1
|
Fang L, Yi X, Shen J, Deng N, Peng X. Gut-brain axis mediated by intestinal content microbiota was associated with Zhishi Daozhi decoction on constipation. Front Cell Infect Microbiol 2025; 15:1539277. [PMID: 39963403 PMCID: PMC11830728 DOI: 10.3389/fcimb.2025.1539277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Background Constipation is a common digestive system disorder, which is closely related to the intestinal flora. Zhishi Daozhi decoction (ZDD) is a traditional Chinese medicine prescription used to treat constipation caused by indigestion. This study is to evaluate the efficacy of ZDD in treating constipation and to elucidate the underlying mechanism. Methods In this study, Kunming mice were administered a high-protein diet (HFHPD) and loperamide hydrochloride injections to induce constipation. The mice then received varying doses (2.4, 4.7, and 9.4 mg/kg) of ZDD for seven days. Following the sampling process, we measured fecal microbial activity. The levels of 5-hydroxytryptamine (5-HT), vasoactive intestinal peptide (VIP), and aquaporin-3 (AQP3) were quantified using enzyme-linked immunosorbent assay. Changes in the gut microbiota were evaluated through 16S rRNA gene sequencing. Additionally, we investigated the correlation between specific microbiota features and the levels of 5-HT, VIP, and AQP3. Results The fecal surface of the mice in the model group (CMM) was rough and dry. The stool of mice in the low-dose ZDD group (CLD), medium-dose ZDD group (CMD), and high-dose ZDD group (CHD) exhibited a smoother texture, closely resembling that of the normal group (CNM). 5-HT levels in the CMM group were significantly lower than in the CNM, CLD, and CHD. VIP levels in the CMD were lower than in the other four groups, and AQP3 levels in CMM showed a decreasing trend. The fecal microbial activity of the CMM group was significantly higher than that of the other groups. Diversity analysis indicated that CMD and CHD treatments were more effective in restoring the intestinal microbiota structure. Potential pathogenic bacteria, including Clostridium, Aerococcus, Jeotgalicoccus, and Staphylococcus were enriched in CMM. In contrast, beneficial bacteria such as Faecalibacterium, Bacillaceae, and Bacillus were more prevalent in the CLD, CMD, and CHD. Correlation analysis revealed that Streptococcus and Enterococcus were positively correlated with VIP, while Succinivibrio showed a negative correlation with 5-HT. Conclusions Constipation induced by HFHPD and loperamide hydrochloride disrupts the structure of the intestinal microbiota. ZDD appears to alleviate constipation, potentially through mechanisms linked to the brain-gut axis and its interaction with the intestinal microbiota. Among the treatment groups, the medium dose of ZDD demonstrated the most effective results.
Collapse
Affiliation(s)
- Leyao Fang
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xin Yi
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Junxi Shen
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Na Deng
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xinxin Peng
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
2
|
Li J, Zhou J, Weng J, Jin F, Shen Y, Qi Y, Jiang M, Hei M. Rapidly progressive necrotizing enterocolitis: Risk factors and a predictive model. Pediatr Res 2025; 97:1058-1064. [PMID: 39147906 DOI: 10.1038/s41390-024-03482-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Rapidly progressive necrotizing enterocolitis (RP-NEC) is a particular subtype of NEC known for its rapid progression and high mortality rate. The objective of this study was to establish a predictive model for RP-NEC. METHODS This was a retrospective single-center cohort study. Patients were newborn infants with NEC (Bell's stage ≥ IIB) admitted from January 1, 2016 to December 31, 2023. The primary outcome was RP-NEC defined as the need for surgical intervention and/or death within 48 hours of the onset of NEC. RESULTS Totally 334 newborn infants were included, among which 82 (24.6%) were RP-NEC cases with a gestation age 34.1 (31.0, 37.0) weeks and birth weight 2100 (1413, 2800) g. Plasma sodium <135 mmol/L, C-reactive protein ≥10 mg/L, platelet count <100 × 109/L, lymphocyte count <1.5 × 109/L, pH <7.2 in blood gas, and ascites at NEC onset were identified as independent risk factors for RP-NEC. The model established presented an AUC value of 0.983 (95% CI 0.97-0.99). The calibration curve for validation was applied revealing a slope close to unity while the Hosmer-Lemeshow test yielded χ2 = 2.550 (p = 0.636). CONCLUSION The predictive model established on the above 6 items of RP-NEC is highly promising. IMPACT Currently, there is a paucity of research on this specific type of severe necrotizing enterocolitis (NEC) characterized by rapid progression. Our study was to investigate the risk factors associated with surgical intervention and/or death within 48 hours following onset in infants with NEC, establish a predictive model for infants with rapidly progressive NEC. The new data presented in this study was the ROC curve combining the above factors as well as hyponatremia.
Collapse
Affiliation(s)
- Jicheng Li
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
- National Center for Children's Health, Beijing, 100045, China
| | - Jingjing Zhou
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
- National Center for Children's Health, Beijing, 100045, China
| | - Jingwen Weng
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
- National Center for Children's Health, Beijing, 100045, China
| | - Fei Jin
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
- National Center for Children's Health, Beijing, 100045, China
| | - Yanhua Shen
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
- National Center for Children's Health, Beijing, 100045, China
| | - Yujie Qi
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
- National Center for Children's Health, Beijing, 100045, China
| | - Min Jiang
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China.
- National Center for Children's Health, Beijing, 100045, China.
| | - Mingyan Hei
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China.
- National Center for Children's Health, Beijing, 100045, China.
| |
Collapse
|
3
|
Agibalova T, Hempel A, Maurer HC, Ragab M, Ermolova A, Wieland J, Waldherr Ávila de Melo C, Heindl F, Giller M, Fischer JC, Tschurtschenthaler M, Kohnke-Ertel B, Öllinger R, Steiger K, Demir IE, Saur D, Quante M, Schmid RM, Middelhoff M. Vasoactive intestinal peptide promotes secretory differentiation and mitigates radiation-induced intestinal injury. Stem Cell Res Ther 2024; 15:348. [PMID: 39380035 PMCID: PMC11462795 DOI: 10.1186/s13287-024-03958-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Vasoactive intestinal peptide (VIP) is a neuronal peptide with prominent distribution along the enteric nervous system. While effects of VIP on intestinal motility, mucosal vasodilation, secretion, and mucosal immune cell function are well-studied, the direct impact of VIP on intestinal epithelial cell turnover and differentiation remains less understood. Intestinal stem and progenitor cells are essential for the maintenance of intestinal homeostasis and regeneration, and their functions can be modulated by factors of the stem cell niche, including neuronal mediators. Here, we investigated the role of VIP in regulating intestinal epithelial homeostasis and regeneration following irradiation-induced injury. METHODS Jejunal organoids were derived from male and female C57Bl6/J, Lgr5-EGFP-IRES-CreERT2 or Lgr5-EGFP-IRES-CreERT2/R26R-LSL-TdTomato mice and treated with VIP prior to analysis. Injury conditions were induced by exposing organoids to 6 Gy of irradiation (IR). To investigate protective effects of VIP in vivo, mice received 12 Gy of abdominal IR followed by intraperitoneal injections of VIP. RESULTS We observed that VIP promotes epithelial differentiation towards a secretory phenotype predominantly via the p38 MAPK pathway. Moreover, VIP prominently modulated epithelial proliferation as well as the number and proliferative activity of Lgr5-EGFP+ progenitor cells under homeostatic conditions. In the context of acute irradiation injury in vitro, we observed that IR injury renders Lgr5-EGFP+ progenitor cells more susceptible to VIP-induced modulations, which coincided with the strong promotion of epithelial regeneration by VIP. Finally, the observed effects translate into an in vivo model of abdominal irradiation, where VIP showed to prominently mitigate radiation-induced injury. CONCLUSIONS VIP prominently governs intestinal homeostasis by regulating epithelial progenitor cell proliferation and differentiation and promotes intestinal regeneration following acute irradiation injury.
Collapse
Affiliation(s)
- Tatiana Agibalova
- Department of Internal Medicine II, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Anneke Hempel
- Department of Internal Medicine II, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - H Carlo Maurer
- Department of Internal Medicine II, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Mohab Ragab
- Department of Internal Medicine II, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Anastasia Ermolova
- Department of Internal Medicine II, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Jessica Wieland
- Department of Internal Medicine II, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Caroline Waldherr Ávila de Melo
- Department of Internal Medicine II, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Fabian Heindl
- Department of Internal Medicine II, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Maximilian Giller
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Julius Clemens Fischer
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Markus Tschurtschenthaler
- Division of Translational Cancer Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Chair of Translational Cancer Research and Institute of Experimental Cancer Therapy, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Birgit Kohnke-Ertel
- Department of Internal Medicine II, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Rupert Öllinger
- Institute of Molecular Oncology and Functional Genomics, Center for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Else Kröner Clinician Scientist Professor for Translational Pancreatic Surgery, Munich, Germany
| | - Dieter Saur
- Division of Translational Cancer Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Chair of Translational Cancer Research and Institute of Experimental Cancer Therapy, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Michael Quante
- Department of Internal Medicine II, Faculty of Medicine, Freiburg University Medical Center, University of Freiburg, Freiburg, Germany
| | - Roland M Schmid
- Department of Internal Medicine II, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Moritz Middelhoff
- Department of Internal Medicine II, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany.
| |
Collapse
|
4
|
Dong X, Zhan Y, Yang M, Li S, Zheng H, Gao Y. miR-30c affects the pathogenesis of ulcerative colitis by regulating target gene VIP. Sci Rep 2024; 14:3472. [PMID: 38342939 PMCID: PMC10859366 DOI: 10.1038/s41598-024-54092-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/08/2024] [Indexed: 02/13/2024] Open
Abstract
MicroRNAs play a crucial role in regulating the epithelial barrier and immune response, which are implicated in the pathogenesis of ulcerative colitis (UC). This study aimed to investigate the role and molecular mechanism of miR-30c in the pathogenesis of UC using a dextran sulfate sodium salt (DSS)-induced colitis model, which is similar to ulcerative colitis. Wild-type (WT) and miR-30c knockout (KO) mice were assigned to either control or DSS-treated groups to evaluate the influence of aberrant miR-30c expression on UC pathogenesis. The disease activity index, inflammatory factors, and the extent of pathological and histological damage in colon tissues were analyzed. The effect of miR-30c on vasoactive intestinal peptide (VIP) gene expression was validated through luciferase reporter assay, qRT-PCR, Western blotting, and immunohistochemistry. The results showed that miR-30c KO mice with DSS-induced colitis model showed more severe phenotypes: significantly higher disease activity indices, significant body weight loss, reduced length of the colon of mice, increased number of aberrant crypt structures, reduced mucus secretion, and significant differences in inflammatory factors. These findings suggested that the absence of miR-30c might promote DSS-induced colitis, and the targe-regulatory effect of miR-30c on VIP might play an important role in the development of colitis.
Collapse
Affiliation(s)
- Xiang Dong
- School of Life Science, Laboratory Animal Center, Bengbu Medical College, No. 2600 Donghai Road, Bengbu, 233030, China
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, China
| | - Yuling Zhan
- School of Life Science, Laboratory Animal Center, Bengbu Medical College, No. 2600 Donghai Road, Bengbu, 233030, China
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, China
| | - Minghui Yang
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, China
- School of Basic Courses, Bengbu Medical College, Bengbu, China
| | - Suwan Li
- School of Life Science, Laboratory Animal Center, Bengbu Medical College, No. 2600 Donghai Road, Bengbu, 233030, China
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, China
| | - Hailun Zheng
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yu Gao
- School of Life Science, Laboratory Animal Center, Bengbu Medical College, No. 2600 Donghai Road, Bengbu, 233030, China.
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, China.
- Laboratory Animal Center, Bengbu Medical College, Bengbu, China.
| |
Collapse
|
5
|
Wiese JJ, Manna S, Kühl AA, Fascì A, Elezkurtaj S, Sonnenberg E, Bubeck M, Atreya R, Becker C, Weixler B, Siegmund B, Patankar JV, Prüß MS, Schumann M. Myenteric Plexus Immune Cell Infiltrations and Neurotransmitter Expression in Crohn's Disease and Ulcerative Colitis. J Crohns Colitis 2024; 18:121-133. [PMID: 37565754 PMCID: PMC10821712 DOI: 10.1093/ecco-jcc/jjad122] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND AND AIMS Pain is a cardinal symptom in inflammatory bowel disease [IBD]. An important structure in the transduction of pain signalling is the myenteric plexus [MP]. Nevertheless, IBD-associated infiltration of the MP by immune cells lacks in-depth characterisation. Herein, we decipher intra- and periganglionic immune cell infiltrations in Crohn´s disease [CD] and ulcerative colitis [UC] and provide a comparison with murine models of colitis. METHODS Full wall specimens of surgical colon resections served to examine immune cell populations by either conventional immuno-histochemistry or immunofluorescence followed by either bright field or confocal microscopy. Results were compared with equivalent examinations in various murine models of intestinal inflammation. RESULTS Whereas the MP morphology was not significantly altered in IBD, we identified intraganglionic IBD-specific B cell- and monocyte-dominant cell infiltrations in CD. In contrast, UC-MPs were infiltrated by CD8+ T cells and revealed a higher extent of ganglionic cell apoptosis. With regard to the murine models of intestinal inflammation, the chronic dextran sulphate sodium [DSS]-induced colitis model reflected CD [and to a lesser extent UC] best, as it also showed increased monocytic infiltration as well as a modest B cell and CD8+ T cell infiltration. CONCLUSIONS In CD, MPs were infiltrated by B cells and monocytes. In UC, mostly CD8+ cytotoxic T cells were found. The chronic DSS-induced colitis in the mouse model reflected best the MP-immune cell infiltrations representative for IBD.
Collapse
Affiliation(s)
- Jakob J Wiese
- Medizinische Klinik m. S. Gastroenterologie, Infektiologie und Rheumatologie, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Subhakankha Manna
- Medizinische Klinik m. S. Gastroenterologie, Infektiologie und Rheumatologie, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anja A Kühl
- Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, iPATH.Berlin, Berlin, Germany
| | - Alberto Fascì
- Medizinische Klinik m. S. Gastroenterologie, Infektiologie und Rheumatologie, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sefer Elezkurtaj
- Institute of Pathology, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Elena Sonnenberg
- Medizinische Klinik m. S. Gastroenterologie, Infektiologie und Rheumatologie, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marvin Bubeck
- Department of Internal Medicine 1, University Hospital Erlangen, Faculty of Medicine, Erlangen, Germany
| | - Raja Atreya
- Department of Internal Medicine 1, University Hospital Erlangen, Faculty of Medicine, Erlangen, Germany
| | - Christoph Becker
- Department of Internal Medicine 1, University Hospital Erlangen, Faculty of Medicine, Erlangen, Germany
| | - Benjamin Weixler
- Klinik für Allgemein- und Viszeralchirurgie, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Britta Siegmund
- Medizinische Klinik m. S. Gastroenterologie, Infektiologie und Rheumatologie, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jay V Patankar
- Department of Internal Medicine 1, University Hospital Erlangen, Faculty of Medicine, Erlangen, Germany
| | - Magdalena S Prüß
- Medizinische Klinik m. S. Gastroenterologie, Infektiologie und Rheumatologie, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin, – Berlin, BIH Biomedical Innovation Academy, BIH, Charité Clinician Scientist Program, 10178 Berlin, Germany
| | - Michael Schumann
- Medizinische Klinik m. S. Gastroenterologie, Infektiologie und Rheumatologie, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin, – Berlin, BIH Biomedical Innovation Academy, BIH, Charité Clinician Scientist Program, 10178 Berlin, Germany
| |
Collapse
|
6
|
Zhang RB, Ren L, Ding DP, Wang HD, Peng J, Zheng K. Protective Effect of the SIRT1-Mediated NF-κB Signaling Pathway against Necrotizing Enterocolitis in Neonatal Mice. Eur J Pediatr Surg 2023; 33:386-394. [PMID: 36379465 DOI: 10.1055/s-0042-1758157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To discover the mechanism of the sirtuin 1 (SIRT1)-mediated nuclear factor-κB (NF-κB) pathway in the protection against necrotizing enterocolitis (NEC) in neonatal mice. MATERIALS AND METHODS Neonatal mice were treated with EX527 (an inhibitor of SIRT1) and/or pyrrolidine dithiocarbamate (PDTC, an inhibitor of NF-κB). The survival rate of the mice was recorded. Hematoxylin and eosin (HE) staining was performed to observe the pathological changes in the intestines. Furthermore, western blotting, enzyme-linked immunosorbent assay, and real-time quantitative polymerase chain reaction were conducted to measure the protein and gene expression, while corresponding kits were used to detect the levels of oxidative stress indicators. RESULTS PDTC increased the survival rate of NEC mice. When compared with the NEC+ EX527 + PDTC group, the histological NEC score was higher in the NEC + EX527 group but lower in the NEC + PDTC group. SIRT1 expression in the intestines of NEC mice was downregulated, with an increase in p65 nuclear translocation. Additionally, malondialdehyde increased and glutathione peroxidase decreased in the intestines of NEC mice, with the upregulation of interleukin (IL)-6, IL-1β, and tumor necrosis factor-α, as well as the downregulation of ZO-1, occludin, and claudin-4 in the intestines. However, the above changes could be improved by PDTC, which could be further reversed by EX527. CONCLUSION SIRT1 can mitigate inflammation and the oxidative stress response and improve intestinal permeability by mediating the NF-κB pathway, playing an important role in the alleviation of NEC.
Collapse
Affiliation(s)
- Rui-Bo Zhang
- Department of Pediatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Lan Ren
- Department of Pediatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - De-Ping Ding
- Department of Infectious Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Heng-Dong Wang
- Department of Pediatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Juan Peng
- Department of Blood Transfusion, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Kun Zheng
- Department of Pediatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| |
Collapse
|
7
|
Fan Z, Jia W. Lactobacillus casei-Derived Postbiotics Elevate the Bioaccessibility of Proteins via Allosteric Regulation of Pepsin and Trypsin and Introduction of Endopeptidases. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37410960 DOI: 10.1021/acs.jafc.3c02125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
The potential of probiotics to benefit digestion has been widely reported, while its utilization in high-risk patients and potential adverse reactions have focused interest on postbiotics. A variable data-independent acquisition (vDIA)-based spatial-omics strategy integrated with unsupervised variational autoencoders was applied to profile the functional mechanism underlying the action of Lactobacillus casei-derived postbiotic supplementation in goat milk digestion in an infant digestive system, from a metabolomics-peptidomics-proteomics perspective. Amide and olefin derivatives were proved to elevate the activities of pepsin and trypsin through hydrogen bonding and hydrophobic forces based on allosteric effects, and recognition of nine endopeptidases and their cleavage to serine, proline, and aspartate were introduced by postbiotics, thereby promoting the generation of hydrophilic peptides and elevating the bioaccessibility of goat milk protein. The peptides originating from αs1-casein, β-casein, β-lactoglobulin, Ig-like domain-containing protein, κ-casein, and serum amyloid A protein, with multiple bioactivities including angiotensin I-converting enzyme (ACE)-inhibitory, osteoanabolic, dipeptidyl peptidase IV (DPP-IV) inhibitory, antimicrobial, bradykinin-potentiating, antioxidant, and anti-inflammatory activities, were significantly increased in the postbiotic supplementation group, which was also considered to potentially prevent necrotizing enterocolitis through inhibiting the multiplication of pathogenic bacteria and blocking signal transducer and activator of transcription 1 and nuclear factor kappa-light-chain-enhancer of activated B cells inflammatory pathways. This research deepened the understanding of the mechanism underlying the postbiotics affecting goat milk digestion, which established a critical groundwork for the clinical application of postbiotics in infant complementary foods.
Collapse
Affiliation(s)
- Zibian Fan
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Wei Jia
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
- Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an 710021, China
| |
Collapse
|
8
|
Yi X, Zhou K, Jiang P, Deng N, Peng X, Tan Z. Brain-bacteria-gut axis and oxidative stress mediated by intestinal mucosal microbiota might be an important mechanism for constipation in mice. 3 Biotech 2023; 13:192. [PMID: 37205176 PMCID: PMC10185723 DOI: 10.1007/s13205-023-03580-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 04/21/2023] [Indexed: 05/21/2023] Open
Abstract
Intestinal microbiota disorder was associated with constipation. This study investigated the microbiota-gut-brain axis and oxidative stress mediated by intestinal mucosal microbiota in mice with spleen deficiency constipation. The Kunming mice were randomly divided into the control (MC) group and the constipation (MM) group. The spleen deficiency constipation model was established by gavage with Folium sennae decoction and controlled diet and water intake. The body weight, spleen and thymus index, 5-Hydroxytryptamine (5-HT) and Superoxide Dismutase (SOD) content were significantly lower in the MM group than the MC group, the content of vasoactive intestinal peptide (VIP) and malondialdehyde (MDA) content were significantly higher than the MC group. The Alpha diversity of intestinal mucosal bacteria was not changed but beta diversity was changed in mice with spleen deficiency constipation. Compared to the MC group, the relative abundance of Proteobacteria was an upward trend and the Firmicutes/Bacteroidota (F/B) value was a downward trend in the MM group. There was a significant difference in the characteristic microbiota between the two groups. In the MM group, Brevinema, Akkermansia, Parasutterella, Faecalibaculum, Aeromonas, Sphingobium, Actinobacillus, and other pathogenic bacteria were enriched. Meanwhile, there was a certain relationship between the microbiota and gastrointestinal neuropeptide and oxidative stress indicators. The community structure of intestinal mucosal bacteria in mice with spleen deficiency constipation was changed, which was characterized by the reduction of F/B value and enrichment of Proteobacteria. Microbiota-gut-brain axis may be important for spleen deficiency constipation.
Collapse
Affiliation(s)
- Xin Yi
- The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208 China
| | - Kang Zhou
- The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208 China
| | - Ping Jiang
- The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208 China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007 China
| | - Na Deng
- The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208 China
| | - Xinxin Peng
- The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208 China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007 China
| | - Zhoujin Tan
- The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208 China
| |
Collapse
|
9
|
Yi X, Zhou K, Deng N, Cai Y, Peng X, Tan Z. Simo decoction curing spleen deficiency constipation was associated with brain-bacteria-gut axis by intestinal mucosal microbiota. Front Microbiol 2023; 14:1090302. [PMID: 36846756 PMCID: PMC9947565 DOI: 10.3389/fmicb.2023.1090302] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 01/20/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Simo decoction (SMD) is a traditional prescription for treating gastrointestinal diseases. More and more evidences prove that SMD can treat constipation by regulating intestinal microbiota and related oxidative stress indicators, but the specific mechanism is still unclear. METHODS A network pharmacological analysis was used to predict the medicinal substances and potential targets of SMD to alleviate constipation. Then, 15 male mice were randomly divided into normal group (MN group), natural recovery group (MR group), and SMD treatment group (MT group). Constipation model mice were constructed by gavage of Folium sennae decoction and control of diet and drinking water, and SMD was used for intervention after successful modeling. The levels of 5-hydroxytryptamine (5-HT), vasoactive intestinal peptide (VIP), superoxide dismutase (SOD), malondialdehyde (MDA), and fecal microbial activities were measured, and the intestinal mucosal microbiota was sequenced. RESULT Network pharmacology analysis showed that a total of 24 potential active components were obtained from SMD, and 226 target proteins were obtained after conversion. Meanwhile, we obtained 1,273 and 424 disease-related targets in the GeneCards database and the DisGeNET database, respectively. After combination and deduplication, the disease targets shared 101 targets with the potential active components of SMD. When the mice were intervened with SMD, the 5-HT, VIP, MDA, SOD content, and microbial activity in MT group were close to MN group, and Chao 1 and ACE in MT group were significantly higher than that in MR group. In the Linear discriminant analysis Effect Size (LEfSe) analysis, the abundance of beneficial bacteria such as Bacteroides, Faecalibacterium, Alistipes, Subdoligranulum, Lactiplantibacillus, and Phascolarctobacterium in MT group increased. At the same time, there were some associations between microbiota and brain-gut peptides and oxidative stress indicators. CONCLUSION SMD can promote intestinal health and relieve constipation through brain-bacteria-gut axis associating with intestinal mucosal microbiota and alleviate oxidative stress.
Collapse
Affiliation(s)
- Xin Yi
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Kang Zhou
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Na Deng
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Ying Cai
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xinxin Peng
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Zhoujin Tan
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
10
|
An L, Li J, Liu B, Hui J, Zhang Q, Zhang X, Wang Q. Amniotic fluid stem cell attenuated necrotizing enterocolitis progression by promoting Rspo3/AMPKα axis. Immunobiology 2023:152336. [PMID: 37173190 DOI: 10.1016/j.imbio.2023.152336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 12/28/2022] [Accepted: 01/09/2023] [Indexed: 01/17/2023]
Abstract
R-spondin 3 (Rspo3) is involved in various cellular processes. The alteration of Rspo3 participates in the differentiation of intestinal epithelial cells which are the crucial effector cells during necrotizing enterocolitis (NEC) development. Amniotic fluid stem cells (AFSCs) were recently indicated as a potential approach for NEC therapy. This study aimed to illustrate the regulatory role and mechanism of Rspo3 in the pathogenesis of NEC and whether AFSCs therapy would impact NEC by mediating Rspo3. First, the alteration of Rspo3 was investigated in the serum and tissues of NEC patients, and an in vitro cell model induced by LPS. A gain-of-function assay was conducted to explore the function of Rspo3 in NEC. Through the analysis of adenosine 5'-monophosphate-activated protein kinase α (AMPKα) activation, the mechanism of Rspo3-mediated NEC progression was demonstrated. Finally, AFSCs were used to coculture human intestinal epithelial cells (HIECs) and the impacts on NEC development were also explored. The results found that Rspo3 was dramatically depressed during NEC progression and reversing Rspo3 expression ameliorated LPS-induced injury, inflammation, oxidative stress and tight junction dysregulation in HIECs. Besides, Rspo3 overexpression reversed AMPKα inactivation induced by NEC and an AMPKα inhibitor, Compound C, blocked the effect of Rspo3 overexpression on NEC. AFSCs treatment was beneficial for NEC therapy by restoring Rspo3 expression which was counteracted by exosome inhibitor. Generally, AFSCs attenuated NEC progression by promoting the Rspo3/AMPKα axis which might exert via the secretion of exosomes. Our conclusions might be valuable for NEC diagnosis and therapy.
Collapse
|
11
|
Xie X, Geng C, Li X, Liao J, Li Y, Guo Y, Wang C. Roles of gastrointestinal polypeptides in intestinal barrier regulation. Peptides 2022; 151:170753. [PMID: 35114316 DOI: 10.1016/j.peptides.2022.170753] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/29/2022] [Accepted: 01/30/2022] [Indexed: 12/17/2022]
Abstract
The intestinal barrier is a dynamic entity that is organized as a multilayer system and includes various intracellular and extracellular elements. The gut barrier functions in a coordinated manner to impede the passage of antigens, toxins, and microbiome components and simultaneously preserves the balanced development of the epithelial barrier and the immune system and the acquisition of tolerance to dietary antigens and intestinal pathogens.Numerous scientific studies have shown a significant association between gut barrier damage and gastrointestinal and extraintestinal diseases such as inflammatory bowel disease, celiac disease and hepatic fibrosis. Various internal and external factors regulate the intestinal barrier. Gastrointestinal peptides originate from enteroendocrine cells in the luminal digestive tract and are critical gut barrier regulators. Recent studies have demonstrated that gastrointestinal peptides have a therapeutic effect on digestive tract diseases, enhancing epithelial barrier activity and restoring the gut barrier. This review demonstrates the roles and mechanisms of gastrointestinal polypeptides, especially somatostatin (SST) and vasoactive intestinal peptide (VIP), in intestinal barrier regulation.
Collapse
Affiliation(s)
- Xiaoxi Xie
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Chong Geng
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Xiao Li
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China; Division of Digestive Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Juan Liao
- Non-communicable Diseases Research Center, West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
| | - Yanni Li
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Yaoyu Guo
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Chunhui Wang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Chandramowlishwaran P, Raja S, Maheshwari A, Srinivasan S. Enteric Nervous System in Neonatal Necrotizing Enterocolitis. Curr Pediatr Rev 2022; 18:9-24. [PMID: 34503418 DOI: 10.2174/1573396317666210908162745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/26/2021] [Accepted: 06/08/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The pathophysiology of necrotizing enterocolitis (NEC) is not clear, but increasing information suggests that the risk and severity of NEC may be influenced by abnormalities in the enteric nervous system (ENS). OBJECTIVE The purpose of this review was to scope and examine the research related to ENS-associated abnormalities that have either been identified in NEC or have been noted in other inflammatory bowel disorders (IBDs) with histopathological abnormalities similar to NEC. The aim was to summarize the research findings, identify research gaps in existing literature, and disseminate them to key knowledge end-users to collaborate and address the same in future studies. METHODS Articles that met the objectives of the study were identified through an extensive literature search in the databases PubMed, EMBASE, and Scopus. RESULTS The sources identified through the literature search revealed that: (1) ENS may be involved in NEC development and post-NEC complications, (2) NEC development is associated with changes in the ENS, and (3) NEC-associated changes could be modulated by the ENS. CONCLUSION The findings from this review identify the enteric nervous as a target in the development and progression of NEC. Thus, factors that can protect the ENS can potentially prevent and treat NEC and post-NEC complications. This review serves to summarize the existing literature and highlights a need for further research on the involvement of ENS in NEC.
Collapse
Affiliation(s)
- Pavithra Chandramowlishwaran
- Department of Medicine, Emory University School of Medicine, Decatur, GA, USA.,Gastroenterology Research, Atlanta VA Medical Center, Decatur, GA, USA
| | - Shreya Raja
- Department of Medicine, Emory University School of Medicine, Decatur, GA, USA.,Gastroenterology Research, Atlanta VA Medical Center, Decatur, GA, USA
| | - Akhil Maheshwari
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA
| | - Shanthi Srinivasan
- Department of Medicine, Emory University School of Medicine, Decatur, GA, USA.,Gastroenterology Research, Atlanta VA Medical Center, Decatur, GA, USA
| |
Collapse
|
13
|
Song X, Pi S, Gao Y, Zhou F, Yan S, Chen Y, Qiao L, Dou X, Shao D, Xu C. The Role of Vasoactive Intestinal Peptide and Mast Cells in the Regulatory Effect of Lactobacillus casei ATCC 393 on Intestinal Mucosal Immune Barrier. Front Immunol 2021; 12:723173. [PMID: 34899686 PMCID: PMC8657605 DOI: 10.3389/fimmu.2021.723173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/09/2021] [Indexed: 01/07/2023] Open
Abstract
Vasoactive intestinal peptide (VIP) plays an important role in the neuro-endocrine-immune system. Mast cells (MCs) are important immune effector cells. This study was conducted to investigate the protective effect of L. casei ATCC 393 on Enterotoxigenic Escherichia coli (ETEC) K88-induced intestinal mucosal immune barrier injury and its association with VIP/MC signaling by in vitro experiments in cultures of porcine mucosal mast cells (PMMCs) and in vivo experiments using VIP receptor antagonist (aVIP) drug. The results showed that compared with the ETEC K88 and lipopolysaccharides (LPS)-induced model groups, VIP pretreatment significantly inhibited the activation of MCs and the release of β-hexosaminidase (β-hex), histamine and tryptase. Pretreatment with aVIP abolished the protective effect of L. casei ATCC 393 on ETEC K88-induced intestinal mucosal immune barrier dysfunction in C57BL/6 mice. Also, with the blocking of VIP signal transduction, the ETEC K88 infection increased serum inflammatory cytokines, and the numbers of degranulated MCs in ileum, which were decreased by administration of L. casei ATCC 393. In addition, VIP mediated the regulatory effect of L. casei ATCC 393 on intestinal microbiota in mice. These findings suggested that VIP may mediate the protective effect of L.casei ATCC 393 on intestinal mucosal immune barrier dysfunction via MCs.
Collapse
Affiliation(s)
- Xiaofan Song
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Shanyao Pi
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yueming Gao
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Fengxia Zhou
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Shuqi Yan
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yue Chen
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Lei Qiao
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xina Dou
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Dongyan Shao
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Chunlan Xu
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
14
|
Griffiths V, Al Assaf N, Khan R. Review of claudin proteins as potential biomarkers for necrotizing enterocolitis. Ir J Med Sci 2021; 190:1465-1472. [PMID: 33492576 PMCID: PMC8521514 DOI: 10.1007/s11845-020-02490-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022]
Abstract
Background Claudin proteins are a component of tight junctions found in cell-cell adhesion complexes. A central feature of necrotizing enterocolitis (NEC) is intestinal permeability, with changes to claudin proteins potentially contributing to intestinal instability, inflammation, and the progression of NEC. A current area of interest is the development of a novel, noninvasive biomarker for the detection of NEC in neonates at risk of developing this disease, in order to reduce morbidity and mortality through earlier intervention. Aims This review aims to explore the relevance of claudin proteins in the pathophysiology of NEC and their potential usefulness as a biomarker. Methods This review was conducted using the search terms “claudin” + “necrotizing enterocolitis”, with 27 papers selected for review. Results Claudin proteins appear to have a role in the stability of the gut epithelium through the regulation of intestinal permeability, maturity, and inflammation. Formula feeding has been shown to promote inflammation and result in changes to claudin proteins, while breastfeeding and certain nutritional supplements lead to reduced inflammation and improved intestinal stability as demonstrated through changes to claudin protein expression. Preliminary studies in human neonates suggest that urinary claudin measurements may be used to predict the development of NEC. Conclusions Alterations to claudin proteins may reflect changes seen to intestinal permeability and inflammation in the context of NEC. Further research is necessary to understand the relevance of claudin proteins in the pathophysiology of NEC and their use as a biomarker.
Collapse
Affiliation(s)
- Victoria Griffiths
- Graduate Entry Medical School, University of Limerick, Limerick, Ireland.
| | - Niazy Al Assaf
- Department of Neonatology, University Maternity Hospital Limerick, Limerick, Ireland
| | - Rizwan Khan
- Department of Neonatology, University Maternity Hospital Limerick, Limerick, Ireland
| |
Collapse
|
15
|
Peptidomics Analysis Discloses That Novel Bioactive Peptides Participate in Necrotizing Enterocolitis in a Rat Model. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4705149. [PMID: 33490244 PMCID: PMC7790586 DOI: 10.1155/2020/4705149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 12/06/2020] [Accepted: 12/17/2020] [Indexed: 11/17/2022]
Abstract
Necrotizing enterocolitis (NEC) is a common devastating gastrointestinal disease in premature infants, the molecular mechanisms of which have not been fully elucidated. Recently, endogenous peptides have garnered much attention owing to their role in diagnosis and treatment. However, changes in the peptide expression of NEC intestinal tissues remain poorly understood. In the present study, a comparative peptidomics profiling analysis was performed between NEC and control intestinal tissues via liquid chromatography-tandem mass spectrometry (LC-MS). In total, 103 upregulated and 73 downregulated peptides were identified in the intestinal tissues (fold change ≥ 1.5, p < 0.05). Bioinformatics analysis revealed that these differentially expressed peptides were significantly associated with NEC pathophysiology, including apoptosis, the TGF-β signaling pathway, the Wnt signaling pathway, and the MAPK signaling pathway. Furthermore, two putative peptides could inhibit apoptosis and promote the migration of intestinal epithelial cells induced by lipopolysaccharide; these peptides were derived from the protein domains MT1 and EZRI, respectively. In conclusion, our study revealed that endogenous peptides are involved in the pathophysiologic mechanism of NEC; nevertheless, further exploration is required in this regard.
Collapse
|
16
|
Hwang JT, Yu JW, Nam HJ, Song SK, Sung WY, Kim, Y, Cho JH. Suppressive Effects of a Truncated Inhibitor K562 Protein-Derived Peptide on Two Proinflammatory Cytokines, IL-17 and TNF-α. J Microbiol Biotechnol 2020; 30:1810-1818. [PMID: 32958733 PMCID: PMC9728226 DOI: 10.4014/jmb.2004.04062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Inhibitor K562 (IK) protein was first isolated from the culture medium of K562 cells, a leukemia cell line, and is an inhibitory regulator of interferon-γ-induced major histocompatibility complex class II expression. Recently, exogenous truncated IK (tIK) protein showed potential as a therapeutic agent for inflammation-related diseases. In this study, we designed a novel putative anti-inflammatory peptide derived from tIK protein based on homology modeling of the human interleukin-10 (hIL-10) structure, and investigated whether the peptide exerted inhibitory effects against proinflammatory cytokines such as IL-17 and tumor necrosis factor-α (TNF-α). The peptide contains key residues involved in binding hIL-10 to the IL-10 receptor, and exerted strong inhibitory effects on IL- 17 (43.8%) and TNF-α (50.7%). In addition, we used circular dichroism spectroscopy to confirm that the peptide is usually present in a random coil configuration in aqueous solution. In terms of toxicity, the peptide was found to be biologically safe. The mechanisms by which the short peptide derived from human tIK protein exerts inhibitory effects against IL-17 and TNF-α should be explored further. We also evaluated the feasibility of using this novel peptide in skincare products.
Collapse
Affiliation(s)
- Jong Tae Hwang
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea
| | - Ji Won Yu
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea,Department of Biology, Kongju National University, Kongju 3588, Republic of Korea
| | - Hee Jin Nam
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea
| | - Sun Kwang Song
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea
| | - Woo Yong Sung
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea
| | - Yongae Kim,
- Department of Chemistry, Hankuk University of Foreign Studies, Yongin 1705, Republic of Korea
| | - Jang-Hee Cho
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea,Corresponding author Phone: +82-31-695-7959 Fax: +82-31-695-7986 E-mail:
| |
Collapse
|
17
|
Xu Y, Chen F. Antioxidant, Anti-Inflammatory and Anti-Apoptotic Activities of Nesfatin-1: A Review. J Inflamm Res 2020; 13:607-617. [PMID: 33061526 PMCID: PMC7532075 DOI: 10.2147/jir.s273446] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/29/2020] [Indexed: 12/12/2022] Open
Abstract
Nesfatin-1, a newly identified energy-regulating peptide, is widely expressed in the central and peripheral tissues, and has a variety of physiological activities. A large number of recent studies have shown that nesfatin-1 exhibits antioxidant, anti-inflammatory, and anti-apoptotic properties and is involved in the occurrence and progression of various diseases. This review summarizes current data focusing on the therapeutic effects of nesfatin-1 under different pathophysiological conditions and the mechanisms underlying its antioxidant, anti-inflammatory, and anti-apoptotic activities.
Collapse
Affiliation(s)
- Yayun Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, People's Republic of China
| | - Feihu Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, People's Republic of China
| |
Collapse
|
18
|
Dulari Jayawardena, Arivarasu N Anbazhagan, Priyamvada S, Kumar A, Saksena S, Onyuksel H, Pradeep K Dudeja. Colonic delivery of vasoactive intestinal peptide nanomedicine alleviates colitis and shows promise as an oral capsule. Nanomedicine (Lond) 2020; 15:2459-2474. [PMID: 32975467 DOI: 10.2217/nnm-2020-0280] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aim: To evaluate the efficacy of locally delivered nanomedicine, vasoactive intestinal peptide in sterically stabilized micelles (VIP-SSM) to the colon and conduct in vitro release studies of a potential oral formulation. Materials & methods: Intracolonic instillation of VIP-SSM was tested in a mouse model of dextran sulfate sodium-induced colitis. Based on the effective mouse dose, human equivalent dose containing nanomedicine powder was filled into enteric coated capsules for in vitro release testing. Results: Colonic delivery of VIP-SSM significantly alleviated colitis. VIP-SSM containing capsules completely dissolved at colonic pH allowing micelles to reform with active VIP. Capsule formulations exhibited reproducible release profiles when stored up to 6 weeks demonstrating stability. Conclusion: VIP-SSM is an effective nanomedicine formulation which appears to have potential for oral treatment of colitis in humans. [Formula: see text].
Collapse
Affiliation(s)
- Dulari Jayawardena
- Department of Medicine, College of Medicine, University of Illinois at Chicago, IL 60612, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, IL 60612, USA
| | - Arivarasu N Anbazhagan
- Department of Medicine, College of Medicine, University of Illinois at Chicago, IL 60612, USA
| | - Shubha Priyamvada
- Department of Medicine, College of Medicine, University of Illinois at Chicago, IL 60612, USA
| | - Anoop Kumar
- Department of Medicine, College of Medicine, University of Illinois at Chicago, IL 60612, USA.,Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Seema Saksena
- Department of Medicine, College of Medicine, University of Illinois at Chicago, IL 60612, USA.,Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Hayat Onyuksel
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, IL 60612, USA
| | - Pradeep K Dudeja
- Department of Medicine, College of Medicine, University of Illinois at Chicago, IL 60612, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, IL 60612, USA.,Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
19
|
Song Y, Kim M, Kim Y. Homology Modeling and Optimized Expression of Truncated IK Protein, tIK, as an Anti-Inflammatory Peptide. Molecules 2020; 25:molecules25194358. [PMID: 32977406 PMCID: PMC7583991 DOI: 10.3390/molecules25194358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 11/24/2022] Open
Abstract
Rheumatoid arthritis, caused by abnormalities in the autoimmune system, affects about 1% of the population. Rheumatoid arthritis does not yet have a proper treatment, and current treatment has various side effects. Therefore, there is a need for a therapeutic agent that can effectively treat rheumatoid arthritis without side effects. Recently, research on pharmaceutical drugs based on peptides has been actively conducted to reduce negative effects. Because peptide drugs are bio-friendly and bio-specific, they are characterized by no side effects. Truncated-IK (tIK) protein, a fragment of IK protein, has anti-inflammatory effects, including anti-rheumatoid arthritis activity. This study focused on the fact that tIK protein phosphorylates the interleukin 10 receptor. Through homology modeling with interleukin 10, short tIK epitopes were proposed to find the essential region of the sequence for anti-inflammatory activity. TH17 differentiation experiments were also performed with the proposed epitope. A peptide composed of 18 amino acids with an anti-inflammatory effect was named tIK-18mer. Additionally, a tIK 9-mer and a 14-mer were also found. The procedure for the experimental expression of the proposed tIK series (9-mer, 14-mer, and 18-mer) using bacterial strain is discussed.
Collapse
Affiliation(s)
| | | | - Yongae Kim
- Correspondence: ; Tel.: +82-2-2173-8705; Fax: +82-31-330-4566
| |
Collapse
|
20
|
Wang Y, Sims CE, Allbritton NL. Enterochromaffin Cell-Enriched Monolayer Platform for Assaying Serotonin Release from Human Primary Intestinal Cells. Anal Chem 2020; 92:12330-12337. [PMID: 32819098 DOI: 10.1021/acs.analchem.0c02016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Enteroendocrine (EE) cells within the intestinal epithelium produce a range of hormones that have key roles in modulating satiety and feeding behavior in humans. The regulation of hormone release from EE cells as a potential therapeutic strategy to treat metabolic disorders is highly sought after by the pharmaceutical industry. However, functional studies are limited by the scarcity of EE cells (or surrogates) in both in vivo and in vitro systems. Enterochromaffin (EC) cells are a subtype of EE cells that produce serotonin (5HT). Here, we explored simple strategies to enrich EC cells in in vitro monolayer systems derived from human primary intestinal stem cells. During differentiation of the monolayers, the EC cell lineage was significantly altered by both the culture method [air-liquid interface (ALI) vs submerged] and the presence of vasoactive intestinal peptide (VIP). Compared with traditional submerged cultures without VIP, VIP-assisted ALI culture significantly boosted the number of EC cells and their 5HT secretion by up to 430 and 390%, respectively. The method also increased the numbers of other subtypes of EE cells such as L cells. Additionally, this method generated monolayers with enhanced barrier integrity, so that directional (basal or apical) 5HT secretion was measurable. For all donor tissues, the enriched EC cells improved the signal-to-background ratio and reliability of 5HT release assays. The enhancement in the 5HT secretion behavior was consistent over time from a single donor, but significant variation in the amount of secreted 5HT was present among tissues derived from five different donors. To demonstrate the utility of the EC-enriched monolayer system, 13 types of pungent food ingredients were screened for their ability to stimulate 5HT secretion. Curcumin found in the spice turmeric derived from the Curcuma longa plant was found to be the most potent secretagogue. This EC-enriched cell monolayer platform can provide a valuable analytical tool for the high-throughput screening of nutrients and gut microbial components that alter the secretion of 5HT.
Collapse
Affiliation(s)
- Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States.,Department of Bioengineering, University of Washington, Seattle, Washington 98105, United States
| | - Christopher E Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States.,Department of Bioengineering, University of Washington, Seattle, Washington 98105, United States
| | - Nancy L Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States.,Department of Bioengineering, University of Washington, Seattle, Washington 98105, United States
| |
Collapse
|
21
|
Vasoactive Intestinal Polypeptide in the Carotid Body-A History of Forty Years of Research. A Mini Review. Int J Mol Sci 2020; 21:ijms21134692. [PMID: 32630153 PMCID: PMC7370131 DOI: 10.3390/ijms21134692] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022] Open
Abstract
Vasoactive intestinal polypeptide (VIP) consists of 28 amino acid residues and is widespread in many internal organs and systems. Its presence has also been found in the nervous structures supplying the carotid body not only in mammals but also in birds and amphibians. The number and distribution of VIP in the carotid body clearly depends on the animal species studied; however, among all the species, this neuropeptide is present in nerve fibers around blood vessels and between glomus cell clusters. It is also known that the number of nerves containing VIP located in the carotid body may change under various pathological and physiological factors. The knowledge concerning the functioning of VIP in the carotid body is relatively limited. It is known that VIP may impact the glomus type I cells, causing changes in their spontaneous discharge, but the main impact of VIP on the carotid body is probably connected with the vasodilatory effects of this peptide and its influence on blood flow and oxygen delivery. This review is a concise summary of forty years of research concerning the distribution of VIP in the carotid body.
Collapse
|
22
|
Zmora O, Gutzeit O, Segal L, Boulos S, Millo Z, Ginsberg Y, Khatib N, Dabbah-Assad F, Fainaru O, Weiner Z, Beloosesky R. Prophylactic antenatal N-Acetyl Cysteine administration combined with postnatal administration can decrease mortality and injury markers associated with necrotizing enterocolitis in a rat model. PLoS One 2020; 15:e0233612. [PMID: 32479520 PMCID: PMC7263616 DOI: 10.1371/journal.pone.0233612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 05/08/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease of neonates, especially premature neonates. To date, there is no prophylactic treatment against NEC, except breast milk and slow increase in enteral feeding, and there is no antenatal prophylaxis. AIMS To assess possible protective effects of antenatal N-Acetyl Cysteine (NAC) against the intestinal pathophysiological changes associated with NEC in a rat model of NEC and against its associated mortality. METHODS Newborn Sprague-Dawley rats were divided into 5 groups: control (n = 33); NEC (n = 32)-subjected to hypoxia and formula feeding for 4 days to induce NEC; NEC-NAC (n = 34)-with induced NEC and concomitant postnatal NAC administration; NAC-NEC (n = 33)-born to dams treated with NAC for the last 3 days of pregnancy starting at gestational age of 18 days, and then subjected to induced NEC after birth; NAC-NEC-NAC (n = 36)-subjected to induced NEC with both prenatal and postnatal NAC treatment. At day of life 5, weight and survival of pups in the different groups were examined, and pups were euthanized. Ileal TNF-α, IL-6, IL-1β, IL-10, NFkB p65, iNOS and cleaved caspase 3 protein levels (western blot) and mRNA expression (RT-PCR) were compared between groups. RESULTS Pup mortality was significantly reduced in the NAC-NEC-NAC group compared to NEC (11% vs. 34%, P<0.05). Ileal protein levels and mRNA expression of all injury markers tested except IL-10 were significantly increased in NEC compared to control. These markers were significantly reduced in all NAC treatment groups (NEC-NAC, NAC-NEC, and NAC-NEC-NAC) compared to NEC. The most pronounced decrease was observed in the NAC-NEC NAC group. CONCLUSIONS Antenatal NAC decreases injury markers and mortality associated with NEC in a rat model. Antenatal administration of NAC may present a novel approach for NEC prophylaxis in pregnancies with risk for preterm birth.
Collapse
Affiliation(s)
- Osnat Zmora
- Department of Pediatric Surgery, Shamir Medical Center, Zerifin, Israel
| | - Ola Gutzeit
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Linoy Segal
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Sari Boulos
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Zvika Millo
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Yuval Ginsberg
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Nizar Khatib
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Fadwa Dabbah-Assad
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Ofer Fainaru
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Zeev Weiner
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Ron Beloosesky
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| |
Collapse
|
23
|
Abstract
PURPOSE Butyrate is a short-chain fatty acid produced in the intestine. It is controversial whether butyrate is protective or destructive for the intestinal epithelium in the development of diseases like necrotizing enterocolitis (NEC), and its mechanism of action remains unclear. We aimed to determine the effect of butyrate on the intestinal epithelium by studying its effects on intestinal epithelial cells (IEC-18) exposed to injury and in vivo by investigating the effects on the intestine in an experimental model of NEC. METHODS A) In vitro study: Butyrate was given to normal IEC-18 to determine the dose triggering injury. Based on above results, low dose butyrate (1 mM) was given to H2O2-injured cells to determine its effect against inflammation. B) In vivo study: NEC was induced by hypoxia and gavage feeding between postnatal day P5 and P9 (n = 8). Breastfed mice were used as control (n = 7). Butyrate (150 mM) was administered by enema on P6 in NEC (n = 6). Distal ileum was harvested on P9. RESULTS High dose (16 mM) butyrate upregulated inflammatory marker IL-6, while low dose butyrate protected cells from injury by reducing IL-6 expression. Similarly, compared with NEC alone, NEC mice who received butyrate had reduced intestinal damage, reduced IL-6 and NF-ĸB expression, and increased intestinal tight junction marker Claudin-7. CONCLUSION Butyrate has opposite effects depending on the dose administered. Butyrate can protect cells from H2O2-induced injury and can in vivo protect the intestine from NEC. This beneficial effect is because of downregulation of inflammation and enhancement of intestinal barrier.
Collapse
|