1
|
Li RJ, Xu JY, Wang X, Liao LJ, Wei X, Xie P, Xu WY, Xu ZY, Xie SH, Jiang YY, Huang L, Wang LY, Huang GR, Huang YQ. Therapeutic effect of demethylated hydroxylated phillygenin derivative on Helicobacter pylori infection. Front Microbiol 2023; 14:1071603. [PMID: 37275170 PMCID: PMC10235509 DOI: 10.3389/fmicb.2023.1071603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 05/04/2023] [Indexed: 06/07/2023] Open
Abstract
Modifying and transforming natural antibacterial products is a novel idea for developing new efficacious compounds. Phillygenin has an inhibitory effect on H. pylori. The aim of the present study was to prepare a phillygenin derivative (PHI-Der) through demethylation and hydroxylation. The minimum inhibitory concentration of 18 strains of H. pylori from different sources was 8-32 μg/mL in vitro, and the activity increased 2-8 times than that of phillygenin. PHI-Der could significantly inhibit the colonization of H. pylori in vivo, reduce the inflammatory response, and promote the repair of inflammatory damage. Further, we used SwissTargetPrediction to predict that its main targets are ALOX5, MCL1, and SLC6A4, and find that it can inhibit bacterial biofilm formation and reduce bacterial infection of cells. It can enhance the intracellular oxidative capacity of H. pylori to inhibit H. pylori growth. Further, it could prevent the oxidation of H. pylori-infected cells and reduce the inflammatory response, which plays a role in protection. In conclusion, compared to phillygenin, PHI-Der had better antibacterial activity and was more effective in treating H. pylori infection. It has characteristics of high safety, specificity, resistance to drug resistance and better antibacterial activity than phillygenin, it's a good antioxidant for host cells.
Collapse
Affiliation(s)
- Ru-Jia Li
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
- Clinical Laboratory of 980 Hospital of PLA Joint Logistics Support Force (Bethune International Peace Hospital), Shijiazhuang, Hebei, China
| | - Jia-yin Xu
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
| | - Xue Wang
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
| | - Li-juan Liao
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
| | - Xian Wei
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
| | - Ping Xie
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
| | - Wen-yan Xu
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
| | - Zhen-yi Xu
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
| | - Shuo-hua Xie
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
| | - Yu-ying Jiang
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
| | - Liang Huang
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
| | - Lu-yao Wang
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
| | - Gan-rong Huang
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
| | - Yan-Qiang Huang
- Key Laboratory of the Prevention and Treatment of Drug Resistant Microbial Infecting (Youjiang Medical University for Nationalities), Education Department of Guangxi Zhuang Autonomous Region, Baise, China
| |
Collapse
|
2
|
Singh R, Zogg H, Ro S. Role of microRNAs in Disorders of Gut-Brain Interactions: Clinical Insights and Therapeutic Alternatives. J Pers Med 2021; 11:1021. [PMID: 34683162 PMCID: PMC8541612 DOI: 10.3390/jpm11101021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Disorders of gut-brain interactions (DGBIs) are heterogeneous in nature and intertwine with diverse pathophysiological mechanisms. Regular functioning of the gut requires complex coordinated interplay between a variety of gastrointestinal (GI) cell types and their functions are regulated by multiple mechanisms at the transcriptional, post-transcriptional, translational, and post-translational levels. MicroRNAs (miRNAs) are small non-coding RNA molecules that post-transcriptionally regulate gene expression by binding to specific mRNA targets to repress their translation and/or promote the target mRNA degradation. Dysregulation of miRNAs might impair gut physiological functions leading to DGBIs and gut motility disorders. Studies have shown miRNAs regulate gut functions such as visceral sensation, gut immune response, GI barrier function, enteric neuronal development, and GI motility. These biological processes are highly relevant to the gut where neuroimmune interactions are key contributors in controlling gut homeostasis and functional defects lead to DGBIs. Although extensive research has explored the pathophysiology of DGBIs, further research is warranted to bolster the molecular mechanisms behind these disorders. The therapeutic targeting of miRNAs represents an attractive approach for the treatment of DGBIs because they offer new insights into disease mechanisms and have great potential to be used in the clinic as diagnostic markers and therapeutic targets. Here, we review recent advances regarding the regulation of miRNAs in GI pacemaking cells, immune cells, and enteric neurons modulating pathophysiological mechanisms of DGBIs. This review aims to assess the impacts of miRNAs on the pathophysiological mechanisms of DGBIs, including GI dysmotility, impaired intestinal barrier function, gut immune dysfunction, and visceral hypersensitivity. We also summarize the therapeutic alternatives for gut microbial dysbiosis in DGBIs, highlighting the clinical insights and areas for further exploration. We further discuss the challenges in miRNA therapeutics and promising emerging approaches.
Collapse
Affiliation(s)
| | | | - Seungil Ro
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, 1664 North Virginia Street, Reno, NV 89557, USA; (R.S.); (H.Z.)
| |
Collapse
|
3
|
Singh R, Wei L, Ghoshal UC. Micro-organic basis of functional gastrointestinal (GI) disorders: Role of microRNAs in GI pacemaking cells. Indian J Gastroenterol 2021; 40:102-110. [PMID: 33738768 DOI: 10.1007/s12664-021-01159-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Rajan Singh
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, 89557, USA
| | - Lai Wei
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, 89557, USA
| | - Uday C Ghoshal
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226 014, India.
| |
Collapse
|
4
|
Beversdorf DQ, Shah A, Jhin A, Noel-MacDonnell J, Hecht P, Ferguson BJ, Bruce D, Tilley M, Talebizadeh Z. microRNAs and Gene-Environment Interactions in Autism: Effects of Prenatal Maternal Stress and the SERT Gene on Maternal microRNA Expression. Front Psychiatry 2021; 12:668577. [PMID: 34290629 PMCID: PMC8288023 DOI: 10.3389/fpsyt.2021.668577] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
Background: Genetics and environment both are critical in autism spectrum disorder (ASD), but their interaction (G × E) is less understood. Numerous studies have shown higher incidence of stress exposures during pregnancies with children later diagnosed with ASD. However, many stress-exposed mothers have unaffected children. The serotonin transporter (SERT) gene affects stress reactivity. Two independent samples have shown that the association between maternal stress exposure and ASD is greatest with maternal presence of the SERT short (S)-allele (deletion in the promoter region). MicroRNAs play a regulatory role in the serotonergic pathway and in prenatal stress and are therefore potential mechanistic targets in this setting. Design/methods: We profiled microRNA expression in blood from mothers of children with ASD, with known stress exposure during pregnancy. Samples were divided into groups based on SERT genotypes (LL/LS/SS) and prenatal stress level (high/low). Results: Two thousand five hundred mature microRNAs were examined. The ANOVA analysis showed differential expression (DE) of 119 microRNAs; 90 were DE in high- vs. low-stress groups (stress-dependent). Two (miR-1224-5p, miR-331-3p) were recently reported by our group to exhibit stress-dependent expression in rodent brain samples from embryos exposed to prenatal stress. Another, miR-145-5p, is associated with maternal stress. Across SERT genotypes, with high stress exposure, 20 significantly DE microRNAs were detected, five were stress-dependent. These microRNAs may be candidates for stress × SERT genotype interactions. This is remarkable as these changes were from mothers several years after stress-exposed pregnancies. Conclusions: Our study provides evidence for epigenetic alterations in relation to a G × E model (prenatal maternal stress × SERT gene) in ASD.
Collapse
Affiliation(s)
- David Q Beversdorf
- Departments of Radiology, Neurology, and Psychological Sciences, William and Nancy Thompson Endowed Chair in Radiology, University of Missouri, Columbia, MO, United States.,Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, United States
| | - Ayten Shah
- Children's Mercy Hospital, Kansas City, MO, United States
| | - Allison Jhin
- Kansas City University, Kansas City, MO, United States
| | - Janelle Noel-MacDonnell
- Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
| | - Patrick Hecht
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, United States
| | - Bradley J Ferguson
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, United States.,Health Psychology, Radiology, and Thompson Center for Autism and Neurodevelopmental Disorders, University of Missouri, Columbia, MO, United States
| | - Danielle Bruce
- Department of Biology, Central Methodist University, Fayette, MO, United States
| | - Michael Tilley
- Department of Biology, Central Methodist University, Fayette, MO, United States
| | - Zohreh Talebizadeh
- Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
| |
Collapse
|
5
|
Tanaka F, Takashima S, Nadatani Y, Otani K, Hosomi S, Kamata N, Taira K, Nagami Y, Tanigawa T, Fukumoto S, Watanabe T, Murakami Y, Kawada N, Fujiwara Y. Exosomal hsa-miR-933 in Gastric Juice as a Potential Biomarker for Functional Dyspepsia. Dig Dis Sci 2020; 65:3493-3501. [PMID: 31974910 DOI: 10.1007/s10620-020-06096-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/20/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND MicroRNAs (miRNAs) in exosomes represent disease-specific profiles and are applied as biomarkers in oncology. However, in functional dyspepsia (FD), the role of exosomal miRNAs has not been fully elucidated. AIMS To investigate exosomal miRNAs as potential biomarkers of FD using liquid biopsy. METHODS This retrospective cohort study included 11 subjects with FD and 11 age- and sex-matched healthy controls (HCs). We collected gastric juice and isolated exosomal miRNAs. In a discovery cohort, expression levels of 2565 miRNAs were evaluated by 3D-Gene® microarray. miRNA expression profiles from exosomes of subjects with FD and HCs were compared by two normalization methods: (1) global normalization and (2) normalization by internal control. Subsequently, in a validation cohort, the expression levels of miRNAs were validated by quantitative reverse transcription PCR (RT-qPCR). RESULTS Through microarray analysis using the two methods, we identified 39 miRNAs that were consistently and significantly downregulated in FD cases compared with those in HCs. Of these, 12 miRNAs (hsa-miR-933, hsa-miR-345-5p, hsa-miR-708-5p, hsa-miR-203a-3p, hsa-miR-619-5p, hsa-miR-4294, hsa-miR-4481, hsa-miR-196a-5p, hsa-miR-3918, hsa-miR-372-3p, hsa-miR-658, and hsa-miR-3654) were further validated by RT-qPCR. Our results indicated that hsa-miR-933 was significantly downregulated in FD compared with HCs (0.317 ± 0.205-fold, P = 0.0317). Furthermore, the expression level of hsa-miR-933 was negatively associated with dyspepsia score and the frequency of epigastric pain and/or burning (P < 0.01, r = - 0.835; P = 0.0280, r = - 0.688, respectively). CONCLUSIONS Exosomal hsa-miR-933 in gastric juice could be a candidate biomarker for FD.
Collapse
Affiliation(s)
- Fumio Tanaka
- Department of Gastroenterology, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan.
- Department of Premier Preventive Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan.
| | - Shingo Takashima
- Department of Gastroenterology, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yuji Nadatani
- Department of Gastroenterology, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
- Department of Premier Preventive Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Koji Otani
- Department of Gastroenterology, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
- Department of Premier Preventive Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Shuhei Hosomi
- Department of Gastroenterology, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Noriko Kamata
- Department of Gastroenterology, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Koichi Taira
- Department of Gastroenterology, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yasuaki Nagami
- Department of Gastroenterology, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Tetsuya Tanigawa
- Department of Gastroenterology, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Shinya Fukumoto
- Department of Premier Preventive Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Toshio Watanabe
- Department of Gastroenterology, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yoshiki Murakami
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Norifumi Kawada
- Department of Premier Preventive Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan
- Department of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Yasuhiro Fujiwara
- Department of Gastroenterology, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| |
Collapse
|
6
|
Martínez C, Lasitschka F, Thöni C, Wohlfarth C, Braun A, Granzow M, Röth R, Dizdar V, Rappold GA, Hausken T, Langeland N, Hanevik K, Niesler B. Comparative expression profiling in the intestine of patients with Giardia-induced postinfectious functional gastrointestinal disorders. Neurogastroenterol Motil 2020; 32:e13868. [PMID: 32391639 DOI: 10.1111/nmo.13868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/13/2020] [Accepted: 04/07/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND A Giardia outbreak in Bergen, Norway, caused postinfectious functional gastrointestinal disorders (PI-FGIDs). Despite the devastating effects of this outbreak, it presented a unique chance to investigate the implication on the dysregulation of genetic pathways in PI-FGID. METHODS We performed the first comparative expression profiling of miRNAs and their potential target genes in microdissected rectal biopsies from 20 Giardia-induced PI-FGID patients vs 18 healthy controls by nCounter analysis. Subsequently, candidates were validated on protein level by immunostaining. KEY RESULTS miRNA profiling on rectal biopsy samples from 5 diarrhea-predominant PI-IBS cases compared to 10 healthy controls revealed differential expression in the epithelial layer. The top five regulated miRNAs were implicated in GI disease, inflammatory response, and immunological disease. Subsequently, these miRNAs and 100 potential mRNA targets were examined in 20 PI-FGID cases and 18 healthy controls in both the mucosal epithelium and the lamina propria. Although deregulation of the selected miRNAs could not be verified in the larger sample set, mRNAs involved in barrier function were downregulated in the epithelium. Pro-inflammatory genes and genes implicated in epigenetic modifications were upregulated in the lamina propria. Immunostaining for selected candidates on 17 PI-FGID cases and 16 healthy controls revealed increased tryptase levels as well as a decreased and aberrant subcellular expression of occludin. CONCLUSIONS AND INFERENCES Genes relevant to immune and barrier function as well as stress response and epigenetic modulation are differentially expressed in PI-FGIDs and may contribute to disease manifestation.
Collapse
Affiliation(s)
- Cristina Martínez
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany.,Lleida Institute for Biomedical Research Dr. Pifarré Foundation (IRBLleida), Lleida, Spain.,Genes in Irritable Bowel Syndrome (GENIEUR) Research Network Europe, Heidelberg, Germany
| | - Felix Lasitschka
- Institute of Pathology, Heidelberg University, Heidelberg, Germany
| | - Cornelia Thöni
- Institute of Pathology, Heidelberg University, Heidelberg, Germany
| | - Carolin Wohlfarth
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Alexander Braun
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Martin Granzow
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Ralph Röth
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany.,nCounter Core Facility Heidelberg, Institute of Human Genetics, Heidelberg, Germany
| | - Vernesa Dizdar
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gudrun A Rappold
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany.,nCounter Core Facility Heidelberg, Institute of Human Genetics, Heidelberg, Germany
| | - Trygve Hausken
- Genes in Irritable Bowel Syndrome (GENIEUR) Research Network Europe, Heidelberg, Germany.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Nina Langeland
- Genes in Irritable Bowel Syndrome (GENIEUR) Research Network Europe, Heidelberg, Germany.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kurt Hanevik
- Genes in Irritable Bowel Syndrome (GENIEUR) Research Network Europe, Heidelberg, Germany.,Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Medicine, National Advisory Center for Tropical Infectious Diseases, Haukeland University Hospital, Bergen, Norway
| | - Beate Niesler
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany.,Genes in Irritable Bowel Syndrome (GENIEUR) Research Network Europe, Heidelberg, Germany.,nCounter Core Facility Heidelberg, Institute of Human Genetics, Heidelberg, Germany
| |
Collapse
|
7
|
Chen X, Gao J, Yu Y, Zhao Z, Pan Y. LncRNA FOXD3-AS1 promotes proliferation, invasion and migration of cutaneous malignant melanoma via regulating miR-325/MAP3K2. Biomed Pharmacother 2019; 120:109438. [PMID: 31541886 DOI: 10.1016/j.biopha.2019.109438] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/06/2019] [Accepted: 09/06/2019] [Indexed: 12/22/2022] Open
Abstract
PURPOSE The aim was to study the mechanism of LncRNA FOXD3-AS1 in cutaneous melanoma. METHODS FOXD3-AS1 levels in 47 pairs of melanoma samples were detected. We used qRT-PCR to detect FOXD3-AS1, miR-325 and MAP3K2 expression in different staging samples and cutaneous melanoma cell lines. We used Kaplan-Meier curve to analyze survival rate in patients with FOXD3-AS1 high and low expression. Sh-FOXD3-AS1, miR-325, miR-325 inhibitor and oeMAP3K2 were transfected. The proliferation of A375 and SK-MEL-1 was detected by CCK8 and EdU labeling assay and cell clone formation assay. Dual luciferase reporter assay and pull down assay was used to confirm the binding site of FOXD3-AS1, miR-325 and MAP3K2. Flow cytometry was applied to detect the effect of lncRNA on cell cycle. The migration and invasion ability were detected by transwell assay. RESULTS LncRNA FOXD3-AS1 highly expressed in cutaneous melanoma cells and tissues. Patients with highly expressed LncRNA FOXD3-AS1 were always with shorter overall survival time. When LncRNA FOXD3-AS1 was knockdown, proliferation, invasion and migration of cutaneous malignant melanoma, and tumor weight was inhibited, and cell cycle was arrested. LncRNA FOXD3-AS1 negatively regulated the expression of miR-325, and then improved the level of MAP3K2. MiR-325 was with similarly effects on above biological process, and MAP3K2 overexpression could rescue the influence of sh-FOXD3-AS1. Tumor volume and weight were measured to confirm the effect of sh-FOXD3-AS1 in vivo. CONCLUSION LncRNA FOXD3-AS1 could promote proliferation, invasion and migration of cutaneous malignant melanoma via regulating miR-325/MAP3K2 axis.
Collapse
Affiliation(s)
- Xige Chen
- Department of Dermatology, Weihai Central Hospitai, Weihai 264400, China
| | - Juan Gao
- Department of Rheumatology, Weihai Central Hospitai, Weihai 264400, China
| | - Yanhua Yu
- Department of Dermatology, Weihai Central Hospitai, Weihai 264400, China
| | - Zhengjuan Zhao
- Department of Dermatology, Weihai Central Hospitai, Weihai 264400, China
| | - Yingli Pan
- Department of Dermatology, Weihai Central Hospitai, Weihai 264400, China.
| |
Collapse
|
8
|
Wu R, Zhang PA, Liu X, Zhou Y, Xu M, Jiang X, Yan J, Xu GY. Decreased miR-325-5p Contributes to Visceral Hypersensitivity Through Post-transcriptional Upregulation of CCL2 in Rat Dorsal Root Ganglia. Neurosci Bull 2019; 35:791-801. [PMID: 30980241 DOI: 10.1007/s12264-019-00372-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 01/16/2019] [Indexed: 12/13/2022] Open
Abstract
Chronic visceral hypersensitivity is an important type of chronic pain with unknown etiology and pathophysiology. Recent studies have shown that epigenetic regulation plays an important role in the development of chronic pain conditions. However, the role of miRNA-325-5p in chronic visceral pain remains unknown. The present study was designed to determine the roles and mechanism of miRNA-325-5p in a rat model of chronic visceral pain. This model was induced by neonatal colonic inflammation (NCI). In adulthood, NCI led to a significant reduction in the expression of miRNA-325-5p in colon-related dorsal root ganglia (DRGs), starting to decrease at the age of 4 weeks and being maintained to 8 weeks. Intrathecal administration of miRNA-325-5p agomir significantly enhanced the colorectal distention (CRD) threshold in a time-dependent manner. NCI also markedly increased the expression of CCL2 (C-C motif chemokine ligand 2) in colon-related DRGs at the mRNA and protein levels relative to age-matched control rats. The expression of CXCL12, IL33, SFRS7, and LGI1 was not significantly altered in NCI rats. CCL2 was co-expressed in NeuN-positive DRG neurons but not in glutamine synthetase-positive glial cells. Furthermore, CCL2 was mainly expressed in isolectin B4-binding- and calcitonin gene-related peptide-positive DRG neurons but in few NF-200-positive cells. More importantly, CCL2 was expressed in miR-325-5p-positive DRG neurons. Intrathecal injection of miRNA-325-5p agomir remarkably reduced the upregulation of CCL2 in NCI rats. Administration of Bindarit, an inhibitor of CCL2, markedly raised the CRD threshold in NCI rats in a dose- and time-dependent manner. These data suggest that NCI suppresses miRNA-325-5p expression and enhances CCL2 expression, thus contributing to visceral hypersensitivity in adult rats.
Collapse
Affiliation(s)
- Rui Wu
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, 215600, China
- Department of Physiology and Neurobiology, Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Ping-An Zhang
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, 215600, China
- Department of Physiology and Neurobiology, Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xuelian Liu
- Department of Physiology and Neurobiology, Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Yuan Zhou
- Department of Physiology and Neurobiology, Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Meijie Xu
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, 215600, China
| | - Xinghong Jiang
- Department of Physiology and Neurobiology, Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Jun Yan
- The Second Affiliated of Hospital Soochow University, Suzhou, 215004, China
| | - Guang-Yin Xu
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, 215600, China.
- Department of Physiology and Neurobiology, Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
9
|
Beversdorf DQ, Stevens HE, Margolis KG, Van de Water J. Prenatal Stress and Maternal Immune Dysregulation in Autism Spectrum Disorders: Potential Points for Intervention. Curr Pharm Des 2019; 25:4331-4343. [PMID: 31742491 PMCID: PMC7100710 DOI: 10.2174/1381612825666191119093335] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 11/15/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Genetics is a major etiological contributor to autism spectrum disorder (ASD). Environmental factors, however, also appear to contribute. ASD pathophysiology due to gene x environment is also beginning to be explored. One reason to focus on environmental factors is that they may allow opportunities for intervention or prevention. METHODS AND RESULTS Herein, we review two such factors that have been associated with a significant proportion of ASD risk, prenatal stress exposure and maternal immune dysregulation. Maternal stress susceptibility appears to interact with prenatal stress exposure to affect offspring neurodevelopment. We also explore how maternal stress may interact with the microbiome in the neurodevelopmental setting. Additionally, understanding of the impact of maternal immune dysfunction on ASD has recently been advanced by recognition of specific fetal brain proteins targeted by maternal autoantibodies, and identification of unique mid-gestational maternal immune profiles. This might also be interrelated with maternal stress exposure. Animal models have been developed to explore pathophysiology targeting each of these factors. CONCLUSION We are beginning to understand the behavioral, pharmacopathological, and epigenetic effects related to these interactions, and we are beginning to explore potential mitigating factors. Continued growth in understanding of these mechanisms may ultimately allow for the identification of multiple potential targets for prevention or intervention for this subset of environmental-associated ASD cases.
Collapse
Affiliation(s)
- David Q. Beversdorf
- Departments of Radiology, Neurology, and Psychological Sciences, and The Thompson Center for Neurodevelopmental Disorders, University of Missouri, William and Nancy Thompson Endowed Chair in Radiology
| | - Hanna E. Stevens
- Departments of Psychiatry and Pediatrics, Iowa Neuroscience Institute, University of Iowa
| | - Kara Gross Margolis
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Morgan Stanley Children’s Hospital, Columbia University Medical Center
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, And the MIND Institute, University of California, Davis
| |
Collapse
|
10
|
Suzuki H. New Medical Approach to Functional Dyspepsia, from Core Symposium 3, Japan Gastroenterological Association 2015-2017. Digestion 2018; 97:6-12. [PMID: 29393229 DOI: 10.1159/000484029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In the annual meeting of the Japan Gastroenterological Association (JGA), the scientific organizing committee selected the serial topics for the core symposium. One of the core symposia held during 2015-2017 was entitled "New medical approach to functional dyspepsia (FD)." In 2015, the subtitle of this symposium was "Helicobacter pylori gastritis and FD." In 2016, the subtitle of this symposium was "overlap with other functional GI disorders." In 2017, the subtitle was "therapeutic approach to FD." During these 3 years, a total of 24 presentations were included in Core Symposium 3 and deep and intensive discussions were carried out.
Collapse
|
11
|
Beversdorf DQ, Stevens HE, Jones KL. Prenatal Stress, Maternal Immune Dysregulation, and Their Association With Autism Spectrum Disorders. Curr Psychiatry Rep 2018; 20:76. [PMID: 30094645 PMCID: PMC6369590 DOI: 10.1007/s11920-018-0945-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE OF REVIEW While genetic factors are a major etiological contributor to autism spectrum disorder (ASD), evidence also supports a role for environmental factors. Herein, we will discuss two such factors that have been associated with a significant proportion of ASD risk: prenatal stress exposure and maternal immune dysregulation, and how sex and gender relate to these factors. RECENT FINDINGS Recent evidence suggests that maternal stress susceptibility interacts with prenatal stress exposure to affect offspring neurodevelopment. Additionally, understanding of the impact of maternal immune dysfunction on ASD has recently been advanced by recognition of specific fetal brain proteins targeted by maternal autoantibodies, and identification of unique mid-gestational maternal immune profiles. Animal models have been developed to explore pathophysiology targeting both of these factors, with limited sex-specific effects observed. While prenatal stress and maternal immune dysregulation are associated with ASD, most cases of these prenatal exposures do not result in ASD, suggesting interaction with multiple other risks. We are beginning to understand the behavioral, pharmacopathological, and epigenetic effects related to these interactions, as well as potential mitigating factors. Sex differences of these risks have been understudied but are crucial for understanding the higher prevalence of ASD in boys. Continued growth in understanding of these mechanisms may ultimately allow for the identification of multiple potential points for prevention or intervention, and for a personalized medicine approach for this subset of environmental-associated ASD cases.
Collapse
Affiliation(s)
- David Q. Beversdorf
- Departments of Radiology, Neurology, and Psychological Sciences And The Thompson Center for Neurodevelopmental Disorders, University of Missouri, Columbia, MO, USA,Department of Radiology, University of Missouri, DC 069.10, One Hospital Drive, Columbia, MO 65212, USA
| | - Hanna E. Stevens
- Departments of Psychiatry and Pediatrics, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Karen L. Jones
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, And the MIND Institute, University of California, Davis, Davis, CA, USA
| |
Collapse
|
12
|
Zhang Y, Li Y, Hao Z, Li X, Bo P, Gong W. Association of the Serotonin Receptor 3E Gene as a Functional Variant in the MicroRNA-510 Target Site with Diarrhea Predominant Irritable Bowel Syndrome in Chinese Women. J Neurogastroenterol Motil 2016; 22:272-81. [PMID: 26787495 PMCID: PMC4819866 DOI: 10.5056/jnm15138] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 12/22/2015] [Accepted: 12/31/2015] [Indexed: 12/12/2022] Open
Abstract
Background/Aims The functional variant (rs56109847) in the 3′-untranslated regions (3′-UTR) of the serotonin receptor 3E (HTR3E) gene is associated with female diarrhea predominant irritable bowel syndrome (IBS-D) in British populations. However, the relationship of the polymorphism both to HTR3E expression in the intestine and to the occurrence of Chinese functional gastrointestinal disorders has yet to be examined. Methods Polymerase chain reaction amplification and restriction fragment length polymorphism analyses were employed to detect polymorphisms among Chinese Han women, particularly 107 patients with IBS-D, 99 patients with functional dyspepsia (FD), 115 patients with mixed IBS and 69 patients with IBS-D + FD. We also assessed microRNA-510 (miR-510) and HTR3E expression in human colonic mucosal tissues with immunohistochemistry and other methods. Dual-luciferase reporter assays were conducted to examine the binding ability of miR-510 and HTR3E 3′-UTR. Results Genotyping data showed the variant rs56109847 was significantly associated with IBS-D, but not with FD, mixed-IBS, or FD + IBS-D. HTR3E was abundantly expressed around the colonic mucosal glands but less expressed in the stroma. miR-510 expression decreased, whereas HTR3E expression increased in the colonic mucosal tissue of patients with IBS-D compared with those in controls. HTR3E expression was significantly higher in patients with the GA genotype than that in patients with the GG genotype. The single-nucleotide polymorphisms disrupted the binding site of miR-510 and significantly upregulated luciferase expression in HEK293 and HT-29 cells. Conclusions The single-nucleotide polymorphisms rs56109847 led to reduced microRNA binding and overexpression of the target gene in intestinal cells, thereby increasing IBS-D risk in the Chinese Han population. The decreased expression of miR-510 might contribute to IBS-D.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Chinese and Western Integrative Medicine, Medical College of Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, China
| | - Yaoyao Li
- Department of Chinese and Western Integrative Medicine, Medical College of Yangzhou University, Yangzhou, China.,Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Zhenfeng Hao
- Department of Chinese and Western Integrative Medicine, Medical College of Yangzhou University, Yangzhou, China
| | - Xiangming Li
- Department of Chinese and Western Integrative Medicine, Medical College of Yangzhou University, Yangzhou, China
| | - Ping Bo
- Department of Chinese and Western Integrative Medicine, Medical College of Yangzhou University, Yangzhou, China
| | - Weijuan Gong
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, China.,Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
13
|
Martinucci I, Blandizzi C, de Bortoli N, Bellini M, Antonioli L, Tuccori M, Fornai M, Marchi S, Colucci R. Genetics and pharmacogenetics of aminergic transmitter pathways in functional gastrointestinal disorders. Pharmacogenomics 2016; 16:523-39. [PMID: 25916523 DOI: 10.2217/pgs.15.12] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Functional gastrointestinal disorders (FGIDs) are highly prevalent syndromes, without evident underlying organic causes. Their pathogenesis is multifactorial in nature, with a combination of environmental and genetic factors contributing to their clinical manifestations, for which most of current treatments are not satisfactory. It is acknowledged that amine mediators (noradrenaline, dopamine and serotonin) play pivotal regulatory actions on gut functions and visceral sensation. In addition, drugs of therapeutic interest for FGIDs act on these transmitter pathways. The present article reviews current knowledge on the impact of genetics and pharmacogenetics of aminergic pathways on FGID pathophysiology, clinical presentations, symptom severity and medical management, in an attempt of highlighting the most relevant evidence and point out issues that should be addressed in future investigations.
Collapse
Affiliation(s)
- Irene Martinucci
- Gastroenterology Unit, Department of Translational Research & New Technologies in Medicine, University of Pisa, Via Paradisa 2, I-56124 Pisa, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Kourikou A, Karamanolis GP, Dimitriadis GD, Triantafyllou K. Gene polymorphisms associated with functional dyspepsia. World J Gastroenterol 2015; 21:7672-7682. [PMID: 26167069 PMCID: PMC4491956 DOI: 10.3748/wjg.v21.i25.7672] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/07/2015] [Accepted: 05/21/2015] [Indexed: 02/06/2023] Open
Abstract
Functional dyspepsia (FD) is a constellation of functional upper abdominal complaints with poorly elucidated pathophysiology. However, there is increasing evidence that susceptibility to FD is influenced by hereditary factors. Genetic association studies in FD have examined genotypes related to gastrointestinal motility or sensation, as well as those related to inflammation or immune response. G-protein b3 subunit gene polymorphisms were first reported as being associated with FD. Thereafter, several gene polymorphisms including serotonin transporter promoter, interlukin-17F, migration inhibitory factor, cholecystocynine-1 intron 1, cyclooxygenase-1, catechol-o-methyltransferase, transient receptor potential vanilloid 1 receptor, regulated upon activation normal T cell expressed and secreted, p22PHOX, Toll like receptor 2, SCN10A, CD14 and adrenoreceptors have been investigated in relation to FD; however, the results are contradictory. Several limitations underscore the value of current studies. Among others, inconsistencies in the definitions of FD and controls, subject composition differences regarding FD subtypes, inadequate samples, geographical and ethnical differences, as well as unadjusted environmental factors. Further well-designed studies are necessary to determine how targeted genes polymorphisms, influence the clinical manifestations and potentially the therapeutic response in FD.
Collapse
|
15
|
Li F, Sun G, Yang YS, Cui LH, Peng LH, Guo X, Wang WF, Yan B, Zhang L. Offshore training in navy personnel is associated with uninvestigated dyspepsia. ACTA ACUST UNITED AC 2014; 34:808-814. [PMID: 25480574 DOI: 10.1007/s11596-014-1357-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Revised: 07/01/2014] [Indexed: 10/24/2022]
Abstract
To investigate the known and new factors associated with uninvestigated dyspepsia (UD), we surveyed 8600 Chinese navy personnel with offshore training shorter than 1 month or longer than 9 months per year. All respondents were required to complete a questionnaire covering demographics, the Chinese version of the Rome III survey, eating habits, life styles, and medical and family history. The response rate was 94.3% (8106/8600) with 4899 respondents qualified for analysis, including 1046 with offshore training and 3853 with onshore training. The prevalence of UD was higher in the offshore group than in the onshore group (12.6% vs. 6.9%, P<0.001), with a general prevalence of 8.1%. The subjects with offshore training were more likely to suffer from UD and postprandial distress syndrome (OR=1.955, 95% CI 1.568-2.439, P<0.001 and OR=1.789, 95% CI 1.403-2.303, P<0.001, respectively). The multivariate logistic regression analysis showed UD was associated with offshore training (OR=1.580, 95% CI 1.179-2.118, P=0.002), family history (OR=1.765, 95% CI 1.186-2.626, P=0.005) and smoking (OR=1.270, 95% CI 1.084-1.488, P=0.003), but not with alcohol drinking. The association between dysentery history and UD was undetermined/borderline (P=0.056-0.069). In conclusion, we identified offshore training as a new factor associated with UD, and also confirmed 2 known associated factors, family history and smoking.
Collapse
Affiliation(s)
- Fan Li
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing, 100853, China.,Department of Gastroenterology and Hepatology, Peking University International Hospital, Beijing, 102206, China
| | - Gang Sun
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yun-Sheng Yang
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Li-Hong Cui
- Department of Gastroenterology and Hepatology, The Navy General Hospital of The Chinese People's Liberation Army, Beijing, 100048, China.
| | - Li-Hua Peng
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xu Guo
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wei-Feng Wang
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Bin Yan
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lanjing Zhang
- Department of Pathology, University Medical Center of Princeton, Plainsboro, 08536, USA.,Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, 08903, USA.,Department of Chemical Biology, Ernest Mario School of Pharmacy, Piscataway, 08854, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, 08903, USA
| |
Collapse
|
16
|
Quigley EMM, Lacy BE. Overlap of functional dyspepsia and GERD--diagnostic and treatment implications. Nat Rev Gastroenterol Hepatol 2013; 10:175-86. [PMID: 23296247 DOI: 10.1038/nrgastro.2012.253] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
GERD and functional dyspepsia are the two most prevalent upper gastrointestinal disorders. Gastro-oesophageal reflux is most commonly diagnosed using the cardinal symptoms of heartburn and regurgitation. Patients might also be diagnosed using a questionnaire, after empiric treatment with an acid suppressant, after upper endoscopy or by pH testing. Functional dyspepsia is best diagnosed using symptoms outlined by the Rome committee in conjunction with a normal upper endoscopy. Theoretically, distinguishing these two populations should be easy for all health-care providers. In reality, however, carefully separating out these two populations can be quite difficult, as substantial overlap exists epidemiologically, symptomatically and even diagnostically. This overlap renders precise diagnosis a challenge; given the limited treatment options, the primary goal is to identify those patients who will respond to acid suppressive therapy. Despite the frequency with which functional dyspepsia and GERD overlap, remarkably few studies have investigated this overlap. Most recommendations are based on data derived from separate studies of functional dyspepsia and GERD. A further limitation of existing studies is their failure to differentiate between the various diagnostic categories into which the individual presenting with heartburn might belong.
Collapse
|
17
|
Hu S, Xiao Y, Zhu L, Li L, Hu CY, Jiang X, Xu GY. Neonatal maternal deprivation sensitizes voltage-gated sodium channel currents in colon-specific dorsal root ganglion neurons in rats. Am J Physiol Gastrointest Liver Physiol 2013; 304:G311-21. [PMID: 23139220 DOI: 10.1152/ajpgi.00338.2012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Irritable bowel syndrome (IBS) is a common gastrointestinal disorder characterized by abdominal pain in association with altered bowel movements. The underlying mechanisms of visceral hypersensitivity remain elusive. This study was designed to examine the role for sodium channels in a rat model of chronic visceral hyperalgesia induced by neonatal maternal deprivation (NMD). Abdominal withdrawal reflex (AWR) scores were performed on adult male rats. Colon-specific dorsal root ganglion (DRG) neurons were labeled with DiI and acutely dissociated for measuring excitability and sodium channel current under whole-cell patch-clamp configurations. The expression of Na(V)1.8 was analyzed by Western blot and quantitative real-time PCR. NMD significantly increased AWR scores, which lasted for ~6 wk in an association with hyperexcitability of colon DRG neurons. TTX-resistant but not TTX-sensitive sodium current density was greatly enhanced in colon DRG neurons in NMD rats. Compared with controls, activation curves showed a leftward shift in NMD rats whereas inactivation curves did not differ significantly. NMD markedly accelerated the activation time of peak current amplitude without any changes in inactivation time. Furthermore, NMD remarkably enhanced expression of Na(V)1.8 at protein levels but not at mRNA levels in colon-related DRGs. The expression of Na(V)1.9 was not altered after NMD. These data suggest that NMD enhances TTX-resistant sodium activity of colon DRG neurons, which is most likely mediated by a leftward shift of activation curve and by enhanced expression of Na(V)1.8 at protein levels, thus identifying a specific molecular mechanism underlying chronic visceral pain and sensitization in patients with IBS.
Collapse
Affiliation(s)
- Shufen Hu
- Institute of Neuroscience, Laboratory for Translational Pain Medicine, Department of Neurobiology, Soochow University, Suzhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|