Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Am J Physiol Gastrointest Liver Physiol ; 326(4): G398-G410, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38290993

ABSTRACT

Major esophageal disorders involve obstructive transport of bolus to the stomach, causing symptoms of dysphagia and impaired clearing of the refluxed gastric contents. These may occur due to mechanical constriction of the esophageal lumen or loss of relaxation associated with deglutitive inhibition, as in achalasia-like disorders. Recently, immune inflammation has been identified as an important cause of esophageal strictures and the loss of inhibitory neurotransmission. These disorders are also associated with smooth muscle hypertrophy and hypercontractility, whose cause is unknown. This review investigated immune inflammation in the causation of smooth muscle changes in obstructive esophageal bolus transport. Findings suggest that smooth muscle hypertrophy occurs above the obstruction and is due to mechanical stress on the smooth muscles. The mechanostressed smooth muscles release cytokines and other molecules that may recruit and microlocalize mast cells to smooth muscle bundles, so that their products may have a close bidirectional effect on each other. Acting in a paracrine fashion, the inflammatory cytokines induce genetic and epigenetic changes in the smooth muscles, leading to smooth muscle hypercontractility, hypertrophy, and impaired relaxation. These changes may worsen difficulty in the esophageal transport. Immune processes differ in the first phase of obstructive bolus transport, and the second phase of muscle hypertrophy and hypercontractility. Moreover, changes in the type of mechanical stress may change immune response and effect on smooth muscles. Understanding immune signaling in causes of obstructive bolus transport, type of mechanical stress, and associated smooth muscle changes may help pathophysiology-based prevention and targeted treatment of esophageal motility disorders.NEW & NOTEWORTHY Esophageal disorders such as esophageal stricture or achalasia, and diffuse esophageal spasm are associated with smooth muscle hypertrophy and hypercontractility, above the obstruction, yet the cause of such changes is unknown. This review suggests that smooth muscle obstructive disorders may cause mechanical stress on smooth muscle, which then secretes chemicals that recruit, microlocalize, and activate mast cells to initiate immune inflammation, producing functional and structural changes in smooth muscles. Understanding the immune signaling in these changes may help pathophysiology-based prevention and targeted treatment of esophageal motility disorders.


Subject(s)
Esophageal Achalasia , Esophageal Motility Disorders , Humans , Mast Cells , Manometry , Muscle, Smooth , Inflammation , Cytokines , Hypertrophy
2.
Am J Respir Crit Care Med ; 207(2): 130-137, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36214830

ABSTRACT

Rationale: Gastroesophageal reflux disease (GERD) is commonly associated with atopic disorders, but cause-effect relationships remain unclear. Objectives: We applied Mendelian randomization analysis to explore whether GERD is causally related to atopic disorders of the lung (asthma) and/or skin (atopic dermatitis [AD]). Methods: We conducted two-sample bidirectional Mendelian randomization to infer the magnitude and direction of causality between asthma and GERD, using summary statistics from the largest genome-wide association studies conducted on asthma (Ncases = 56,167) and GERD (Ncases = 71,522). In addition, we generated instrumental variables for AD from the latest population-level genome-wide association study meta-analysis (Ncases = 22,474) and assessed their fidelity and confidence of predicting the likely causal pathway(s) leading to asthma and/or GERD. Measurements and Main Results: Applying three different methods, each method revealed similar magnitude of causal estimates that were directionally consistent across the sensitivity analyses. Using an inverse variance-weighted method, the largest effect size was detected for asthma predisposition to AD (odds ratio [OR], 1.46; 95% confidence interval [CI], 1.34-1.59), followed by AD to asthma (OR, 1.34; 95% CI, 1.24-1.45). A significant association was detected for genetically determined asthma on risk of GERD (OR, 1.06; 95% CI, 1.03-1.09) but not genetically determined AD on GERD. In contrast, GERD equally increased risks of asthma (OR, 1.21; 95% CI, 1.09-1.35) and AD (OR, 1.21; 95% CI, 1.07-1.37). Conclusions: This study uncovers previously unrecognized causal pathways that have clinical implications in European-ancestry populations: 1) asthma is a causal risk for AD, and 2) the predisposition to AD, including asthma, can arise from specific pathogenic mechanisms manifested by GERD.


Subject(s)
Asthma , Dermatitis, Atopic , Gastroesophageal Reflux , Humans , Dermatitis, Atopic/epidemiology , Dermatitis, Atopic/genetics , Mendelian Randomization Analysis , Genome-Wide Association Study , Asthma/epidemiology , Asthma/genetics , Gastroesophageal Reflux/epidemiology , Gastroesophageal Reflux/genetics , Polymorphism, Single Nucleotide
3.
J Clin Med ; 11(22)2022 Nov 10.
Article in English | MEDLINE | ID: mdl-36431141

ABSTRACT

Oropharyngeal and esophageal dysmotility can cause serious clinical complications such as aspiration pneumonia, cachexia, and sarcopenia, with a resulting increase in mortality and disability. The current standard of care for the treatment of SSc-associated swallowing dysfunction is mainly supportive, although severe cases are usually refractory to conventional management. Recent studies have shown that the abnormal production of functional autoantibodies such as anti-cholinergic muscarinic receptor III antibodies may participate in the pathogenesis of SSc-associated gastrointestinal dysmotility and may provide a novel target for therapeutic intervention. We describe two patients with severe and rapid onset of SSc-associated severe swallowing dysfunction and esophageal dysmotility who had failed standard of care therapy, requiring complete enteral and parenteral nutrition. Both patients were positive for the presence of circulating antimuscarinic III receptor antibodies. They were treated with IVIG at a dose of 2 g/Kg/month divided in two consecutive days, for six months. Following IVIG therapy, both patients markedly improved their symptoms as shown by a reduction in their UCLA2.0 score, and achieved an improvement of esophageal motility documented radiologically. Both patients resumed oral feeding and had their feeding tubes removed within the treatment period. None of the patients developed severe adverse events attributable to IVIG, except for low-grade fever during IVIG infusion in one of the cases. These results provide support for the role of functional autoantibodies in the development of SSc-associated gastrointestinal dysfunction.

5.
Neurogastroenterol Motil ; 34(4): e14099, 2022 04.
Article in English | MEDLINE | ID: mdl-33624396

ABSTRACT

BACKGROUND: Data on the neuromodulatory effects of brain-derived neurotrophic factor (BDNF) in the gastrointestinal tract were recently reported, but there are still no data on the presence, distribution, and release of BDNF in the gastrointestinal tract, including the internal anal sphincter (IAS). METHODS: We examined the presence and distribution of BDNF and its receptor TrkB in the different IAS structures (neuronal and non-neuronal) via immunohistochemical and immunocytochemical analyses. We also monitored the release of BDNF in an IAS muscle bath (consisting of smooth muscle cells [SMCs], myenteric plexus, and submucosal plexus) before and after different agonists, and electrical field stimulation in the absence and presence of neurotoxin tetrodotoxin. KEY RESULTS: BDNF/TrkB was found to be present in all layers of the IAS, especially the smooth muscle, mucosa, myenteric plexus, and submucosal plexus. Detailed analyses revealed a significant colocalization between BDNF and TrkB in different structures, especially in the smooth muscle, the SMCs, and both plexuses. Data further showed higher levels of BDNF in the cytosol and that of TrkB toward the periphery of the SMCs. CONCLUSIONS & INFERENCES: These studies showed that BDNF/TrkB was present not only in the enteric nervous system (ENS), but also in the SMCs. For the neuromodulatory effects, BDNF is released locally from the ENS ((myenteric (10.01 ± 0.23 pg/ml) and submucosal plexus (9.05 ± 0.51 pg/ml)) and the SMCs (18.63 ± 1.63 pg/ml). Collectively, these findings have pathophysiological and therapeutic implications regarding the role of BDNF/TrkB in the IAS-associated rectoanal motility disorders.


Subject(s)
Brain-Derived Neurotrophic Factor , Enteric Nervous System , Anal Canal/physiology , Brain-Derived Neurotrophic Factor/pharmacology , Muscle, Smooth , Neurons
6.
Am J Physiol Gastrointest Liver Physiol ; 321(1): G87-G97, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34075793

ABSTRACT

Aging can lead to rectoanal incontinence due to internal anal sphincter (IAS) dysfunction, which is characterized by a decrease in IAS tone and contractility and an increase in nonadrenergic noncholinergic (NANC) relaxation. We aimed to determine whether brain-derived neurotropic factor (BDNF) rescues this aging-associated IAS dysfunction (AAID). To do so, we studied the effects of BDNF on the basal and G protein-coupled receptors (GPCR)-stimulated IAS smooth muscle tone and on NANC relaxation in Fischer 344 rats representing different age groups [26-mo-old (aging) vs. 6-mo-old (young)], before and after tyrosine kinase receptor B (TrkB) antagonist K252a. We also used isolated smooth muscle cells (SMCs) to determine the effects of BDNF before and after different agonists. For some studies, we monitored NO release using smooth muscle perfusates. BDNF reversed AAID by rescuing the basal IAS tone and agonists [thromboxane A2 analog (U46619) and angiotensin II (ANG II)]-induced contractility, and NANC relaxation. These rescue effects of BDNF were selective as K252a attenuated the changes in the IAS without modifying the effects of K+depolarization. Because of the direct association between the basal and GPCR-stimulated IAS tone and RhoA/ROCK activation, we speculate that this pathway in the rescue effects of BDNF. Conversely, our data suggest that aging-associated increased NANC relaxation is reversed by decreased release of NO and decrease in the sensitivity of the released inhibitory neurotransmitter. In summary, BDNF rescue of AAID involves RhoA/ROCK and inhibitory neurotransmission. These data have direct implications for the role of BDNF in the pathophysiology and therapeutic targeting of aging-associated rectoanal motility disorders.NEW & NOTEWORTHY These studies demonstrate that brain-derived neurotropic factor (BDNF) rescues the aging-associated internal anal sphincter (IAS) dysfunction, characterized by a decrease in IAS tone, and increase in non-adrenergic noncholinergic relaxation. We determined the effects of BDNF on the basal and GPCR (TXA2 and ANG II)-stimulated IAS tone, and on NANC relaxation, before and after TrkB inhibitor K252a. BDNF may have an important role in the pathophysiology and therapeutic targeting of certain rectoanal motility disorders.


Subject(s)
Aging/drug effects , Brain-Derived Neurotrophic Factor/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Animals , Brain/drug effects , Brain/metabolism , Male , Muscle Relaxation/drug effects , Muscle Tonus/physiology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Rats , Rats, Inbred F344 , Signal Transduction/physiology , rho-Associated Kinases/metabolism
7.
FASEB J ; 34(2): 2126-2146, 2020 02.
Article in English | MEDLINE | ID: mdl-31909533

ABSTRACT

Bladder dysfunction is associated with the overexpression of the intermediate filament (IF) proteins desmin and vimentin in obstructed bladder smooth muscle (BSM). However, the mechanisms by which these proteins contribute to BSM dysfunction are not known. Previous studies have shown that desmin and vimentin directly participate in signal transduction. In this study, we hypothesized that BSM dysfunction associated with overexpression of desmin or vimentin is mediated via c-Jun N-terminal kinase (JNK). We employed a model of murine BSM tissue in which increased expression of desmin or vimentin was induced by adenoviral transduction to examine the sufficiency of increased IF protein expression to reduce BSM contraction. Murine BSM strips overexpressing desmin or vimentin generated less force in response to KCl and carbachol relative to the levels in control murine BSM strips, an effect associated with increased JNK2 phosphorylation and reduced myosin light chain (MLC20 ) phosphorylation. Furthermore, desmin and vimentin overexpressions did not alter BSM contractility and MLC20 phosphorylation in strips isolated from JNK2 knockout mice. Pharmacological JNK2 inhibition produced results qualitatively similar to those caused by JNK2 knockout. These findings suggest that inhibition of JNK2 may improve diminished BSM contractility associated with obstructive bladder disease.


Subject(s)
Desmin/biosynthesis , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 9/metabolism , Muscle Contraction , Muscle, Smooth/metabolism , Urinary Bladder/metabolism , Vimentin/biosynthesis , Animals , Desmin/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 9/genetics , Muscle, Smooth/cytology , Urinary Bladder/cytology , Vimentin/genetics
8.
Am J Physiol Gastrointest Liver Physiol ; 318(1): G23-G33, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31682160

ABSTRACT

Presently, there are no studies examining the neuromodulatory effects of brain-derived neurotropic factor (BDNF) on the basal internal anal sphincter (IAS) tone and nonadrenergic noncholinergic (NANC) relaxation. To examine this, we determined the neuromuscular effects of BDNF on basal IAS smooth muscle tone and the smooth muscle cells (SMCs) and the effects of NANC nerve stimulation before and after high-affinity receptor tyrosine kinase receptor B (TrkB) antagonist K252a. We also investigated the mechanisms underlying BDNF-augmented increase in the IAS tone and NANC relaxation. We found that BDNF-increased IAS tone and SMC contractility were TTX resistant and attenuated by K252a. TrkB-specific agonist 7,8-dihydroxyflavone, similar to BDNF, also produced a concentration-dependent increase in the basal tone, whereas TrkB inhibitors K252a and ANA-12 produced a decrease in the tone. In addition, BDNF produced leftward shifts in the concentration-response curves with U46619 and ANG II (but not with bethanechol and K+ depolarization), and these shifts were reversed by K252a. Effects of Y27632 and Western blot data indicated that the BDNF-induced increase in IAS tone was mediated via RhoA/ROCK. BDNF-augmented NANC relaxation by electrical field stimulation was found to be mediated via the nitric oxide (NO)/soluble guanylate cyclase (sGC) pathway rather than via increased sensitivity to NO. In conclusion, the net effect of BDNF was that it caused an increase in the basal IAS tone via RhoA/ROCK signaling. BDNF also augmented NANC relaxation via NO/sGC. These findings may have relevance to the role of BDNF in the pathophysiology and therapeutic targeting of the IAS-associated rectoanal motility disorders.NEW & NOTEWORTHY These studies for the first time to our knowledge demonstrate that increased levels of brain-derived neurotrophic factor (BDNF; conceivably released from smooth muscle cells and/or the enteric neurons) has two major effects. First, BDNF augments the internal anal sphincter (IAS) tone via tyrosine kinase receptor B/thromboxane A2-receptor, angiotensin II receptor type 1/RhoA/ROCK signaling; and second, it increases nonadrenergic noncholinergic relaxation via nitric oxide/soluble guanylate cyclase. These studies may have relevance in therapeutic targeting in the anorectal motility disorders associated with the IAS.


Subject(s)
Anal Canal/innervation , Brain-Derived Neurotrophic Factor/pharmacology , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Nitrergic Neurons/metabolism , Nitric Oxide/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Animals , Dose-Response Relationship, Drug , Electric Stimulation , In Vitro Techniques , Male , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism , Receptor, trkB/agonists , Receptor, trkB/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Signal Transduction , Soluble Guanylyl Cyclase/metabolism
9.
Sci Rep ; 9(1): 6759, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31043680

ABSTRACT

Aging-associated decrease in internal anal sphincter (IAS) tone (AADI) is a major contributor in the rectoanal incontinence (RI). To determine the pathogenesis of AADI, we investigated the effect of aging on GPCR activation and related downstream signaling. We particularly investigated two GPCRs that characterize IAS smooth muscle cells (SMCs): thromboxane A2 and angiotensin II type 1. Two groups of Fischer 344 rats (6-month-old [young group] and 26-month-old [old group]) were employed to determine the GPCR function by isometric contraction, the expressions of GPCRs, and their downstream regulatory signaling proteins (regulator of G-protein signaling 2, RGS2; GPCR Kinase 5, GRK5; and ß-arrestin, Arrb2) using RT-PCR, qPCR, and western blot analyses. We used reversible biotinylation to monitor the GPCR trafficking using SMCs. Aging selectively attenuated thromboxane A2 and Ang II-induced IAS contraction. RT-PCR, qPCR, and WB data revealed a significant decrease in the expressions of the GPCRs and increase in the expression of RGS2, GRK5, and Arrb2. The increased GPCR internalization and decreased recycling under aging were validated by reversible biotinylation. We conclude that downregulation of GPCR, accompanied by upregulation of regulatory proteins, plays an important role in receptor desensitization and may be important underlying mechanisms of RI in certain aging patients.


Subject(s)
Aging , Anal Canal/pathology , Fecal Incontinence/epidemiology , Muscle Tonus , Receptor, Angiotensin, Type 1/metabolism , Rectum/pathology , Thromboxane A2/metabolism , Anal Canal/metabolism , Animals , Fecal Incontinence/metabolism , Fecal Incontinence/pathology , Incidence , Male , Muscle Contraction , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Pennsylvania/epidemiology , Rats , Rats, Inbred F344 , Receptor, Angiotensin, Type 1/genetics , Rectum/metabolism , Thromboxane A2/genetics
10.
Am J Pathol ; 189(4): 847-867, 2019 04.
Article in English | MEDLINE | ID: mdl-30707892

ABSTRACT

Caveolins (CAVs) are structural proteins of caveolae that function as signaling platforms to regulate smooth muscle contraction. Loss of CAV protein expression is associated with impaired contraction in obstruction-induced bladder smooth muscle (BSM) hypertrophy. In this study, microarray analysis of bladder RNA revealed down-regulation of CAV1, CAV2, and CAV3 gene transcription in BSM from models of obstructive bladder disease in mice and humans. We identified and characterized regulatory regions responsible for CAV1, CAV2, and CAV3 gene expression in mice with obstruction-induced BSM hypertrophy, and in men with benign prostatic hyperplasia. DNA affinity chromatography and chromatin immunoprecipitation assays revealed a greater increase in binding of GATA-binding factor 6 (GATA-6) and NF-κB to their cognate binding motifs on CAV1, CAV2, and CAV3 promoters in obstructed BSM relative to that observed in control BSM. Knockout of NF-κB subunits, shRNA-mediated knockdown of GATA-6, or pharmacologic inhibition of GATA-6 and NF-κB in BSM increased CAV1, CAV2, and CAV3 transcription and promoter activity. Conversely, overexpression of GATA-6 decreased CAV2 and CAV3 transcription and promoter activity. Collectively, these data provide new insight into the mechanisms by which CAV gene expression is repressed in hypertrophied BSM in obstructive bladder disease.


Subject(s)
Caveolins/antagonists & inhibitors , GATA6 Transcription Factor/metabolism , Hypertrophy/pathology , Muscle, Smooth/pathology , NF-kappa B/metabolism , Transcription, Genetic , Urinary Bladder Neck Obstruction/complications , Aged , Animals , Biomarkers/analysis , Caveolins/genetics , Caveolins/metabolism , GATA6 Transcription Factor/genetics , Gene Expression Profiling , Gene Expression Regulation , Humans , Hypertrophy/etiology , Hypertrophy/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Muscle Contraction , Muscle, Smooth/metabolism , NF-kappa B/genetics , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Urinary Bladder Neck Obstruction/surgery
11.
Eur J Pharmacol ; 827: 22-31, 2018 May 15.
Article in English | MEDLINE | ID: mdl-29524386

ABSTRACT

Extracellular pH is an important physiological determinant of vascular tone that is normally maintained within 7.35-7.45. Any change outside this range leads to severe pathological repercussions. We investigated the unknown effects of extracellular acidosis on relaxation in the superior mesenteric artery (SMA) of goat. SMA rings were employed to maintain isometric contractions at extracellular pH (pHo) 7.4 and 6.8. We analyzed the effect of acidosis (pHo 6.8) compared to physiological pH (pHo 7.4) on three signaling mediators of endothelium-dependent hyperpolarization: nitric oxide (NO), prostaglandin I2 (PGI2), and myoendothelial gap junctions (MEGJ). NO and cyclic guanosine monophosphate (cGMP) levels were compared between normal and acidic pH. Quantitative real-time PCR (qPCR) studies determined the change in expression of vascular connexin (Cx), Cx37, Cx40, and Cx43. Under acidosis, acetyl choline-induced relaxation was augmented in an endothelium-dependent manner via eNOS-NO-cGMP signaling. Conversely, at normal pH, acetyl choline-induced vasorelaxation was mediated primarily via COX-PGI2 pathway. The functional activity of MEGJ was increased under acidosis as evident from increased sensitivity of connexin blockers and upregulated gene and protein expression of connexins. In conclusion, acetyl choline-induced augmented vasorelaxation under acidosis is mediated by NOS-NO-cGMP, with a partial role of MEGJ as EDH mediators in the SMA. Present data suggest a novel role of connexin as therapeutic targets to attenuate the detrimental effect of acidosis on vascular tone.


Subject(s)
Acidosis/pathology , Acidosis/physiopathology , Endothelium, Vascular/metabolism , Gap Junctions/metabolism , Mesenteric Artery, Superior/pathology , Mesenteric Artery, Superior/physiopathology , Vasodilation , Animals , Cell Communication , Cyclic GMP/metabolism , Epoprostenol/metabolism , Goats , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism
12.
Am J Physiol Gastrointest Liver Physiol ; 314(1): G109-G118, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29051187

ABSTRACT

In these studies, we developed a novel approach of in vivo magnetofection for localized delivery of nucleic acids such as micro-RNA-139-5p (miR-139-5p; which is known to target Rho kinase2) to the circular smooth muscle layer of the internal anal sphincter (IAS). The IAS tone is known to play a major role in the rectoanal continence via activation of RhoA-associated kinase (RhoA/ROCK2). These studies established an optimized protocol for efficient gene delivery using an assembly of equal volumes of in vivo PolyMag and miR139-5p or anti-miR-139-5p (100 nM each) injected in the circular smooth muscle layer in the pinpointed areas of the rat perianal region and then incubated for 20 min under magnetic field. Magnetofection efficiency was confirmed and analyzed by confocal microscopy of FITC-tagged siRNA. Using physiological and biochemical approaches, we investigated the effects of miR-139-5p and anti-miR-139-5p on basal intraluminal IAS pressure (IASP), fecal pellet count, IAS tone, agonist-induced contraction, contraction-relaxation kinetics, and RhoA/ROCK2 signaling. Present studies demonstrate that magnetofection-mediated miR-139-5p delivery significantly decreased RhoA/ROCK2, p-MYPT1, and p-MLC20 signaling, leading to decreases in the basal IASP and IAS tone and in rates of contraction and relaxation associated with increase in fecal pellet output. Interestingly, anti-miR-139-5p transfection had opposite effects on these parameters. Collectively, these data demonstrate that magnetofection is a promising novel method of in vivo gene delivery and of nucleotides to the internal anal sphincter for the site-directed and targeted therapy for rectoanal motility disorders. NEW & NOTEWORTHY These studies for the first time demonstrate the success of topical in vivo magnetofection (MF) of nucleic acids using perianal injections. To demonstrate its effectiveness, we used FITC-tagged siRNA via immunofluorescence microcopy and functional and biochemical evidence using miR-139-5p (which is known to target ROCK2). In conclusion, MF allows safe, convenient, efficient, and targeted delivery of oligonucleotides such as siRNAs and microRNAs. These studies have direct therapeutic implications in rectoanal motility disorders especially associated with IAS.


Subject(s)
Anal Canal/metabolism , Antagomirs/administration & dosage , Anus Diseases/therapy , Gastrointestinal Motility , Gene Transfer Techniques , Magnetics/methods , Magnetite Nanoparticles , MicroRNAs/administration & dosage , Animals , Antagomirs/genetics , Antagomirs/metabolism , Anus Diseases/genetics , Anus Diseases/metabolism , Anus Diseases/physiopathology , Defecation , Injections , Kinetics , MicroRNAs/genetics , MicroRNAs/metabolism , Myosin Light Chains/metabolism , Phosphorylation , Pressure , Protein Phosphatase 1/metabolism , Rats, Sprague-Dawley , Signal Transduction , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
13.
Sci Rep ; 7(1): 1477, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28469189

ABSTRACT

The present study focused on the role of microRNA-139-5p (miRNA-139-5p) in the regulation of basal tone in internal anal sphincter (IAS). Applying genome-wide miRNA microarrays on the phenotypically distinct smooth muscle cells (SMCs) within the rat anorectrum, we identified miRNA-139-5p as differentially expressed RNA repressor with highest expression in the purely phasic smooth muscle of anococcygeus (ASM) vs. the truly tonic smooth muscle of IAS. This pattern of miRNA-139-5p expression, previously shown to target ROCK2, was validated by target prediction using ingenuity pathway (IPA) and by qPCR analyses. Immunoblotting, immunocytochemistry (ICC), and functional assays using IAS tissues and cells subjected to overexpression/knockdown of miRNA-139-5p confirmed the inverse relationship between miRNA-139-5p and ROCK2 expressions/IAS tone. Overexpression of miRNA-139-5p caused a decrease, while knockdown by anti-miRNA-139-5p caused an increase in the IAS tone; these tissue contractile responses were confirmed by single-cell contraction using magnetic twisting cytometry (MTC). These findings suggest miRNA-139-5p is capable of significantly influencing the phenotypic tonicity in smooth muscle via ROCK2: a lack of tone in ASM may be associated with the suppression of ROCK2 by high expression of miRNA-139-5p, whereas basal IAS tone may be associated with the persistence of ROCK2 due to low expression of miRNA-139-5p.


Subject(s)
Anal Canal/metabolism , MicroRNAs/genetics , Muscle Hypotonia/genetics , Myocytes, Smooth Muscle/metabolism , rho-Associated Kinases/genetics , Anal Canal/cytology , Animals , Antagomirs/genetics , Antagomirs/metabolism , Cytophotometry , Gene Expression Regulation , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Muscle Contraction/physiology , Muscle Hypotonia/metabolism , Muscle Hypotonia/physiopathology , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/cytology , Oligoribonucleotides/genetics , Oligoribonucleotides/metabolism , Phenotype , Rats , Rats, Sprague-Dawley , Signal Transduction , Single-Cell Analysis , rho-Associated Kinases/metabolism
15.
Am J Physiol Gastrointest Liver Physiol ; 311(5): G964-G973, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27634012

ABSTRACT

A comprehensive genomic and proteomic, computational, and physiological approach was employed to examine the (previously unexplored) role of microRNAs (miRNAs) as regulators of internal anal sphincter (IAS) smooth muscle contractile phenotype and basal tone. miRNA profiling, genome-wide expression, validation, and network analyses were employed to assess changes in mRNA and miRNA expression in IAS smooth muscles from young vs. aging rats. Multiple miRNAs, including rno-miR-1, rno-miR-340-5p, rno-miR-185, rno-miR-199a-3p, rno-miR-200c, rno-miR-200b, rno-miR-31, rno-miR-133a, and rno-miR-206, were found to be upregulated in aging IAS. qPCR confirmed the upregulated expression of these miRNAs and downregulation of multiple, predicted targets (Eln, Col3a1, Col1a1, Zeb2, Myocd, Srf, Smad1, Smad2, Rhoa/Rock2, Fn1, Tagln v2, Klf4, and Acta2) involved in regulation of smooth muscle contractility. Subsequent studies demonstrated an aging-associated increase in the expression of miR-133a, corresponding decreases in RhoA, ROCK2, MYOCD, SRF, and SM22α protein expression, RhoA-signaling, and a decrease in basal and agonist [U-46619 (thromboxane A2 analog)]-induced increase in the IAS tone. Moreover, in vitro transfection of miR-133a caused a dose-dependent increase of IAS tone in strips, which was reversed by anti-miR-133a. Last, in vivo perianal injection of anti-miR-133a reversed the loss of IAS tone associated with age. This work establishes the important regulatory effect of miRNA-133a on basal and agonist-stimulated IAS tone. Moreover, reversal of age-associated loss of tone via anti-miR delivery strongly implicates miR dysregulation as a causal factor in the aging-associated decrease in IAS tone and suggests that miR-133a is a feasible therapeutic target in aging-associated rectoanal incontinence.


Subject(s)
Aging/metabolism , Anal Canal/metabolism , Gene Expression Regulation , MicroRNAs/metabolism , Muscle, Smooth/metabolism , Aging/genetics , Animals , Gene Expression Profiling , Kruppel-Like Factor 4 , MicroRNAs/genetics , Muscle Contraction/physiology , Rats , Rats, Inbred F344 , Signal Transduction/physiology
16.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G1052-60, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27173508

ABSTRACT

Gastrointestinal dysmotility in systemic sclerosis (SSc) is associated with autoantibodies against muscarinic-3 receptor (M3-R). We investigated the temporal course of the site of action of these autoantibodies at the myenteric neurons (MN) vs. the smooth muscle (SM) M3-R in relation to disease duration, and determined the role of intravenous immunoglobulin (IVIG) in reversing these changes. Immunoglobulins purified from SSc patients (SScIgG) were used to assess their differential binding to MN and SM (from rat colon) employing immunohistochemistry (IHC). Effect of SScIgG on neural and direct muscle contraction was determined by cholinergic nerve stimulation and bethanechol-induced SM contraction. Effects of IVIG and its antigen-binding fragment F(ab')2 on SScIgG binding were studied by enzyme-linked immunosorbent assay (ELISA) of rat colonic longitudinal SM myenteric plexus (LSMMP) lysate and to second extracellular loop peptide of M3-R (M3-RL2). SScIgG from all patients demonstrated significantly higher binding to MN than to SM. With progression of SSc duration, binding at MN and SM increased in a linear fashion with a correlation coefficient of 0.696 and 0.726, respectively (P < 0.05). SScIgG-mediated attenuation of neural and direct SM contraction also increased with disease duration. ELISA analysis revealed that IVIG and F(ab')2 significantly reduced SScIgG binding to LSMMP lysate and M3-RL2. Dysmotility in SSc occurs sequentially, beginning with SScIgG-induced blockage of cholinergic neurotransmission (neuropathy), which progresses to inhibition of acetylcholine action at the SM cell (myopathy). IVIG reverses this cholinergic dysfunction at the neural and myogenic receptors by anti-idiotypic neutralization of SScIgG.


Subject(s)
Immunoglobulins, Intravenous/therapeutic use , Receptor, Muscarinic M3/immunology , Scleroderma, Systemic/immunology , Adult , Aged , Animals , Female , Humans , Immunoglobulins, Intravenous/immunology , Male , Middle Aged , Muscle Contraction , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , Myenteric Plexus/cytology , Neurons/metabolism , Neurons/physiology , Protein Binding , Rats , Rats, Sprague-Dawley , Scleroderma, Systemic/therapy
17.
Indian J Gastroenterol ; 35(2): 133-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27053352

ABSTRACT

Hypogammaglobulinemia/common variable immunodeficiency (CVID) may lead to disruption of the gut mucosal immune barrier. Collagenous infiltrative disorders of the intestinal tract (colitis, gastritis, sprue) constitute a relatively new spectrum of gastrointestinal disorders. Our aims were (1) to determine the association between immunoglobulin deficiency state like CVID and collagenous infiltrative disorders of the gut and (2) to study the clinic-pathologic characteristics and treatment outcomes in these patients. A retrospective search was conducted to identify cases with concurrence of these two conditions at an academic center from 2007 to 2013. Four such patients were identified from our database: three with collagenous colitis and one with collagenous gastritis. All patients with collagenous colitis had normal colonic mucosa while the patient with collagenous gastritis had nodular gastric mucosa. Only one patient out of four had decreased plasma cells in the submucosa as expected in low immunoglobulin states. All patients had improvement in their symptoms on immunoglobulin therapy with considerable remission on budesonide. Literature search revealed reporting of four similar patients. In conclusion, (1) the association between collagenous infiltrative disorders of the gut and CVID and its prompt response to immunoglobulins with effective maintenance with budesonide are novel findings. Our study also shows that the presence of plasma cells should not rule out the possibility of CVID. (2) In patients with chronic diarrhea, hypogammaglobulinemia and collagenous colitis/sprue should be considered for the available effective treatments such as immunoglobulins and budesonide.


Subject(s)
Colitis, Collagenous/etiology , Common Variable Immunodeficiency/complications , Adult , Aged , Budesonide/administration & dosage , Budesonide/therapeutic use , Colitis, Collagenous/drug therapy , Common Variable Immunodeficiency/immunology , Female , Humans , Immunoglobulin G/blood , Immunoglobulins, Intravenous/administration & dosage , Immunoglobulins, Intravenous/therapeutic use , Male , Middle Aged , Remission Induction , Retrospective Studies , Treatment Outcome , Young Adult
18.
Am J Physiol Gastrointest Liver Physiol ; 310(7): G449-59, 2016 04 01.
Article in English | MEDLINE | ID: mdl-26822916

ABSTRACT

MicroRNAs (miRNAs) belong to a group of short noncoding RNA molecules with important roles in cellular biology. miRNAs regulate gene expression by repressing translation or degrading the target mRNA. Recently, a growing body of evidence suggests that miRNAs are implicated in many diseases and could be potential biomarkers. Fibrosis and/smooth muscle (SM) dysfunction contributes to the morbidity and mortality associated with several diseases of the gastrointestinal tract (GIT). Currently available therapeutic modalities are unsuccessful in efficiently blocking or reversing fibrosis and/or SM dysfunction. Recent understanding of the role of miRNAs in signaling pathway of fibrogenesis and SM phenotype switch has provided a new insight into translational research. However, much is still unknown about the molecular targets and therapeutic potential of miRNAs in the GIT. This review discusses miRNA biology, pathophysiology of fibrosis, and aging- associated SM dysfunction in relation to the deregulation of miRNAs in the GIT. We also highlight the role of selected miRNAs associated with fibrosis and SM dysfunction-related diseases of the GIT.


Subject(s)
Gastrointestinal Diseases/metabolism , Gastrointestinal Tract/metabolism , MicroRNAs/metabolism , Muscle, Smooth/metabolism , Age Factors , Animals , Epigenesis, Genetic , Fibrosis , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/physiopathology , Gastrointestinal Diseases/therapy , Gastrointestinal Tract/pathology , Gastrointestinal Tract/physiopathology , Gene Expression Regulation , Genetic Markers , Genetic Therapy/methods , Humans , MicroRNAs/genetics , MicroRNAs/therapeutic use , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , Oxidative Stress , Phenotype , Signal Transduction
19.
Ann Gastroenterol ; 28(4): 469-74, 2015.
Article in English | MEDLINE | ID: mdl-26423466

ABSTRACT

BACKGROUND: Despite the surge of new medical and surgical approaches to treat fecal incontinence, the types of sphincter abnormalities in patients with incontinence have not been well characterized. We aimed to categorize anal sphincter dysfunction using anorectal manometry in patients with fecal incontinence as a potential guide for improved treatment. METHODS: A retrospective review of 162 consecutive patients with fecal incontinence referred for anorectal manometry was performed. Resting anal pressure and maximal squeeze pressure were considered as measures of internal anal sphincter and external anal sphincter function respectively. RESULTS: Mean age of the patients was 63 years (13-89); females (81.5%) and males (18.5%). 74% of the patients had sphincter dysfunction on anorectal manometry. Internal anal sphincter dysfunction was present in 62% patients vs. external anal sphincter dysfunction present in 44% patients. 80% females had abnormal manometry vs. 44% in males (P<0.0001). Internal anal sphincter dysfunction was present in 68% females vs. 37% in males (P=0.0026). CONCLUSIONS: Overall, abnormal anorectal manometry studies revealed that internal anal sphincter dysfunction is the most common finding, alone or in combination with external anal sphincter dysfunction. We suggest that anorectal manometry may be important to delineate anal sphincter function prior to using newer therapeutic mechanical devices. Future studies using pharmacological agents to increase internal anal sphincter tone may be of clinical importance. Finally, the classification of fecal incontinence based on the type of sphincter dysfunction may be an improved guide in the selection of newer agents in treating fecal incontinence.

20.
Am J Physiol Gastrointest Liver Physiol ; 309(5): G292-300, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26138467

ABSTRACT

Changes in oxidative stress may affect basal tone and relaxation of the internal anal sphincter (IAS) smooth muscle in aging. We examined this issue by investigating the effects of the oxidative stress inducer 6-anilino-5,8-quinolinedione (LY-83583) in basal as well as U-46619-stimulated tone, and nonadrenergic, noncholinergic (NANC) relaxation in rat IAS. LY-83583, which works via generation of reactive oxygen species in living cells, produced a bimodal effect in IAS tone: lower concentrations (0.1 nM to 10 µM) produced a concentration-dependent increase, while higher concentrations (50-100 µM) produced a decrease in IAS tone. An increase in IAS tone by lower concentrations was associated with an increase in RhoA/Rho kinase (ROCK) activity. This was evident by the increase in RhoA/ROCK in the particulate fractions, in ROCK activity, and in the levels of phosphorylated (p) (Thr696)-myosin phosphatase target subunit 1 and p(Thr18/Ser19)-20-kDa myosin light chain. Conversely, higher concentrations of LY-83583 produced inhibitory effects on RhoA/ROCK. Interestingly, both the excitatory and inhibitory effects of LY-83583 in the IAS were reversed by superoxide dismutase. The excitatory effects of LY-83583 were found to resemble those with neuronal nitric oxide synthase (nNOS) inhibition by l-NNA, since it produced a significant increase in the IAS tone and attenuated NANC relaxation. These effects of LY-83583 and l-NNA were reversible by l-arginine. This suggests the role of nNOS inhibition and RhoA/ROCK activation in the increase in IAS tone by LY-83583. These data have important implications in the pathophysiology and therapeutic targeting of rectoanal disorders, especially associated with IAS dysfunction.


Subject(s)
Anal Canal/metabolism , Oxidative Stress , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Aminoquinolines/pharmacology , Anal Canal/drug effects , Anal Canal/physiology , Animals , Guanylate Cyclase/antagonists & inhibitors , Myosin-Light-Chain Phosphatase/metabolism , Myosins/metabolism , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Vasoconstrictor Agents/pharmacology , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL