Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Neurogastroenterol Motil ; 33(9): e14149, 2021 09.
Article in English | MEDLINE | ID: mdl-33837991

ABSTRACT

BACKGROUND: Normal gastrointestinal motility depends on electrical slow-wave activity generated by interstitial cells of Cajal (ICC) in the tunica muscularis of the gastrointestinal tract. A requirement for HCO3- in extracellular solutions used to record slow waves indicates a role for HCO3- transport in ICC pacemaking. The Slc4a4 gene transcript encoding the electrogenic Na+ /HCO3- cotransporter, NBCe1, is enriched in mouse small intestinal myenteric region ICC (ICC-MY) that generate slow waves. This study aimed to determine how extracellular HCO3- concentrations affect electrical activity in mouse small intestine and to determine the contribution of NBCe1 activity to these effects. METHODS: Immunohistochemistry and sharp electrode electrical recordings were used. KEY RESULTS: The NBCe1 immunoreactivity was localized to ICC-MY of the tunica muscularis. In sharp electrode electrical recordings, removal of HCO3- from extracellular solutions caused significant, reversible, depolarization of the smooth muscle and a reduction in slow-wave amplitude and frequency. In 100 mM HCO3- , the muscle hyperpolarized and slow wave amplitude and frequency increased. The effects of replacing extracellular Na+ with Li+ , an ion that does not support NBCe1 activity, were similar to, but larger than, the effects of removing HCO3- . There were no additional changes to electrical activity when HCO3- was removed from Li+ containing solutions. The Na+ /HCO3- cotransport inhibitor, S-0859 (30µM) significantly reduced the effect of removing HCO3- on electrical activity. CONCLUSIONS & INFERENCES: These studies demonstrate a major role for Na+ /HCO3- cotransport by NBCe1 in electrical activity of mouse small intestine and indicated that regulation of intracellular acid:base homeostasis contributes to generation of normal pacemaker activity in the gastrointestinal tract.


Subject(s)
Bicarbonates/metabolism , Gastrointestinal Motility/physiology , Intestine, Small/physiology , Sodium-Bicarbonate Symporters/metabolism , Animals , Female , Ion Transport/physiology , Male , Mice , Mice, Inbred C57BL
2.
Am J Physiol Gastrointest Liver Physiol ; 320(1): G93-G107, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33112159

ABSTRACT

Interstitial cells of Cajal (ICCs) generate electrical slow waves, which are required for normal gastrointestinal motility. The mechanisms for generation of normal pacemaking are not fully understood. Normal gastrointestinal contractility- and electrical slow-wave activity depend on the presence of extracellular HCO3-. Previous transcriptional analysis identified enrichment of mRNA encoding the electrogenic Na+/HCO3- cotransporter (NBCe1) gene (Slc4a4) in pacemaker myenteric ICCs in mouse small intestine. We aimed to determine the distribution of NBCe1 protein in ICCs of the mouse gastrointestinal tract and to identify the transcripts of the Slc4a4 gene in mouse and human small intestinal tunica muscularis. We determined the distribution of NBCe1 immunoreactivity (NBCe1-IR) by immunofluorescent labeling in mouse and human tissues. In mice, NBCe1-IR was restricted to Kit-positive myenteric ICCs of the stomach and small intestine and submuscular ICCs of the large intestine, that is, the slow wave generating subset of ICCs. Other subtypes of ICCs were NBCe1-negative. Quantitative real-time PCR identified >500-fold enrichment of Slc4a4-207 and Slc4a4-208 transcripts ["IP3-receptor-binding protein released by IP3" (IRBIT)-regulated isoforms] in Kit-expressing cells isolated from KitcreERT2/+, Rpl22tm1.1Psam/Sj mice and from single GFP-positive ICCs from Kittm1Rosay mice. Human jejunal tunica muscularis ICCs were also NBCe1-positive, and SLC4A4-201 and SLC4A4-204 RNAs were >300-fold enriched relative to SLC4A4-202. In summary, NBCe1 protein expressed in ICCs with electrical pacemaker function is encoded by Slc4a4 gene transcripts that generate IRBIT-regulated isoforms of NBCe1. In conclusion, Na+/HCO3- cotransport through NBCe1 contributes to the generation of pacemaker activity in subsets of ICCs.NEW & NOTEWORTHY In this study, we show that the electrogenic Na+/HCO3- cotransporter, NBCe1/Slc4a4, is expressed in subtypes of interstitial cells of Cajal (ICCs) responsible for electrical slow wave generation throughout the mouse gastrointestinal tract and is absent in other types of ICCs. The transcripts of Slc4a4 expressed in mouse ICCs and human gastrointestinal smooth muscle are the regulated isoforms. This indicates a key role for HCO3- transport in generation of gastrointestinal motility patterns.


Subject(s)
Interstitial Cells of Cajal/metabolism , Sodium-Bicarbonate Symporters/metabolism , Sodium/metabolism , Symporters/metabolism , Adult , Aged , Animals , Humans , Intestine, Small/metabolism , Mice, Transgenic , Middle Aged , Muscle, Smooth/physiology , Oocytes/metabolism , RNA-Binding Proteins/metabolism , Ribosomal Proteins/metabolism
3.
Neurogastroenterol Motil ; 33(2): e13994, 2021 02.
Article in English | MEDLINE | ID: mdl-33000540

ABSTRACT

BACKGROUND: Gastrointestinal (GI) motility is a complex physiological process that is critical for normal GI function. Disruption of GI motility frequently occurs in GI diseases or as side effects of therapeutics. Whole gut transit measurements, like carmine red leading-edge transit, in mice form the cornerstone of in vivo preclinical GI motility studies. METHOD: We have developed an easily achievable, labor-saving method to measure whole gut transit time in mice. This approach uses inexpensive, commercially available materials to monitor pellet production over time via high definition cameras capturing time-lapse video for offline analysis. KEY RESULT: We describe the assembly of our automated gut transit setup and validate this approach by comparing the results with loperamide to delay transit and conventional transit measurements. We demonstrate that compared to the control group, the loperamide group had slowed transit, evidenced by a decrease in total pellet production and prolonged whole gut transit time. The control group had an extended transit time compared with the results reported in the literature. Whole gut transit rates accelerated to times comparable to the literature by disrupting cages every 10-15 min to imitate the conventional approach, suggesting that disruption affects the assay and supports the use of an automated approach. CONCLUSION & INFERENCES: A novel automated, inexpensive, and easily assembled whole gut transit setup is labor-saving and allows minimal disruption to animal behavior compared with the conventional approach.


Subject(s)
Gastrointestinal Transit , Time-Lapse Imaging/methods , Animals , Automation, Laboratory , Female , Male , Mice , Mice, Inbred C57BL , Time-Lapse Imaging/instrumentation
4.
Neurogastroenterol Motil ; 33(3): e13993, 2021 03.
Article in English | MEDLINE | ID: mdl-33020982

ABSTRACT

BACKGROUND AND AIM: Muscularis macrophages (MMs) not only mediate the innate immunity, but also functionally interact with cells important for gastrointestinal motility. The aim of this study was to determine the spatial relationship and types of contacts between the MMs and neighboring cells in the muscularis propria of human and mouse stomach, small intestine, and large intestine. METHODS: The distribution and morphology of MMs and their contacts with other cells were investigated by immunohistochemistry and transmission electron microscopy. KEY RESULTS: Immunohistochemistry showed variable shape and number of MMs according to their location in different portions of the muscle coat. By double labeling, a close association between MMs and neighboring cells, that is, neurons, smooth muscle cells, interstitial cells of Cajal (ICCs), telocytes (TCs)/PDGFRα-positive cells, was seen. Electron microscopy demonstrated that in the muscle layers of both animal species, MMs have similar ultrastructural features and have specialized cell-to-cell contacts with smooth muscle cells and TCs/PDGFRα-positive cells but not with ICCs and enteric neurons. CONCLUSION & INFERENCES: This study describes varying patterns of distribution of MMs between different regions of the gut, and reports the presence of distinct and extended cell-to-cell contacts between MMs and smooth muscle cells and between MMs and TCs/PDGFRα-positive cells. In contrast, MMs, although close to ICCs and nerve elements, did not make contact with them. These findings indicate specialized and variable roles for MMs in the modulation of gastrointestinal motility whose significance should be more closely investigated in normal and pathological conditions.


Subject(s)
Gastric Mucosa/cytology , Intercellular Junctions/ultrastructure , Intestinal Mucosa/cytology , Macrophages/cytology , Myocytes, Smooth Muscle/cytology , Telocytes/cytology , Animals , Cell Communication , Enteric Nervous System , Female , Gastric Mucosa/metabolism , Gastric Mucosa/ultrastructure , Humans , Interstitial Cells of Cajal/cytology , Interstitial Cells of Cajal/metabolism , Interstitial Cells of Cajal/ultrastructure , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Macrophages/metabolism , Macrophages/ultrastructure , Male , Mice , Microscopy, Electron, Transmission , Middle Aged , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/ultrastructure , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Telocytes/metabolism , Telocytes/ultrastructure
5.
Cell Mol Gastroenterol Hepatol ; 11(1): 117-145, 2021.
Article in English | MEDLINE | ID: mdl-32771388

ABSTRACT

BACKGROUND & AIMS: Gastric dysfunction in the elderly may cause reduced food intake, frailty, and increased mortality. The pacemaker and neuromodulator cells interstitial cells of Cajal (ICC) decline with age in humans, and their loss contributes to gastric dysfunction in progeric klotho mice hypomorphic for the anti-aging Klotho protein. The mechanisms of ICC depletion remain unclear. Klotho attenuates Wnt (wingless-type MMTV integration site) signaling. Here, we examined whether unopposed Wnt signaling could underlie aging-associated ICC loss by up-regulating transformation related protein TRP53 in ICC stem cells (ICC-SC). METHODS: Mice aged 1-107 weeks, klotho mice, APCΔ468 mice with overactive Wnt signaling, mouse ICC-SC, and human gastric smooth muscles were studied by RNA sequencing, reverse transcription-polymerase chain reaction, immunoblots, immunofluorescence, histochemistry, flow cytometry, and methyltetrazolium, ethynyl/bromodeoxyuridine incorporation, and ex-vivo gastric compliance assays. Cells were manipulated pharmacologically and by gene overexpression and RNA interference. RESULTS: The klotho and aged mice showed similar ICC loss and impaired gastric compliance. ICC-SC decline preceded ICC depletion. Canonical Wnt signaling and TRP53 increased in gastric muscles of klotho and aged mice and middle-aged humans. Overstimulated canonical Wnt signaling increased DNA damage response and TRP53 and reduced ICC-SC self-renewal and gastric ICC. TRP53 induction persistently inhibited G1/S and G2/M cell cycle phase transitions without activating apoptosis, autophagy, cellular quiescence, or canonical markers/mediators of senescence. G1/S block reflected increased cyclin-dependent kinase inhibitor 1B and reduced cyclin D1 from reduced extracellular signal-regulated kinase activity. CONCLUSIONS: Increased Wnt signaling causes age-related ICC loss by up-regulating TRP53, which induces persistent ICC-SC cell cycle arrest without up-regulating canonical senescence markers.


Subject(s)
Aging/physiology , Cellular Senescence/physiology , Interstitial Cells of Cajal/physiology , Stomach/physiology , Adenomatous Polyposis Coli Protein/genetics , Animals , Cell Cycle Checkpoints , Female , Humans , Klotho Proteins/genetics , Male , Mice , Mice, Transgenic , Middle Aged , Models, Animal , Stomach/cytology , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Wnt Signaling Pathway , Young Adult
6.
Neurogastroenterol Motil ; 32(12): e13989, 2020 12.
Article in English | MEDLINE | ID: mdl-32986284

ABSTRACT

BACKGROUND: The gut is the only organ system with intrinsic neural reflexes. Intrinsic primary afferent neurons (IPANs) of the enteric nervous system initiate intrinsic reflexes, form gut-brain connections, and undergo considerable neuroplasticity to cause digestive diseases. They remain inaccessible to study in mice in the absence of a selective marker. Advillin is used as a marker for primary afferent neurons in dorsal root ganglia. The aim of this study was to test the hypothesis that advillin is expressed in IPANs of the mouse jejunum. METHODS: Advillin expression was assessed with immunohistochemistry and using transgenic mice expressing an inducible Cre recombinase under the advillin promoter were used to drive tdTomato and the genetically encoded calcium indicator GCaMP5. These mice were used to characterize the morphology and physiology of advillin-expressing enteric neurons using confocal microscopy, calcium imaging, and whole-cell patch-clamp electrophysiology. KEY RESULTS: Advillin is expressed in about 25% of myenteric neurons of the mouse jejunum, and these neurons demonstrate the requisite properties of IPANs. Functionally, they demonstrate calcium responses following mechanical stimuli of the mucosa and during antidromic action potentials. They have Dogiel type II morphology with neural processes that mostly remain within the myenteric plexus, but also project to the mucosa and express NeuN and calcitonin gene-related peptide (CGRP), but not nNOS. CONCLUSIONS AND INFERENCES: Advillin marks jejunal IPANs providing accessibility to this important neuronal population to study and model digestive disease.


Subject(s)
Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Jejunum/cytology , Jejunum/metabolism , Microfilament Proteins/biosynthesis , Neurons, Afferent/metabolism , Animals , Calcium Signaling/physiology , Enteric Nervous System/chemistry , Jejunum/chemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/genetics , Neurons, Afferent/chemistry
7.
Gut ; 69(5): 868-876, 2020 05.
Article in English | MEDLINE | ID: mdl-31757880

ABSTRACT

OBJECTIVE: This study was designed to evaluate the roles of microRNAs (miRNAs) in slow transit constipation (STC). DESIGN: All human tissue samples were from the muscularis externa of the colon. Expression of 372 miRNAs was examined in a discovery cohort of four patients with STC versus three age/sex-matched controls by a quantitative PCR array. Upregulated miRNAs were examined by quantitative reverse transcription qPCR (RT-qPCR) in a validation cohort of seven patients with STC and age/sex-matched controls. The effect of a highly differentially expressed miRNA on a custom human smooth muscle cell line was examined in vitro by RT-qPCR, electrophysiology, traction force microscopy, and ex vivo by lentiviral transduction in rat muscularis externa organotypic cultures. RESULTS: The expression of 13 miRNAs was increased in STC samples. Of those miRNAs, four were predicted to target SCN5A, the gene that encodes the Na+ channel NaV1.5. The expression of SCN5A mRNA was decreased in STC samples. Let-7f significantly decreased Na+ current density in vitro in human smooth muscle cells. In rat muscularis externa organotypic cultures, overexpression of let-7f resulted in reduced frequency and amplitude of contraction. CONCLUSIONS: A small group of miRNAs is upregulated in STC, and many of these miRNAs target the SCN5A-encoded Na+ channel NaV1.5. Within this set, a novel NaV1.5 regulator, let-7f, resulted in decreased NaV1.5 expression, current density and reduced motility of GI smooth muscle. These results suggest NaV1.5 and miRNAs as novel diagnostic and potential therapeutic targets in STC.


Subject(s)
Constipation/physiopathology , Gene Expression Regulation , MicroRNAs/genetics , Microtubule-Associated Proteins/genetics , Muscle Contraction/genetics , Adult , Aged , Biopsy, Needle , Case-Control Studies , Colon/pathology , Female , Gastrointestinal Motility/genetics , Humans , Immunohistochemistry , Middle Aged , Muscle Contraction/physiology , Muscle, Smooth , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction/methods , Reference Values , Sampling Studies , Up-Regulation
8.
Am J Physiol Gastrointest Liver Physiol ; 317(5): G716-G726, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31482734

ABSTRACT

Macrophage-based immune dysregulation plays a critical role in development of delayed gastric emptying in diabetic mice. Loss of anti-inflammatory macrophages and increased expression of genes associated with pro-inflammatory macrophages has been reported in full-thickness gastric biopsies from gastroparesis patients. We aimed to determine broader protein expression (proteomics) and protein-based signaling pathways in gastric biopsies of diabetic (DG) and idiopathic gastroparesis (IG) patients. Additionally, we determined correlations between protein expressions, gastric emptying, and symptoms. Full-thickness gastric antrum biopsies were obtained from nine DG patients, seven IG patients, and five nondiabetic controls. Aptamer-based SomaLogic tissue scan that quantitatively identifies 1,305 human proteins was used. Protein fold changes were computed, and differential expressions were calculated using Limma. Ingenuity pathway analysis and correlations were carried out. Multiple-testing corrected P < 0.05 was considered statistically significant. Seventy-three proteins were differentially expressed in DG, 132 proteins were differentially expressed in IG, and 40 proteins were common to DG and IG. In both DG and IG, "Role of Macrophages, Fibroblasts and Endothelial Cells" was the most statistically significant altered pathway [DG false discovery rate (FDR) = 7.9 × 10-9; IG FDR = 6.3 × 10-12]. In DG, properdin expression correlated with GCSI bloating (r = -0.99, FDR = 0.02) and expressions of prostaglandin G/H synthase 2, protein kinase C-ζ type, and complement C2 correlated with 4 h gastric retention (r = -0.97, FDR = 0.03 for all). No correlations were found between proteins and symptoms or gastric emptying in IG. Protein expression changes suggest a central role of macrophage-driven immune dysregulation in gastroparesis, specifically, complement activation in diabetic gastroparesis.NEW & NOTEWORTHY This study uses SOMAscan, a novel proteomics assay for determination of altered proteins and associated molecular pathways in human gastroparesis. Seventy-three proteins were changed in diabetic gastroparesis, 132 in idiopathic gastroparesis compared with controls. Forty proteins were common in both. Macrophage-based immune dysregulation pathway was most significantly affected in both diabetic and idiopathic gastroparesis. Proteins involved in the complement and prostaglandin synthesis pathway were associated with symptoms and gastric emptying delay in diabetic gastroparesis.


Subject(s)
Diabetes Complications/genetics , Gastroparesis/genetics , Proteome/genetics , Adult , Aged , Complement C2/genetics , Complement C2/metabolism , Diabetes Complications/metabolism , Diabetes Complications/physiopathology , Endothelial Cells/metabolism , Female , Fibroblasts/metabolism , Gastric Emptying , Gastroparesis/etiology , Gastroparesis/metabolism , Gastroparesis/physiopathology , Humans , Macrophages/metabolism , Male , Middle Aged , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Proteome/metabolism
9.
Am J Physiol Gastrointest Liver Physiol ; 317(2): G141-G146, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31169993

ABSTRACT

Postsurgical gastric dysfunction is common, but the mechanisms are varied and poorly understood. The pylorus normally acts as an electrical barrier isolating gastric and intestinal slow waves. In this report, we present an aberrant electrical conduction pathway arising between the stomach and small intestine, following pyloric excision and surgical anastomosis, as a novel disease mechanism. A patient was referred with postsurgical gastroparesis following antrectomy, gastroduodenostomy, and vagotomy for peptic ulceration. Scintigraphy confirmed markedly abnormal 4-h gastric retention. Symptoms included nausea, vomiting, postprandial distress, and reflux. Intraoperative, high-resolution electrical mapping was performed across the anastomosis immediately before revision gastrectomy, and the resected anastomosis underwent immunohistochemistry for interstitial cells of Cajal. Mapping revealed continuous, stable abnormal retrograde slow-wave propagation through the anastomosis, with slow conduction occurring at the scar (4.0 ± 0.1 cycles/min; 2.5 ± 0.6 mm/s; 0.26 ± 0.15 mV). Stable abnormal retrograde propagation continued into the gastric corpus with tachygastria (3.9 ± 0.2 cycles/min; 1.6 ± 0.5 mm/s; 0.19 ± 0.12 mV). Histology confirmed ingrowth of atypical ICC through the scar, defining an aberrant pathway enabling transanastomotic electrical conduction. In conclusion, a "gastrointestinal aberrant pathway" is presented as a novel proposed cause of postsurgical gastric dysfunction. The importance of aberrant anastomotic conduction in acute and long-term surgical recovery warrants further investigation.NEW & NOTEWORTHY High-resolution gastric electrical mapping was performed during revisional surgery in a patient with severe gastric dysfunction following antrectomy and gastroduodenostomy. The results revealed continuous propagation of slow waves from the duodenum to the stomach, through the old anastomotic scar, and resulting in retrograde-propagating tachygastria. Histology showed atypical interstitial cells of Cajal growth through the anastomotic scar. Based on these results, we propose a "gastrointestinal aberrant pathway" as a mechanism for postsurgical gastric dysfunction.


Subject(s)
Cicatrix , Duodenum , Electric Conductivity , Gastrectomy/adverse effects , Gastric Stump , Gastroparesis , Interstitial Cells of Cajal/pathology , Postoperative Complications , Anastomosis, Surgical/adverse effects , Cicatrix/etiology , Cicatrix/pathology , Cicatrix/physiopathology , Duodenum/innervation , Duodenum/pathology , Duodenum/physiopathology , Electric Impedance , Gastric Emptying , Gastric Stump/innervation , Gastric Stump/pathology , Gastric Stump/physiopathology , Gastroparesis/etiology , Gastroparesis/physiopathology , Gastroparesis/surgery , Humans , Immunohistochemistry , Male , Middle Aged , Monitoring, Intraoperative/methods , Postoperative Complications/physiopathology , Postoperative Complications/surgery , Reoperation/methods
10.
FASEB J ; 33(5): 6632-6642, 2019 05.
Article in English | MEDLINE | ID: mdl-30802137

ABSTRACT

The Ca2+-activated Cl- channel, anoctamin 1 (Ano1, also known as transmembrane protein 16A) contributes to intestinal pacemaking, fluid secretion, cellular excitability, and tissue development. The human ANO1 promoter contains binding sites for the glioma-associated oncogene (Gli) proteins. We investigated regulation of ANO1 transcription by Gli. ANO1 promoter activity was determined using a luciferase reporter system. Binding and functional effects of Glis on ANO1 transcription and expression were demonstrated by chromatin immunoprecipitation, small interfering RNA knockdown, PCR, immunolabeling, and recordings of Ca2+-activated Cl- currents in human embryonic kidney 293 (HEK293) cells. Results from previous genome-wide association studies were used to test ANO1 promoter polymorphisms for association with disease. Gli1 and Gli2 bound to the promoter and repressed ANO1 transcription. Repression depended on Gli binding to a site close to the ANO1 transcriptional start site. Mutation of this site prevented Gli binding and transcriptional repression. Knockdown of Gli expression and inhibition of Gli activity increased expression of ANO1 RNA and Ca2+-activated Cl- currents in HEK293 cells. A single-nucleotide polymorphism prevented Gli binding and showed association with irritable bowel syndrome. We conclude that Gli1 and Gli2 repress ANO1 by a novel mechanism that is independent of Gli cleavage and that has a role in gastrointestinal function.-Mazzone, A., Gibbons, S. J., Eisenman, S. T., Strege, P. R., Zheng, T., D'Amato, M., Ordog, T., Fernandez-Zapico, M. E., Farrugia, G. Direct repression of anoctamin 1 (ANO1) gene transcription by Gli proteins.


Subject(s)
Anoctamin-1/biosynthesis , Gene Expression Regulation , Neoplasm Proteins/biosynthesis , Nuclear Proteins/metabolism , Transcription Initiation Site , Transcription, Genetic , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein Gli2/metabolism , Anoctamin-1/genetics , Calcium/metabolism , Calcium Signaling , HEK293 Cells , Humans , Irritable Bowel Syndrome/genetics , Irritable Bowel Syndrome/metabolism , Mutation , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein Gli2/genetics
12.
Annu Int Conf IEEE Eng Med Biol Soc ; 2019: 2765-2768, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31946466

ABSTRACT

Interstitial cells of Cajal (ICC) generate electrical pacemaker activity in the gastrointestinal (GI) tract known as slow waves, which regulate GI motility. ICC express both the Kit receptor tyrosine kinase protein and a Ca2+-activated Cl--channel, encoded by the anoctamin1 (Ano1) protein, which is an essential contributor to the Ca2+ cycling of ICC and slow wave pacemaking. Recent dye-loading imaging studies have demonstrated Ca2+ transients in ICC in isolated tissue preparations. The main aim of this study was to develop a method that allows Ca2+ transients to be registered to structural ICC network data. Confocal image stacks of ICC labeled for Kit or Ano1 and Ca2+ recording data were processed using a thresholding protocol. The Ca2+ transients were then registered to the ICC structural network. First, a general idea of the placement was found by mapping the field-of-view of the Ca2+ transient data to the distorted tissue that contained the ICC network image. The errors in the registration were then corrected for by warping the internal Ca2+ transient field according to the structural network. In data sets from tissues with induced, targeted knockdown of Ano1 expression in a subset of ICC, agreement between the Ca2+ transient data and structural network was 68 ± 10%. This level of agreement allowed selective extraction of Ca2+ data from Ano1-positive (Ano1+) and Ano1-negative (Ano1-) ICC. In the future, this technique will allow investigation into the functional properties of ICC in relation to the level of knockdown of specific ICC associated proteins.


Subject(s)
Interstitial Cells of Cajal , Animals , Anoctamin-1 , Calcium , Calcium Signaling , Gastrointestinal Motility , Mice
13.
Annu Int Conf IEEE Eng Med Biol Soc ; 2019: 4791-4794, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31946933

ABSTRACT

Gastrointestinal (GI) motility is a key component of digestive health, and it is a complex both rhythmic and arrhythmic process governed by many physiological factors. The ability to accurately track the movements of the GI tissues in vitro over multiple days would provide valuable insights into GI tract physiology. A correlational analysis tracking algorithm was developed and applied to intestinal smooth muscle tissues that were maintained in organotypic culture. Physiologically relevant metrics, such as frequency, amplitude and motility index were independently assayed to quantify smooth muscle contractions. The results were validated by manually detected frequency determined using a standard software package. The algorithm was capable of tracking the changes in contractions of the same tissues over six days. This proof-of-concept study demonstrates the feasibility of long-term tracking of GI motility in organotypic cultures over multiple days. The approach allows study of the effects on GI smooth muscle contractility of direct controlled targeting of the cells and molecules in the GI tunica muscularis.


Subject(s)
Gastrointestinal Motility , Gastrointestinal Tract , Muscle, Smooth , Algorithms , Animals , Muscle Contraction , Rats
14.
BMC Med Genomics ; 11(1): 62, 2018 Aug 07.
Article in English | MEDLINE | ID: mdl-30086735

ABSTRACT

BACKGROUND: Cellular changes described in human gastroparesis have revealed a role for immune dysregulation, however, a mechanistic understanding of human gastroparesis and the signaling pathways involved are still unclear. METHODS: Diabetic gastroparetics, diabetic non-gastroparetic controls, idiopathic gastroparetics and non-diabetic non-gastroparetic controls underwent full-thickness gastric body biopsies. Deep RNA sequencing was performed and pathway analysis of differentially expressed transcripts was done using Ingenuity®. A subset of differentially expressed genes in diabetic gastroparesis was validated in a separate cohort using QT-PCR. RESULTS: 111 genes were differentially expressed in diabetic gastroparesis and 181 in idiopathic gastroparesis with a log2fold difference of | ≥ 2| and false detection rate (FDR) < 5%. Top canonical pathways in diabetic gastroparesis included genes involved with macrophages, fibroblasts and endothelial cells in rheumatoid arthritis, osteoarthritis pathway and differential regulation of cytokine production in macrophages and T helper cells by IL-17A and IL-17F. Top canonical pathways in idiopathic gastroparesis included genes involved in granulocyte adhesion and diapedesis, agranulocyte adhesion and diapedesis, and role of macrophages, fibroblasts and endothelial cells in rheumatoid arthritis. Sixty-five differentially expressed genes (log2fold difference | ≥ 2|, FDR < 5%) were common in both diabetic and idiopathic gastroparesis with genes in the top 5 canonical pathways associated with immune signaling. 4/5 highly differentially expressed genes (SGK1, APOLD1, CXCR4, CXCL2, and FOS) in diabetic gastroparesis were validated in a separate cohort of patients using RT-PCR. Immune profile analysis revealed that genes associated with M1 (pro inflammatory) macrophages were enriched in tissues from idiopathic gastroparesis tissues compared to controls (p < 0.05). CONCLUSIONS: Diabetic and idiopathic gastroparesis have both unique and overlapping transcriptomic signatures. Innate immune signaling likely plays a central role in pathogenesis of human gastroparesis.


Subject(s)
Diabetes Complications/genetics , Diabetes Complications/immunology , Gastroparesis/genetics , Gastroparesis/immunology , Gene Expression Profiling , Adult , Diabetes Complications/pathology , Female , Gastroparesis/pathology , Humans , Male , Middle Aged , Signal Transduction/genetics , Young Adult
16.
Gastroenterology ; 154(8): 2122-2136.e12, 2018 06.
Article in English | MEDLINE | ID: mdl-29501441

ABSTRACT

BACKGROUND & AIMS: Muscularis propria macrophages lie close to cells that regulate gastrointestinal motor function, including interstitial cells of Cajal (ICC) and myenteric neurons. In animal models of diabetic gastroparesis, development of delayed gastric emptying has been associated with loss of macrophages that express cytoprotective markers and reduced networks of ICC. Mice with long-term diabetes and normal gastric emptying have macrophages that express anti-inflammatory markers and have normal gastric ICC. Mice homozygous for the osteopetrosis spontaneous mutation in the colony-stimulating factor 1 gene (Csf1op/op) do not have macrophages; when they are given streptozotocin to induce diabetes, they do not develop delayed gastric emptying. We investigated whether population of the gastric muscularis propria of diabetic Csf1op/op mice with macrophages is necessary to change gastric emptying, ICC, and myenteric neurons and investigated the macrophage-derived factors that determine whether diabetic mice do or do not develop delayed gastric emptying. METHODS: Wild-type and Csf1op/op mice were given streptozotocin to induce diabetes. Some Csf1op/op mice were given daily intraperitoneal injections of CSF1 for 7 weeks; gastric tissues were collected and cellular distributions were analyzed by immunohistochemistry. CD45+, CD11b+, F4/80+ macrophages were dissociated from gastric muscularis propria, isolated by flow cytometry and analyzed by quantitative real-time polymerase chain reaction. Cultured gastric muscularis propria from Csf1op/op mice was exposed to medium that was conditioned by culture with bone marrow-derived macrophages from wild-type mice. RESULTS: Gastric muscularis propria from Csf1op/op mice given CSF1 contained macrophages; 11 of 15 diabetic mice given CSF1 developed delayed gastric emptying and had damaged ICC. In non-diabetic Csf1op/op mice, administration of CSF1 reduced numbers of gastric myenteric neurons but did not affect the proportion of nitrergic neurons or ICC. In diabetic Csf1op/op mice given CSF1 that developed delayed gastric emptying, the proportion of nitrergic neurons was the same as in non-diabetic wild-type controls. Medium conditioned by macrophages previously exposed to oxidative injury caused damage to ICC in cultured gastric muscularis propria from Csf1op/op mice; neutralizing antibodies against IL6R or TNF prevented this damage to ICC. CD45+, CD11b+, and F4/80+ macrophages isolated from diabetic wild-type mice with delayed gastric emptying expressed higher levels of messenger RNAs encoding inflammatory markers (IL6 and inducible nitric oxide synthase) and lower levels of messenger RNAs encoding markers of anti-inflammatory cells (heme oxygenase 1, arginase 1, and FIZZ1) than macrophages isolated from diabetic mice with normal gastric emptying. CONCLUSIONS: In studies of Csf1op/op and wild-type mice with diabetes, we found delayed gastric emptying to be associated with increased production of inflammatory factors, and reduced production of anti-inflammatory factors, by macrophages, leading to loss of ICC.


Subject(s)
Diabetes Mellitus, Experimental/complications , Gastric Emptying/physiology , Gastroparesis/physiopathology , Macrophages/physiology , Stomach/physiopathology , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Gastroparesis/etiology , Humans , Immunohistochemistry , Interstitial Cells of Cajal/physiology , Macrophage Colony-Stimulating Factor/genetics , Mice , Muscle, Smooth/cytology , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , Mutation , Stomach/cytology , Stomach/pathology , Streptozocin/toxicity
17.
Am J Physiol Gastrointest Liver Physiol ; 314(4): G494-G503, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29167113

ABSTRACT

The SCN5A-encoded voltage-gated mechanosensitive Na+ channel NaV1.5 is expressed in human gastrointestinal smooth muscle cells and interstitial cells of Cajal. NaV1.5 contributes to smooth muscle electrical slow waves and mechanical sensitivity. In predominantly Caucasian irritable bowel syndrome (IBS) patient cohorts, 2-3% of patients have SCN5A missense mutations that alter NaV1.5 function and may contribute to IBS pathophysiology. In this study we examined a racially and ethnically diverse cohort of IBS patients for SCN5A missense mutations, compared them with IBS-negative controls, and determined the resulting NaV1.5 voltage-dependent and mechanosensitive properties. All SCN5A exons were sequenced from somatic DNA of 252 Rome III IBS patients with diverse ethnic and racial backgrounds. Missense mutations were introduced into wild-type SCN5A by site-directed mutagenesis and cotransfected with green fluorescent protein into HEK-293 cells. NaV1.5 voltage-dependent and mechanosensitive functions were studied by whole cell electrophysiology with and without shear force. Five of 252 (2.0%) IBS patients had six rare SCN5A mutations that were absent in 377 IBS-negative controls. Six of six (100%) IBS-associated NaV1.5 mutations had voltage-dependent gating abnormalities [current density reduction (R225W, R433C, R986Q, and F1293S) and altered voltage dependence (R225W, R433C, R986Q, G1037V, and F1293S)], and at least one kinetic parameter was altered in all mutations. Four of six (67%) IBS-associated SCN5A mutations (R225W, R433C, R986Q, and F1293S) resulted in altered NaV1.5 mechanosensitivity. In this racially and ethnically diverse cohort of IBS patients, we show that 2% of IBS patients harbor SCN5A mutations that are absent in IBS-negative controls and result in NaV1.5 channels with abnormal voltage-dependent and mechanosensitive function. NEW & NOTEWORTHY The voltage-gated Na+ channel NaV1.5 contributes to smooth muscle physiology and electrical slow waves. In a racially and ethnically mixed irritable bowel syndrome cohort, 2% had mutations in the NaV1.5 gene SCN5A. These mutations were absent in irritable bowel syndrome-negative controls. Most mutant NaV1.5 channels were loss of function in voltage dependence or mechanosensitivity.


Subject(s)
Gastrointestinal Tract , Irritable Bowel Syndrome , Myocytes, Smooth Muscle/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Adult , Aged , Channelopathies/genetics , Channelopathies/physiopathology , Electrophysiological Phenomena/genetics , Female , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Gastrointestinal Tract/physiopathology , Genetic Predisposition to Disease , Humans , Irritable Bowel Syndrome/genetics , Irritable Bowel Syndrome/metabolism , Irritable Bowel Syndrome/pathology , Male , Middle Aged , Mutation, Missense , NAV1.5 Voltage-Gated Sodium Channel/genetics , Patch-Clamp Techniques
18.
Exp Physiol ; 103(1): 40-57, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28971566

ABSTRACT

NEW FINDINGS: What is the central question of this study? The aim was to investigate the roles of extracellular chloride in electrical slow waves and resting membrane potential of mouse jejunal smooth muscle by replacing chloride with the impermeant anions gluconate and isethionate. What is the main finding and its importance? The main finding was that in smooth muscle cells, the resting Cl- conductance is low, whereas transmembrane Cl- movement in interstitial cells of Cajal (ICCs) is a major contributor to the shape of electrical slow waves. Furthermore, the data confirm that ICCs set the smooth muscle membrane potential and that altering Cl- homeostasis in ICCs can alter the smooth muscle membrane potential. Intracellular Cl- homeostasis is regulated by anion-permeable channels and transporters and contributes to excitability of many cell types, including smooth muscle and interstitial cells of Cajal (ICCs). Our aims were to investigate the effects on electrical activity in mouse jejunal muscle strips of replacing extracellular Cl- (Cl-o ) with the impermeant anions gluconate and isethionate. On reducing Cl-o , effects were observed on electrical slow waves, with small effects on smooth muscle membrane voltage (Em ). Restoration of Cl- hyperpolarized smooth muscle Em proportional to the change in Cl-o concentration. Replacement of 90% of Cl-o with gluconate reversibly abolished slow waves in five of nine preparations. Slow waves were maintained in isethionate. Gluconate and isethionate substitution had similar concentration-dependent effects on peak amplitude, frequency, width at half peak amplitude, rise time and decay time of residual slow waves. Gluconate reduced free ionized Ca2+ in Krebs solutions to 0.13 mm. In Krebs solutions containing normal Cl- and 0.13 mm free Ca2+ , slow wave frequency was lower, width at half peak amplitude was smaller, and decay time was faster. The transient hyperpolarization following restoration of Cl-o was not observed in W/Wv mice, which lack pacemaker ICCs in the small intestine. We conclude that in smooth muscle cells, the resting Cl- conductance is low, whereas transmembrane Cl- movement in ICCs plays a major role in generation or propagation of slow waves. Furthermore, these data support a role for ICCs in setting smooth muscle Em and that altering Cl- homeostasis in ICCs can alter smooth muscle Em .


Subject(s)
Chlorides/physiology , Extracellular Fluid/physiology , Interstitial Cells of Cajal/physiology , Jejunum/physiology , Membrane Potentials/physiology , Muscle, Smooth/physiology , Animals , Chlorides/pharmacology , Extracellular Fluid/drug effects , Female , Interstitial Cells of Cajal/drug effects , Jejunum/cytology , Jejunum/drug effects , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Muscle, Smooth/drug effects , Organ Culture Techniques
19.
PLoS One ; 12(11): e0187772, 2017.
Article in English | MEDLINE | ID: mdl-29161307

ABSTRACT

BACKGROUND: Idiopathic and diabetic gastroparesis in Homo sapiens cause significant morbidity. Etiology or risk factors have not been clearly identified. Failure to sustain elevated heme oxygenase-1 (HO1) expression is associated with delayed gastric emptying in diabetic mice and polymorphisms in the HO1 gene (HMOX1, NCBI Gene ID:3162) are associated with worse outcomes in other diseases. AIM: Our hypothesis was that longer polyGT alleles are more common in the HMOX1 genes of individuals with gastroparesis than in controls without upper gastrointestinal motility disorders. METHODS: Repeat length was determined in genomic DNA. Controls with diabetes (84 type 1, 84 type 2) and without diabetes (n = 170) were compared to diabetic gastroparetics (99 type 1, 72 type 2) and idiopathic gastroparetics (n = 234). Correlations of repeat lengths with clinical symptom sub-scores on the gastroparesis cardinal symptom index (GCSI) were done. Statistical analyses of short (<29), medium and long (>32) repeat alleles and differences in allele length were used to test for associations with gastroparesis. RESULTS: The distribution of allele lengths was different between groups (P = 0.016). Allele lengths were longest in type 2 diabetics with gastroparesis (29.18±0.35, mean ± SEM) and longer in gastroparetics compared to non-diabetic controls (28.50±0.14 vs 27.64±0.20 GT repeats/allele, P = 0.0008). Type 2 diabetic controls had longer alleles than non-diabetic controls. In all gastroparetic groups, allele lengths were longer in African Americans compared to other racial groups, differences in the proportion of African Americans in the groups accounted for the differences between gastroparetics and controls. Diabetic gastroparetics with 1 or 2 long alleles had worse GCSI nausea sub-scores (3.30±0.23) as compared to those with 0 long alleles (2.66±0.12), P = 0.022. CONCLUSIONS: Longer poly-GT repeats in the HMOX1 gene are more common in African Americans with gastroparesis. Nausea symptoms are worse in subjects with longer alleles.


Subject(s)
Diabetes Mellitus/genetics , Gastroparesis/genetics , Heme Oxygenase-1/genetics , Tandem Repeat Sequences/genetics , Adult , Black or African American/genetics , Aged , Alleles , Animals , Diabetes Complications/genetics , Diabetes Complications/pathology , Diabetes Mellitus/pathology , Female , Gastric Emptying/genetics , Gastroparesis/pathology , Humans , Male , Mice , Middle Aged , Polymorphism, Genetic
20.
Gastroenterology ; 153(2): 521-535.e20, 2017 08.
Article in English | MEDLINE | ID: mdl-28438610

ABSTRACT

BACKGROUND & AIMS: Depletion of interstitial cells of Cajal (ICCs) is common in diabetic gastroparesis. However, in approximately 20% of patients with diabetes, gastric emptying (GE) is accelerated. GE also occurs faster in obese individuals, and is associated with increased blood levels of glucose in patients with type 2 diabetes. To understand the fate of ICCs in hyperinsulinemic, hyperglycemic states characterized by rapid GE, we studied mice with mutation of the leptin receptor (Leprdb/db), which in our colony had accelerated GE. We also investigated hyperglycemia-induced signaling in the ICC lineage and ICC dependence on glucose oxidative metabolism in mice with disruption of the succinate dehydrogenase complex, subunit C gene (Sdhc). METHODS: Mice were given breath tests to analyze GE of solids. ICCs were studied by flow cytometry, intracellular electrophysiology, isometric contractility measurement, reverse-transcription polymerase chain reaction, immunoblot, immunohistochemistry, enzyme-linked immunosorbent assays, and metabolite assays; cells and tissues were manipulated pharmacologically and by RNA interference. Viable cell counts, proliferation, and apoptosis were determined by methyltetrazolium, Ki-67, proliferating cell nuclear antigen, bromodeoxyuridine, and caspase-Glo 3/7 assays. Sdhc was disrupted in 2 different strains of mice via cre recombinase. RESULTS: In obese, hyperglycemic, hyperinsulinemic female Leprdb/db mice, GE was accelerated and gastric ICC and phasic cholinergic responses were increased. Female KitK641E/+ mice, which have genetically induced hyperplasia of ICCs, also had accelerated GE. In isolated cells of the ICC lineage and gastric organotypic cultures, hyperglycemia stimulated proliferation by mitogen-activated protein kinase 1 (MAPK1)- and MAPK3-dependent stabilization of ets variant 1-a master transcription factor for ICCs-and consequent up-regulation of v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog (KIT) receptor tyrosine kinase. Opposite changes occurred in mice with disruption of Sdhc. CONCLUSIONS: Hyperglycemia increases ICCs via oxidative metabolism-dependent, MAPK1- and MAPK3-mediated stabilization of ets variant 1 and increased expression of KIT, causing rapid GE. Increases in ICCs might contribute to the acceleration in GE observed in some patients with diabetes.


Subject(s)
DNA-Binding Proteins/physiology , Gastric Emptying/physiology , Hyperglycemia/physiopathology , Interstitial Cells of Cajal/cytology , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins c-kit/physiology , Transcription Factors/physiology , Animals , Female , Humans , Interstitial Cells of Cajal/physiology , Mice , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Receptors, Leptin/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...