activation in myenteric neurons. N46 also concentration dependently inhibited peristaltic reflexes in isolated preparations of distal colon (IC50  = 83.3 ± 1.3 µM). CONCLUSIONS & INFERENCES: These data suggest that PKG1α is present and functionally important in IPANs and visceral afferent nociceptive neurons.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Enteric Nervous System/metabolism , Neurons, Afferent/metabolism , Protein Isoforms/metabolism , Animals , Female , Gastrointestinal Motility/physiology , Guinea Pigs , Intestines/metabolism , Male , Myenteric Plexus/metabolism , Proto-Oncogene Proteins c-fos/metabolism
16.
Gut Microbes ; 13(1): 1866974, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33459114

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder characterized primarily by motor and non-motor gastrointestinal (GI) deficits. GI symptoms' including compromised intestinal barrier function often accompanies altered gut microbiota composition and motor deficits in PD. Therefore, in this study, we set to investigate the role of gut microbiota and epithelial barrier dysfunction on motor symptom generation using a rotenone-induced mouse model of PD. We found that while six weeks of 10 mg/kg of chronic rotenone administration by oral gavage resulted in loss of tyrosine hydroxylase (TH) neurons in both germ-free (GF) and conventionally raised (CR) mice, the decrease in motor strength and coordination was observed only in CR mice. Chronic rotenone treatment did not disrupt intestinal permeability in GF mice but resulted in a significant change in gut microbiota composition and an increase in intestinal permeability in CR mice. These results highlight the potential role of gut microbiota in regulating barrier dysfunction and motor deficits in PD.


Subject(s)
Gastrointestinal Diseases/pathology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , Parkinson Disease/pathology , Rotenone/toxicity , Tight Junctions/pathology , Animals , Brain-Gut Axis , Disease Models, Animal , Dysbiosis/microbiology , Dystonic Disorders/congenital , Dystonic Disorders/pathology , Female , Germ-Free Life/physiology , Male , Mice , Tight Junctions/drug effects , Tyrosine 3-Monooxygenase/metabolism
17.
J Autism Dev Disord ; 49(1): 349-362, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30350113

ABSTRACT

Gastrointestinal dysfunction in children with autism spectrum disorder (ASD) is common and associated with problem behaviors. This study describes the development of a brief, parent-report screen that relies minimally upon the child's ability to report or localize pain for identifying children with ASD at risk for one of three common gastrointestinal disorders (functional constipation, functional diarrhea, and gastroesophageal reflux disease). In a clinical sample of children with ASD, this 17-item screen identified children having one or more of these disorders with a sensitivity of 84%, specificity of 43%, and a positive predictive value of 67%. If found to be valid in an independent sample of children with ASD, the screen will be useful in both clinical practice and research.


Subject(s)
Autism Spectrum Disorder/epidemiology , Gastrointestinal Diseases/epidemiology , Health Surveys/methods , Child , Female , Health Surveys/standards , Humans , Male , Parents
SELECTION OF CITATIONS
SEARCH DETAIL