1
|
Alhendi A, Naser SA. MCT4 inhibition attenuates inflammatory response to Mycobacterium avium paratuberculosis infection and restores intestinal epithelial integrity in vitro. Front Immunol 2025; 16:1562100. [PMID: 40297589 PMCID: PMC12034541 DOI: 10.3389/fimmu.2025.1562100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Mycobacterium avium paratuberculosis (MAP) plays a significant role in Crohn's disease (CD). Monocarboxylate transporter 4 (MCT4) is a proton-coupled symporter of lactate that facilitates the inflammatory shift in macrophages and increases their reliance on glycolysis. MCT4 is also involved in the negative regulation of intestinal epithelial barrier function. Methods In this in vitro study, we examined the role of MCT4 in macrophages and its effect on intestinal epithelial homeostasis during MAP infection. We used cultured THP-1 macrophages infected with a clinical strain of MAP (UCF4) as well as intestinal cell lines, Caco-2 and HT-29. MCT4 was inhibited using α-cyano-4-hydroxycinnamic acid (CHCα). Results Infection of THP-1 cells with MAP upregulated MCT4 expression (2 folds) and resulted in a significant increase in lactate export (1.3 folds), TNFα (13.8 folds), and IL-6 (1.3) via TLR2 activation. Consequently, intestinal damage markers were also upregulated, including MUC2 (2.5 folds), NOX-1 (2 folds), SERPINE1 (2.1 folds), IL-6 (1.6 folds), and CLDN2 (1.4 folds). Inhibition of MCT4 during MAP infection with CHCα significantly reduced TNF-α and IL-6 levels. This effect on macrophages restored baseline oxidative status and mucin production in HT-29 intestinal cells. Moreover, MCT4 inhibition in a MAP-infected THP-1-Caco-2 co-culture system restored IL-6 and SERPINE1 to normal levels and enhanced tight junction protein, TJP1 (ZO-1), expression. Conclusion Collectively, this study revealed the significant role of MCT4 in CD pathophysiology during MAP infection and highlighted MCT4 as a potential therapeutic target for CD treatment.
Collapse
Affiliation(s)
| | - Saleh A. Naser
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| |
Collapse
|
2
|
Kim A, Gorman H, Moreau F, McManus M, Dufour A, Chadee K. Human Mucin-2 Mucin-Producing Colonic Goblet-Like Cells Secrete the Chemokine CXCL8 by Activating Multiple Proinflammatory Pathways in Response to Entamoeba histolytica. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00080-X. [PMID: 40122459 DOI: 10.1016/j.ajpath.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025]
Abstract
The mucus layer produced by highly stressed goblet cells forms a protective shield in the gut to protect the underlying mucosal epithelial cells from external threats. Hypersecretion and depletion of mucin-2 (MUC2) mucin from goblet cells is characteristic of symptomatic Entamoeba histolytica infections. It was hypothesized that MUC2 depleted goblet cells could mount a second line of innate host defense by producing proinflammatory cytokines. To investigate this, it was determined whether E. histolytica could stimulate proinflammatory responses in wild-type (WT) high MUC2 mucin-producing goblet-like cells and in CRISPR-Cas9 gene-edited MUC2KO cells. In response to live E. histolytica and soluble E. histolytica proteins, WT and, to a lesser extent, MUC2KO cells produced high levels of CXCL8. Entamoeba histolytica temporally induced greater levels of CXCL8 mRNA expression and protein secretion in WT versus MUC2KO cells, which was abrogated with alleviation of endoplasmic reticulum stress with the NADPH-oxidase inhibitor diphenyleneiodonium chloride. WT cells produced elevated reactive oxygen species that induced longer half-lives of CXCL8 transcripts, which was abrogated with diphenyleneiodonium chloride. Western blotting and proteomic analyses revealed that WT cells, but not MUC2KO cells, were basally primed to respond to external stressors and responded to E. histolytica through rapid activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase, mitogen-activated protein kinase/p38, and phosphatidylinositol 3-kinase/Akt pathways, to induce CXCL8. These results suggest that colonic goblet-like cells defend against E. histolytica infections by hypersecreting mucus and to produce the chemokine, CXCL8, to recruit neutrophils.
Collapse
Affiliation(s)
- Ariel Kim
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada
| | - Hayley Gorman
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada
| | - France Moreau
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada
| | - Mackenzie McManus
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada
| | - Antoine Dufour
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada
| | - Kris Chadee
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada.
| |
Collapse
|
3
|
Di Mattia M, Sallese M, Lopetuso LR. Unfolded protein response: An essential element of intestinal homeostasis and a potential therapeutic target for inflammatory bowel disease. Acta Physiol (Oxf) 2025; 241:e14284. [PMID: 39822064 DOI: 10.1111/apha.14284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Different physiological and pathological situations can produce alterations in the cell's endoplasmic reticulum (ER), leading to a condition known as ER stress, which can trigger an intricate intracellular signal transduction system known as the unfolded protein response (UPR). UPR is primarily tailored to restore proteostasis and ER equilibrium; otherwise, if ER stress persists, it can cause programmed cell death as a cytoprotective mechanism and drive inflammatory processes. Therefore, since intestinal cells strongly rely on UPR for their biological functions and unbalanced UPR has been linked to inflammatory, metabolic, and immune disorders, here we discussed the role of the UPR within the intestinal tract, focusing on the UPR contribution to inflammatory bowel disease development. Importantly, we also highlighted the promising potential of UPR components as therapeutic targets for intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Michele Sallese
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Loris Riccardo Lopetuso
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
4
|
Zhang L, Li J, Wan Q, Bu C, Jin W, Yuan F, Zhou W. Intestinal stem cell-derived extracellular vesicles ameliorate necrotizing enterocolitis injury. Mol Cell Probes 2025; 79:101997. [PMID: 39645054 DOI: 10.1016/j.mcp.2024.101997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/09/2024]
Abstract
The therapeutic potential of intestinal stem cell-derived extracellular vesicles (ISCs-EVs) in necrotizing enterocolitis (NEC) remains largely unexplored. This research aims to investigate the therapeutic effects of ISCs-EVs on NEC. Lgr5-positive ISCs were screened from the small intestine of mice by flow cytometry, and ISCs-EVs were isolated by density gradient centrifugation. Subsequently, ISCs-EVs were identified through transmission electron microscopy, nanoparticle tracking analysis, and western blotting. Subsequently, we evaluated the efficacy of ISCs-EVs in a mouse model of NEC and found that they enhanced survival (more than 20 %), reduced intestinal damage (restore the number of intestinal crypts and decrease the expression of MPO and cleaved-caspase 3 in intestinal tissues), promoted angiogenesis (the mRNA expression of VEGF was increased by approximately 35 %), and mitigated inflammation (decreased the level of MUC1, p-NF-κB, IL-6 and TNF-α). Furthermore, in vitro assessments demonstrated that ISCs-EVs reduced apoptosis (P < 0.01) and stimulated proliferation (P < 0.05) of IEC-6 cells, while enhancing mucin secretion in LS174T cells. In summary, our study provides a comprehensive assessment of the therapeutic effects of ISCs-EVs on NEC, using both animal and cell models. This highlights their potential for use in NEC treatment.
Collapse
Affiliation(s)
- Le Zhang
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, 214023, Wuxi, Jiangsu, China; Key Laboratory of Birth Defects, Children's Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jiahong Li
- Department of Pediatric Laboratory, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, 214023, Wuxi, Jiangsu, China
| | - Qiwen Wan
- Department of Pediatric Surgery, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, 214023, Wuxi, Jiangsu, China
| | - Chaozhi Bu
- State Key Laboratory of Reproductive Medicine, Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, 214002, Wuxi, Jiangsu, China
| | - Weilai Jin
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, 214023, Wuxi, Jiangsu, China.
| | - Fuqiang Yuan
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, 214023, Wuxi, Jiangsu, China.
| | - Wenhao Zhou
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Birth Defects, Children's Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
Tozzi M, Fiore A, Travaglione S, Marcon F, Rainaldi G, Germinario EAP, Laterza I, Donati S, Macchia D, Spada M, Leoni O, Quattrini MC, Pietraforte D, Tomasoni S, Torrigiani F, Verin R, Matarrese P, Gambardella L, Spadaro F, Carollo M, Pietrantoni A, Carlini F, Panebianco C, Pazienza V, Colella F, Lucchetti D, Sgambato A, Sistigu A, Moschella F, Guidotti M, Vincentini O, Maroccia Z, Biffoni M, De Angelis R, Bracci L, Fabbri A. E. Coli cytotoxic necrotizing factor-1 promotes colorectal carcinogenesis by causing oxidative stress, DNA damage and intestinal permeability alteration. J Exp Clin Cancer Res 2025; 44:29. [PMID: 39876002 PMCID: PMC11776187 DOI: 10.1186/s13046-024-03271-w] [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: 11/23/2024] [Accepted: 12/31/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Bacterial toxins are emerging as promising hallmarks of colorectal cancer (CRC) pathogenesis. In particular, Cytotoxic Necrotizing Factor 1 (CNF1) from E. coli deserves special consideration due to the significantly higher prevalence of this toxin gene in CRC patients with respect to healthy subjects, and to the numerous tumor-promoting effects that have been ascribed to the toxin in vitro. Despite this evidence, a definitive causal link between CNF1 and CRC was missing. Here we investigated whether CNF1 plays an active role in CRC onset by analyzing pro-carcinogenic key effects specifically induced by the toxin in vitro and in vivo. METHODS Viability assays, confocal microscopy of γH2AX and 53BP1 molecules and cytogenetic analysis were carried out to assess CNF1-induced genotoxicity on non-neoplastic intestinal epithelial cells. Caco-2 monolayers and 3D Caco-2 spheroids were used to evaluate permeability alterations specifically induced by CNF1, either in the presence or in the absence of inflammation. In vivo, an inflammatory bowel disease (IBD) model was exploited to evaluate the carcinogenic potential of CNF1. Immunohistochemistry and immunofluorescence stainings of formalin-fixed paraffin-embedded (FFPE) colon tissue were carried out as well as fecal microbiota composition analysis by 16 S rRNA gene sequencing. RESULTS CNF1 induces the release of reactive oxidizing species and chromosomal instability in non-neoplastic intestinal epithelial cells. In addition, CNF1 modifies intestinal permeability by directly altering tight junctions' distribution in 2D Caco-2 monolayers, and by hindering the differentiation of 3D Caco-2 spheroids with an irregular arrangement of these junctions. In vivo, repeated intrarectal administration of CNF1 induces the formation of dysplastic aberrant crypt foci (ACF), and produces the formation of colorectal adenomas in an IBD model. These effects are accompanied by the increased neutrophilic infiltration in colonic tissue, by a mixed pro-inflammatory and anti-inflammatory cytokine milieu, and by the pro-tumoral modulation of the fecal microbiota. CONCLUSIONS Taken together, our results support the hypothesis that the CNF1 toxin from E. coli plays an active role in colorectal carcinogenesis. Altogether, these findings not only add new knowledge to the contribution of bacterial toxins to CRC, but also pave the way to the implementation of current screening programs and preventive strategies.
Collapse
Affiliation(s)
- Michela Tozzi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessia Fiore
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Sara Travaglione
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Marcon
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Gabriella Rainaldi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Angela Pia Germinario
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Ilenia Laterza
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Donati
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Daniele Macchia
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Spada
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Omar Leoni
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Sofia Tomasoni
- Department of Comparative Biomedicine and Food Science, BCA-University of Padua, Legnaro, PD, Italy
| | - Filippo Torrigiani
- Department of Comparative Biomedicine and Food Science, BCA-University of Padua, Legnaro, PD, Italy
| | - Ranieri Verin
- Department of Comparative Biomedicine and Food Science, BCA-University of Padua, Legnaro, PD, Italy
| | - Paola Matarrese
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Maria Carollo
- Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | | | - Francesca Carlini
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Concetta Panebianco
- Division of Gastroenterology, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, FG, Italy
| | - Valerio Pazienza
- Division of Gastroenterology, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, FG, Italy
| | - Filomena Colella
- Multiplex Spatial Profiling Center, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Donatella Lucchetti
- Multiplex Spatial Profiling Center, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Sgambato
- Multiplex Spatial Profiling Center, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonella Sistigu
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Federica Moschella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Guidotti
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Olimpia Vincentini
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Zaira Maroccia
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Roberta De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Bracci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Alessia Fabbri
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
6
|
Wang L, Jin L, Zhang L, Huang X, Li Z, Li Z, Li K, Xu Y, Di S, Cui S, Wang X. Exploring the Link Between Mucin 2 and Weaning Stress-Related Diarrhoea in Piglets. Int J Mol Sci 2025; 26:599. [PMID: 39859316 PMCID: PMC11765659 DOI: 10.3390/ijms26020599] [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: 12/07/2024] [Revised: 01/01/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
To explore the relationship between intestinal mucin 2 (MUC2) and weaning-induced diarrhoea in piglets, we analysed Min and Landrace piglets. The piglets were divided into a healthy weaned group, a weaned diarrhoea group, and a healthy unweaned control group. Intestinal tissues were collected, and goblet cell numbers, sizes, and degrees of intestinal injury were observed and recorded. Intestinal tissue MUC2 mRNA and protein expression were analysed via quantitative real-time PCR (qRT-PCR) and Western blotting. Min pigs presented significantly lower diarrhoea rates and intestinal injury scores than Landrace pigs (p < 0.01). The intestinal injury scores in the weaned diarrhoea group were significantly greater than those in the unweaned groups (p < 0.05), with Min pigs consistently exhibiting lower injury scores than Landrace pigs. Specifically, unweaned Min pigs presented significantly greater duodenal MUC2 mRNA (p < 0.05), and weaned healthy Min pigs presented notably greater expression in both the duodenum and jejunum (p < 0.01). These findings reveal enhanced intestinal protection against weaning stress and diarrhoea in Min pigs, with elevated MUC2 levels likely contributing to lower injury scores and milder symptoms, thus highlighting the influence of genetic differences.
Collapse
Affiliation(s)
- Li Wang
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin 150030, China; (L.W.); (L.J.); (L.Z.); (Z.L.); (Z.L.); (K.L.); (Y.X.); (S.D.)
| | - Long Jin
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin 150030, China; (L.W.); (L.J.); (L.Z.); (Z.L.); (Z.L.); (K.L.); (Y.X.); (S.D.)
| | - Liulian Zhang
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin 150030, China; (L.W.); (L.J.); (L.Z.); (Z.L.); (Z.L.); (K.L.); (Y.X.); (S.D.)
| | - Xuankai Huang
- Branch of Animal Husbandry and Veterinary, Heilongjiang Academy of Agricultural Sciences, 2 Heyi St., Longsha District, Qiqihaer 161005, China;
| | - Ziyu Li
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin 150030, China; (L.W.); (L.J.); (L.Z.); (Z.L.); (Z.L.); (K.L.); (Y.X.); (S.D.)
| | - Zhimin Li
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin 150030, China; (L.W.); (L.J.); (L.Z.); (Z.L.); (Z.L.); (K.L.); (Y.X.); (S.D.)
| | - Ke Li
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin 150030, China; (L.W.); (L.J.); (L.Z.); (Z.L.); (Z.L.); (K.L.); (Y.X.); (S.D.)
| | - Yuan Xu
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin 150030, China; (L.W.); (L.J.); (L.Z.); (Z.L.); (Z.L.); (K.L.); (Y.X.); (S.D.)
| | - Shengwei Di
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin 150030, China; (L.W.); (L.J.); (L.Z.); (Z.L.); (Z.L.); (K.L.); (Y.X.); (S.D.)
| | - Shiquan Cui
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin 150030, China; (L.W.); (L.J.); (L.Z.); (Z.L.); (Z.L.); (K.L.); (Y.X.); (S.D.)
| | - Xibiao Wang
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin 150030, China; (L.W.); (L.J.); (L.Z.); (Z.L.); (Z.L.); (K.L.); (Y.X.); (S.D.)
| |
Collapse
|
7
|
Zhang JY, Li XY, Li DX, Zhang ZH, Hu LQ, Sun CX, Zhang XN, Wu M, Liu LT. Endoplasmic reticulum stress in intestinal microecology: A controller of antineoplastic drug-related cardiovascular toxicity. Biomed Pharmacother 2024; 181:117720. [PMID: 39631125 DOI: 10.1016/j.biopha.2024.117720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
Endoplasmic reticulum (ER) stress is extensively studied as a pivotal role in the pathological processes associated with intestinal microecology. In antineoplastic drug treatments, ER stress is implicated in altering the permeability of the mechanical barrier, depleting the chemical barrier, causing dysbiosis, exacerbating immune responses and inflammation in the immune barrier. Enteric dysbiosis and intestinal dysfunction significantly affect the circulatory system in various heart disorders. In antineoplastic drug-related cardiovascular (CV) toxicity, ER stress constitutes a web of relationships in the host-microbiome symbiotic regulatory loop. Therefore, understanding the holobiont perspective will help de-escalate spatial and temporal restrictions. This review investigates the role of ER stress-mediated gut microecological alterations in antineoplastic treatment-induced CV toxicity.
Collapse
Affiliation(s)
- Jing-Yi Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiao-Ya Li
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - De-Xiu Li
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Zi-Hao Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lan-Qing Hu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Chang-Xin Sun
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiao-Nan Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| | - Min Wu
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Long-Tao Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| |
Collapse
|
8
|
Pasam T, Padhy HP, Dandekar MP. Lactobacillus Helveticus Improves Controlled Cortical Impact Injury-Generated Neurological Aberrations by Remodeling of Gut-Brain Axis Mediators. Neurochem Res 2024; 50:3. [PMID: 39541016 DOI: 10.1007/s11064-024-04251-4] [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: 06/04/2024] [Revised: 08/15/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Considerable studies augured the potential of gut microbiota-based interventions in brain injury-associated complications. Based on our earlier study results, we envisaged the sex-specific neuroprotective effect of Lactobacillus helveticus by remodeling of gut-brain axis. In this study, we investigated the effect of L. helveticus on neurological complications in a mouse model of controlled cortical impact (CCI). Adult, male and female, C57BL/6 mice underwent CCI surgery and received L. helveticus treatment for six weeks. Sensorimotor function was evaluated via neurological severity score and rotarod test. Long-term effects on anxiety-like behavior and cognition were assessed using the elevated-zero maze (EZM) and novel object recognition test (NORT). Brain perilesional area, blood, colon, and fecal samples were collected post-CCI for molecular biology analysis. CCI-operated mice displayed significant neurological impairments at 1-, 3-, 5-, and 7-days post-injury (dpi) and exhibited altered behavior in EZM and NORT compared to sham-operated mice. However, these behavioral changes were ameliorated in mice receiving L. helveticus. GFAP, Iba-1, TNF-α, and IL-1β expressions and corticotrophin-releasing hormone (CRH) levels were elevated in the perilesional cortex of CCI-operated male/female mice. These elevated biomarkers and decreased BDNF levels in both male/female mice were modified by L. helveticus treatment. Additionally, L. helveticus treatment restored altered short-chain fatty acids (SCFAs) levels in fecal samples and improved intestinal integrity but did not affect decreased plasma levels of progesterone and testosterone in CCI mice. These results indicate that L. helveticus exerts beneficial effects in the CCI mouse model by mitigating inflammation and remodeling of gut microbiota-brain mediators.
Collapse
Affiliation(s)
- Tulasi Pasam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Hara Prasad Padhy
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Manoj P Dandekar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
9
|
Liu S, Yin J, Wan D, Yin Y. The Role of Iron in Intestinal Mucus: Perspectives from Both the Host and Gut Microbiota. Adv Nutr 2024; 15:100307. [PMID: 39341502 PMCID: PMC11533511 DOI: 10.1016/j.advnut.2024.100307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/10/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024] Open
Abstract
Although research on the role of iron in host immunity has a history spanning decades, it is only relatively recently that attention has been directed toward the biological effects of iron on the intestinal mucus layer, prompted by an evolving understanding of the role of this material in immune defense. The mucus layer, secreted by intestinal goblet cells, covers the intestinal epithelium, and given its unique location, interactions between the host and gut microbiota, as well as among constituent microbiota, occur frequently within the mucus layer. Iron, as an essential nutrient for the vast majority of life forms, regulates immune responses from both the host and microbial perspectives. In this review, we summarize the iron metabolism of both the host and gut microbiota and describe how iron contributes to intestinal mucosal homeostasis via the intestinal mucus layer with respect to both host and constituent gut microbiota. The findings described herein offer a new perspective on iron-mediated intestinal mucosal barrier function.
Collapse
Affiliation(s)
- Shuan Liu
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China
| | - Dan Wan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
10
|
Zheng M, Xie C, Ye D, Chen Y, Wang Z, Wang L, Xiong F, Zhang S, He Q, Wu H, Wu Z, Zhou H, Li L, Xing J, Miao X. Qingzhuan dark tea polysaccharides-zinc alleviates dextran sodium sulfate-induced ulcerative colitis. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:7617-7628. [PMID: 38785267 DOI: 10.1002/jsfa.13597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/27/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Qingzhuan dark tea polysaccharides (QDTP) have been complexed with Zinc (Zn) to form the Qingzhuan dark tea polysaccharides-Zinc (QDTP-Zn) complex. The present study investigated the protective effects of QDTP-Zn on ulcerative colitis (UC) in mice. The UC mouse model was induced using dextran sodium sulfate (DSS), followed by oral administration of QDTP-Zn (0.2 and 0.4 g kg-1 day-1). RESULTS QDTP-Zn demonstrated alleviation of UC symptoms in mice, as evidenced by a decrease in disease activity index scores. QDTP-Zn also regulated colon tissue injury by upregulating ZO-1 and occludin protein expression, at the same time as downregulating tumor necrosis factor-α and interleukin-6β levels. Furthermore, QDTP-Zn induced significant alterations in the abundance of bacteroidetes and firmicutes and notably increased levels of short-chain fatty acids (SCFAs), particularly acetic acid, propionic acid, and butyric acid. CONCLUSION In summary, QDTP-Zn exhibits therapeutic potential in alleviating enteritis by fortifying the colonic mucosal barrier, mitigating inflammation and modulating intestinal microbiota and SCFAs levels. Thus, QDTP-Zn holds promise as a functional food for both the prevention and treatment of UC. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Min Zheng
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- Hubei Industrial Technology Research Institute of Intelligent Health, Xianning, China
| | - Chen Xie
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- Hubei Industrial Technology Research Institute of Intelligent Health, Xianning, China
| | - Dan Ye
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- Hubei Industrial Technology Research Institute of Intelligent Health, Xianning, China
| | - Yong Chen
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- Hubei Industrial Technology Research Institute of Intelligent Health, Xianning, China
| | - Ziyao Wang
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- Hubei Industrial Technology Research Institute of Intelligent Health, Xianning, China
| | - Le Wang
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- Hubei Industrial Technology Research Institute of Intelligent Health, Xianning, China
| | - Fang Xiong
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- Hubei Industrial Technology Research Institute of Intelligent Health, Xianning, China
| | - Sheng Zhang
- Xianning Center for Disease Control and Prevention, Xianning, China
| | - Qiang He
- Xianning Public Inspection Center of Hubei Province, Xianning, China
| | - Hui Wu
- Xianning Public Inspection Center of Hubei Province, Xianning, China
| | - Zhinong Wu
- Xianning Central Hospital, Xianning, China
| | - Hongfu Zhou
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- Hubei Industrial Technology Research Institute of Intelligent Health, Xianning, China
| | - Ling Li
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- Hubei Industrial Technology Research Institute of Intelligent Health, Xianning, China
| | - Jun Xing
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- Hubei Industrial Technology Research Institute of Intelligent Health, Xianning, China
| | - Xiaolei Miao
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
11
|
Zheng ZJ, Zhang HY, Hu YL, Li Y, Wu ZH, Li ZP, Chen DR, Luo Y, Zhang XJ, Li C, Wang XY, Xu D, Qiu W, Li HP, Liao XP, Ren H, Sun J. Sleep Deprivation Induces Gut Damage via Ferroptosis. J Pineal Res 2024; 76:e12987. [PMID: 38975671 DOI: 10.1111/jpi.12987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024]
Abstract
Sleep deprivation (SD) has been associated with a plethora of severe pathophysiological syndromes, including gut damage, which recently has been elucidated as an outcome of the accumulation of reactive oxygen species (ROS). However, the spatiotemporal analysis conducted in this study has intriguingly shown that specific events cause harmful damage to the gut, particularly to goblet cells, before the accumulation of lethal ROS. Transcriptomic and metabolomic analyses have identified significant enrichment of metabolites related to ferroptosis in mice suffering from SD. Further analysis revealed that melatonin could rescue the ferroptotic damage in mice by suppressing lipid peroxidation associated with ALOX15 signaling. ALOX15 knockout protected the mice from the serious damage caused by SD-associated ferroptosis. These findings suggest that melatonin and ferroptosis could be targets to prevent devastating gut damage in animals exposed to SD. To sum up, this study is the first report that proposes a noncanonical modulation in SD-induced gut damage via ferroptosis with a clearly elucidated mechanism and highlights the active role of melatonin as a potential target to maximally sustain the state during SD.
Collapse
Affiliation(s)
- Zi-Jian Zheng
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Hai-Yi Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Ya-Lin Hu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Yan Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Zhi-Hong Wu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Zhi-Peng Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Dong-Rui Chen
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Yang Luo
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Xiao-Jing Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Cang Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Xiao-Yu Wang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Dan Xu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | - Xiao-Ping Liao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Hao Ren
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Jian Sun
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| |
Collapse
|
12
|
Lim C, Dandrieux JRS, Ploeg R, Nowell CJ, Firestone SM, Mansfield CS. Evaluation of Ki-67, goblet cell and MUC2 mucin RNA expression in dogs with lymphoplasmacytic and granulomatous colitis. Vet Immunol Immunopathol 2024; 271:110740. [PMID: 38537313 DOI: 10.1016/j.vetimm.2024.110740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/20/2024] [Accepted: 02/29/2024] [Indexed: 04/29/2024]
Abstract
Intestinal mucus barrier disruption may occur with chronic inflammatory enteropathies. The lack of studies evaluating mucus health in dogs with chronic colitis arises from inherent challenges with assessment of the intestinal mucus layer. It is therefore unknown if reduced goblet cell (GBC) numbers and/or mucin 2 (MUC2) expression, which are responsible for mucus production and secretion, correlate with inflammation severity in dogs with granulomatous colitis (GC) or lymphocytic-plasmacytic colitis (LPC). It is undetermined if Ki-67 immunoreactivity, which has been evaluated in dogs with small intestinal inflammation, similarly correlates to histologic severity in GC and LPC. Study objectives included comparing Ki-67 immunoreactivity, GBC population and MUC2 expression in dogs with GC, LPC and non-inflamed colon; and exploring the use of ribonucleic acid (RNAscope®) in-situ hybridization (ISH) to evaluate MUC2 expression in canine colon. Formalin-fixed endoscopic colonic biopsies were obtained from 48 dogs over an eight-year period. A blinded pathologist reviewed all biopsies. Dogs were classified into the GC (n=19), LPC (n=19) or no colitis (NC) (n=10) group based on final histopathological diagnosis. Ki-67 immunohistochemistry, Alcian-Blue/PAS staining to highlight GBCs, and RNAscope® ISH using customized canine MUC2-targeted probes were performed. At least five microscopic fields per dog were selected to measure Ki-67 labelling index (KI67%), GBC staining percentage (GBC%) and MUC2 expression (MUC2%) using image analysis software. Spearman's correlation coefficients were used to determine associations between World Small Animal Veterinary Association histologic score (WHS) and measured variables. Linear regression models were used to compare relationships between WHS with KI67%, GBC%, and MUC2%; and between GBC% and MUC2%. Median WHS was highest in dogs with GC. Median KI67% normalised to WHS was highest in the NC group (6.69%; range, 1.70-23.60%). Median GBC% did not correlate with colonic inflammation overall. Median MUC2% normalised to WHS in the NC group (10.02%; range, 3.05-39.09%) was two- and three-fold higher than in the GC and LPC groups respectively. With increased colonic inflammation, despite minimal changes in GBC% overall, MUC2 expression markedly declined in the LPC group (-27.4%; 95%-CI, -49.8, 5.9%) and mildly declined in the GC and NC groups. Granulomatous colitis and LPC likely involve different pathways regulating MUC2 expression. Decreased MUC2 gene expression is observed in dogs with chronic colitis compared to dogs without colonic signs. Changes in MUC2 expression appear influenced by GBC activity rather than quantity in GC and LPC.
Collapse
Affiliation(s)
- Chelsea Lim
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Australia.
| | - Julien R S Dandrieux
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Australia; The University of Edinburgh, Royal (Dick) School of Veterinary Studies, United Kingdom
| | - Richard Ploeg
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Australia; CSIRO - Geelong, Australian Centre for Disease Preparedness (ACDP), Australia
| | - Cameron J Nowell
- Monash Institute of Pharmaceutical Sciences, Monash University, Australia
| | - Simon M Firestone
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Australia
| | - Caroline S Mansfield
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Australia; The University of Queensland, School of Veterinary Science, Australia
| |
Collapse
|
13
|
Kassai S, de Vos P. Gastrointestinal barrier function, immunity, and neurocognition: The role of human milk oligosaccharide (hMO) supplementation in infant formula. Compr Rev Food Sci Food Saf 2024; 23:e13271. [PMID: 38284595 DOI: 10.1111/1541-4337.13271] [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: 06/30/2023] [Revised: 10/06/2023] [Accepted: 10/27/2023] [Indexed: 01/30/2024]
Abstract
Breastmilk is seen as the gold standard for infant nutrition as it provides nutrients and compounds that stimulate gut barrier, immune, and brain development to the infant. However, there are many instances where it is not possible for an infant to be fed with breastmilk, especially for the full 6 months recommended by the World Health Organization. In such instances, infant formula is seen as the next best approach. However, infant formulas do not contain human milk oligosaccharides (hMOs), which are uniquely present in human milk as the third most abundant solid component. hMOs have been linked to many health benefits, such as the development of the gut microbiome, the immune system, the intestinal barrier, and a healthy brain. This paper reviews the effects of specific hMOs applied in infant formula on the intestinal barrier, including the not-often-recognized intestinal alkaline phosphatase system that prevents inflammation. Additionally, impact on immunity and the current proof for effects in neurocognitive function and the corresponding mechanisms are discussed. Recent studies suggest that hMOs can alter gut microbiota, modulate intestinal immune barrier function, and promote neurocognitive function. The hMOs 2'-fucosyllactose and lacto-N-neotetraose have been found to have positive effects on the development of infants and have been deemed safe for use in formula. However, their use has been limited due to their cost and complexity of synthesis. Thus, although many benefits have been described, complex hMOs and combinations of hMOs with other oligosaccharides are the best approach to stimulate gut barrier, immune, and brain development and for the prevention of disease.
Collapse
Affiliation(s)
- Sonia Kassai
- Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
14
|
Bai J, Deng S, Zhang X, Dai Z, Ji Y, Zeng S, Ren F, Yang Y, Wu Z. Cinnamaldehyde alleviates zearalenone-induced LS174T cell apoptosis, barrier dysfunction and mucin reduction through JNK/NF-κB signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 263:115276. [PMID: 37499382 DOI: 10.1016/j.ecoenv.2023.115276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/04/2023] [Accepted: 07/17/2023] [Indexed: 07/29/2023]
Abstract
As a natural aldehyde organic compound, cinnamaldehyde (CA) is one of the main components of cinnamon essential oil with multiple bioactivities. In this study, we investigated the protective effects of CA on zearalenone (ZEA)-induced apoptosis, barrier dysfunction and mucin reduction, as well as underlying mechanisms in LS174T cells. In the present study, cells pre-treated with or without CA for 24 h were left untreated or subjected to ZEA for indicated time points Our results showed that 10 μM CA significantly prevented ZEA-induced cell viability decline, reversed ZEA-induced increase of the LDH level, cell cycle disruption and apoptosis in LS174T cells. Periodic acid-schiff (PAS) staining analysis showed that CA significantly alleviated the reduction of mucin secretion in LS174T cells caused by ZEA exposure. Western blot analysis showed that CA significantly reversed ZEA-induced reduction of the expression of mucin 2 (MUC2) and tight junction (TJ) proteins (claudin-1, claudin-3, ZO-1 and ZO-2) in LS174T cells. Notably, CA can significantly reduce the upregulation of the main effector of MAPK and NF-κB signaling pathways in LS174T cells. Further study showed that CA protects cells against ZEA-induced cellular damage through JNK/NF-κB signaling pathway in LS174T cells. Supplementation with CA might be an potential strategy to alleviate the damaging effect of ZEA on epithelial cells.
Collapse
Affiliation(s)
- Jun Bai
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, PR China
| | - Siwei Deng
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, PR China
| | - Xinyu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, PR China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, PR China
| | - Yun Ji
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, PR China
| | - Shenming Zeng
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, PR China
| | - Fazheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, PR China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, PR China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, PR China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, PR China.
| |
Collapse
|
15
|
Inaba R, Vujakovic S, Bergstrom K. The gut mucus network: A dynamic liaison between microbes and the immune system. Semin Immunol 2023; 69:101807. [PMID: 37478802 DOI: 10.1016/j.smim.2023.101807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 06/24/2023] [Accepted: 07/08/2023] [Indexed: 07/23/2023]
Abstract
A complex mucus network made up of large polymers of the mucin-family glycoprotein MUC2 exists between the large intestinal microbial mass and epithelial and immune cells. This has long been understood as an innate immune defense barrier against the microbiota and other luminal threats that reinforces the barrier function of the epithelium and limits microbiota contact with the tissues. However, past and recent studies have provided new evidence of how critical the mucus network is to act as a 'liaison' between host and microbe to mediate anti-inflammatory, mutualistic interactions with the microbiota and protection from pathogens. This review summarizes historical and recent insights into the formation of the gut mucus network, how the microbes and immune system influence mucus, and in turn, how the mucus influences immune responses to the microbiota.
Collapse
Affiliation(s)
- Rain Inaba
- Department of Biology, University of British Columbia, Okanagan Campus, 3187 University Way, Kelowna V1V 1V7, British Columbia, Canada
| | - Sara Vujakovic
- Department of Biology, University of British Columbia, Okanagan Campus, 3187 University Way, Kelowna V1V 1V7, British Columbia, Canada
| | - Kirk Bergstrom
- Department of Biology, University of British Columbia, Okanagan Campus, 3187 University Way, Kelowna V1V 1V7, British Columbia, Canada.
| |
Collapse
|
16
|
Chen T, Tang R, Lin J, Kuo W, Yang I, Liang Y, Lin F. The synthesis and evaluation of thiolated alginate as the barrier to block nutrient absorption on small intestine for body-weight control. Bioeng Transl Med 2023; 8:e10382. [PMID: 37693067 PMCID: PMC10487312 DOI: 10.1002/btm2.10382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/01/2022] [Accepted: 07/21/2022] [Indexed: 11/12/2022] Open
Abstract
Obesity is the most common health concern all over the world. However, till now, there is no promising way to manage obesity or body-weight control. The aim of the study is to develop an edible gel as a health supplement that temporarily attaches to the mucus of the intestines, forming an absorption barrier to block the nutrients. We modify the alginate with the thiol group as thiolated alginate (TA) that may stay on the mucosa layer for a much longer time to reduce nutrient absorption. In this study, the TA is synthesized successfully and proved a good mucosal adhesion to serve as a barrier for nutrient absorption both in vitro and in vivo. The results of in vivo imaging system (IVIS) show that the synthesized TA can be exiled from the gastrointestinal tract within 24 h. The animal study shows that the TA by daily oral administration can effectively reduce body weight and fat deposition. The biosafety is evaluated in vitro at the cellular level, based on ISO-10993, and further checked by animal study. We do believe that the TA could have a greater potential to be developed into a safe health supplement to manage obesity and for body-weight control.
Collapse
Affiliation(s)
- Tzu‐Chien Chen
- Department of Biomedical Engineering, College of Medicine and College of EngineeringNational Taiwan UniversityTaipeiTaiwan
| | - Rui‐Chian Tang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research InstitutesZhunan, Miaoli CountyTaiwan
| | - Jhih‐Ni Lin
- Department of Biomedical Engineering, College of Medicine and College of EngineeringNational Taiwan UniversityTaipeiTaiwan
| | - Wei‐Ting Kuo
- Department of Biomedical Engineering, College of Medicine and College of EngineeringNational Taiwan UniversityTaipeiTaiwan
| | - I‐Hsuan Yang
- Department of Biomedical Engineering, College of Medicine and College of EngineeringNational Taiwan UniversityTaipeiTaiwan
| | - Ya‐Jyun Liang
- Department of Biomedical Engineering, College of Medicine and College of EngineeringNational Taiwan UniversityTaipeiTaiwan
| | - Feng‐Huei Lin
- Department of Biomedical Engineering, College of Medicine and College of EngineeringNational Taiwan UniversityTaipeiTaiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research InstitutesZhunan, Miaoli CountyTaiwan
| |
Collapse
|
17
|
Gorman H, Moreau F, Dufour A, Chadee K. IgGFc-binding protein and MUC2 mucin produced by colonic goblet-like cells spatially interact non-covalently and regulate wound healing. Front Immunol 2023; 14:1211336. [PMID: 37359538 PMCID: PMC10285406 DOI: 10.3389/fimmu.2023.1211336] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
The colonic mucus bilayer is the first line of innate host defense that at the same time houses and nourishes the commensal microbiota. The major components of mucus secreted by goblet cells are MUC2 mucin and the mucus-associated protein, FCGBP (IgGFc-binding protein). In this study, we determine if FCGBP and MUC2 mucin were biosynthesized and interacted together to spatially enhance the structural integrity of secreted mucus and its role in epithelial barrier function. MUC2 and FCGBP were coordinately regulated temporally in goblet-like cells and in response to a mucus secretagogue but not in CRISPR-Cas9 gene-edited MUC2 KO cells. Whereas ~85% of MUC2 was colocalized with FCGBP in mucin granules, ~50% of FCGBP was diffusely distributed in the cytoplasm of goblet-like cells. STRING-db v11 analysis of the mucin granule proteome revealed no protein-protein interaction between MUC2 and FCGBP. However, FCGBP interacted with other mucus-associated proteins. FCGBP and MUC2 interacted via N-linked glycans and were non-covalently bound in secreted mucus with cleaved low molecular weight FCGBP fragments. In MUC2 KO, cytoplasmic FCGBP was significantly increased and diffusely distributed in wounded cells that healed by enhanced proliferation and migration within 2 days, whereas, in WT cells, MUC2 and FCGBP were highly polarized at the wound margin which impeded wound closure by 6 days. In DSS colitis, restitution and healed lesions in Muc2+/+ but not Muc2-/- littermates, were accompanied by a rapid increase in Fcgbp mRNA and delayed protein expression at 12- and 15-days post DSS, implicating a potential novel endogenous protective role for FCGBP in wound healing to maintain epithelial barrier function.
Collapse
Affiliation(s)
- Hayley Gorman
- Department of Microbiology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - France Moreau
- Department of Microbiology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Antoine Dufour
- Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Kris Chadee
- Department of Microbiology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
18
|
Liu Y, Yu Z, Zhu L, Ma S, Luo Y, Liang H, Liu Q, Chen J, Guli S, Chen X. Orchestration of MUC2 - The key regulatory target of gut barrier and homeostasis: A review. Int J Biol Macromol 2023; 236:123862. [PMID: 36870625 DOI: 10.1016/j.ijbiomac.2023.123862] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
The gut mucosa of human is covered by mucus, functioning as a crucial defense line for the intestine against external stimuli and pathogens. Mucin2 (MUC2) is a subtype of secretory mucins generated by goblet cells and is the major macromolecular component of mucus. Currently, there is an increasing interest on the investigations of MUC2, noting that its function is far beyond a maintainer of the mucus barrier. Moreover, numerous gut diseases are associated with dysregulated MUC2 production. Appropriate production level of MUC2 and mucus contributes to gut barrier function and homeostasis. The production of MUC2 is regulated by a series of physiological processes, which are orchestrated by various bioactive molecules, signaling pathways and gut microbiota, etc., forming a complex regulatory network. Incorporating the latest findings, this review provided a comprehensive summary of MUC2, including its structure, significance and secretory process. Furthermore, we also summarized the molecular mechanisms of the regulation of MUC2 production aiming to provide developmental directions for future researches on MUC2, which can act as a potential prognostic indicator and targeted therapeutic manipulation for diseases. Collectively, we elucidated the micro-level mechanisms underlying MUC2-related phenotypes, hoping to offer some constructive guidance for intestinal and overall health of mankind.
Collapse
Affiliation(s)
- Yaxin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Zihan Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Lanping Zhu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Shuang Ma
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Yang Luo
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Huixi Liang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Qinlingfei Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Jihua Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Sitan Guli
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China.
| |
Collapse
|
19
|
Hussein EM, Muhammad MAA, Hussein AM, Elzagawy SM, Zaki WM, Temsah AG, Badr MS, Alabbassy MM. Levels of Genetic Variants Among Symptomatic Blastocystis Subtypes and their Relationship to Mucosal Immune Surveillance in the Precancerous Colons of Experimentally Infected Rats. Acta Parasitol 2023; 68:70-83. [PMID: 36380160 PMCID: PMC10011339 DOI: 10.1007/s11686-022-00628-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE The relationship between the genetic diversity of Blastocystis and immune surveillance in precancerous colons with blastocystosis is still under investigation. This study aimed to identify the genetic Blastocystis variants among 54 symptomatic human isolates and their relationship to mucosal immune surveillance in the precancerous polyps of experimentally infected rats. METHODS Polymerase chain reaction and high-resolution melting (PCR/HRM) curves discriminated human symptomatic Blastocystis isolates into subtypes (STs)/intrasubtypes, which were orally administered to rats to induce experimental infection. Then, the mucosal immune responses of the infected colons were evaluated in relation to polyp formation through immunostaining to identify mucus MUC2 and determine mucosal immune cell (goblet, lymphocyte and mast) counts, secretory IgA levels and parasitic intestinal invasion. RESULTS ST1, ST3, and ST4 were found in 18.5% (10/54), 54.7% (29/54), and 27.8% (15/54) of the samples, respectively. Then, the HRM curve discriminated ST3 into the wild, mutant, and heterozygous [17/54 (31.5%), 5/54 (9.3%), and 7/54 (12.9%)] intrasubtypes. ST1 and ST4 had no genetic variations. Precancerous polyps were detected in the colons of 40.5% of the infected rats. ST1 constituted 14.7% of these cases, while the wild, mutant, and heterozygous intrasubtypes of ST3 showed polyps in 12.9%, 5.5%, and 5.5% of cases, respectively. Only 1.9% of the polyps were related to ST4. MUC2 showed weak immunostaining in 44.5% of the infected colons, and 38.9% were polyp inducers. Low goblet cell numbers and high interepithelial lymphocyte counts were significantly associated with polyp formation, particularly with ST1 and wild ST3. Among the polyp inducers, high numbers of mast cells were detected in wild ST3 and ST4, while a low number was found with heterozygous ST3. The level of secretory IgA was low in polyp-inducing STs. Most of the results were statistically significant. CONCLUSION Immunosurveillance showed a potential relationship between ST1 and the ST3 intrasubtypes and precancerous polyps. This relationship may provide insight into the prevention and/or development of new immunotherapeutic strategies to combat colorectal cancer.
Collapse
Affiliation(s)
- Eman M Hussein
- Medical Parasitology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| | - Muhammad A A Muhammad
- Pathology Department, Faculty of Medicine, Suez Canal, University, Ismailia, 41522, Egypt
| | - Abdalla M Hussein
- Bio-Physics Department, Faculty of Science, Al-Azhar University, Cairo, 11652, Egypt
| | - Sherine M Elzagawy
- Medical Parasitology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Wafaa M Zaki
- Medical Parasitology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Ashraf G Temsah
- Medical Parasitology Department, Faculty of Medicine, Damietta Branch, AL Azhar University, Damietta, Egypt
| | - Mohamed S Badr
- Medical Genetic Centre, Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Maha M Alabbassy
- Medical Parasitology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
20
|
Chen H, Sun HM, Wu B, Sun TY, Han LZ, Wang G, Shang YF, Yang S, Zhou DS. Artesunate delays the dysfunction of age-related intestinal epithelial barrier by mitigating endoplasmic reticulum stress/unfolded protein response. Mech Ageing Dev 2023; 210:111760. [PMID: 36476344 DOI: 10.1016/j.mad.2022.111760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
The impairment of the intestinal epithelial barrier and subsequent bacterial translocation are common in aging individuals, contributory to several local and systematic disorders. However, the underlying mechanism of the age-related degeneration has not been fully understood. In this study, we demonstrated that the intestinal KIT signaling declined and de-activated with aging, parallel with epithelial barrier dysfunction. Endoplasmic reticulum stress (ERS)/unfolded protein response (UPR) was obviously increased during aging. The ERS and its downstream IRE1α were highly activated in the aging colonic epithelium. Furthermore, by the use of Tunicamycin (Tm)-induced ERS mouse and cell models, we uncovered that the activity of the ERS/IRE1α accelerated the protein degradation of KIT via ubiquitin-proteasome pathway. The deficiency of KIT signaling further reduced the transcription of the tight junction protein Claudin-3. Of significance, Artesunate (ART) could be capable of ameliorating the detrimental effect of ERS/IRE1α, indicated by the re-gained KIT and Claudin-3 expressions and the restoration of the intestinal epithelial barrier. In conclusion, our present study provided novel evidence elucidating the ERS/IRE1α-induced loss of KIT and Claudin-3 in the aging colonic epithelium and also shed light on the protective effect of Artesunate on the intestinal epithelial barrier by blocking ERS/IRE1α activity during aging.
Collapse
Affiliation(s)
- Hong Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Hai-Mei Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing 100069, China
| | - Bo Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing 100069, China
| | - Ting-Yi Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing 100069, China
| | - Li-Zhuang Han
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Guan Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yun-Fei Shang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Shu Yang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing 100069, China.
| | - De-Shan Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing 100069, China.
| |
Collapse
|
21
|
Kiparissi F, Dastamani A, Palm L, Azabdaftari A, Campos L, Gaynor E, Grünewald S, Uhlig HH, Kleta R, Böckenhauer D, Jones KDJ. Phosphomannomutase 2 (PMM2) variants leading to hyperinsulinism-polycystic kidney disease are associated with early-onset inflammatory bowel disease and gastric antral foveolar hyperplasia. Hum Genet 2023; 142:697-704. [PMID: 36773065 PMCID: PMC10181953 DOI: 10.1007/s00439-023-02523-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 01/19/2023] [Indexed: 02/12/2023]
Abstract
Phosphomannomutase 2 (PMM2) deficiency causes Congenital Disorder of Glycosylation (PMM2-CDG), but does not have a recognised association with Inflammatory Bowel Disease (IBD). A distinct clinical syndrome of hyperinsulinism and autosomal recessive polycystic kidney disease (HIPKD) arises in the context of a specific variant in the PMM2 promotor, either in homozygosity, or compound heterozygous with a deleterious PMM2 variant. Here, we describe the development of IBD in three patients with PMM2-HIPKD, with onset of IBD at 0, 6, and 10 years of age. In each case, intestinal inflammation coincided with the unusual finding of gastric antral foveolar hyperplasia. IBD disease was of variable severity at onset but well controlled with conventional and first-line biologic treatment approaches. The organ-level pattern of disease manifestations in PMM2-HIPKD-IBD may reflect a loss of cis-acting regulatory control by hepatocyte nuclear factor 4 alpha (HNF4A). Analysis of published transcriptomic data suggests that IBD most likely arises due to an impact on epithelial cellular function. We identify a specific pattern of variation in PMM2 as a novel association of early-onset IBD with distinctive gastric pathology.
Collapse
Affiliation(s)
- Fevronia Kiparissi
- Department of Paediatric Gastroenterology & Nutrition, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Antonia Dastamani
- Department of Paediatric Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Liina Palm
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Aline Azabdaftari
- Translational Gastroenterology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Luis Campos
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Edward Gaynor
- Department of Paediatric Gastroenterology & Nutrition, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Stephanie Grünewald
- Department of Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Holm H Uhlig
- Translational Gastroenterology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.,Department of Paediatrics and Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Robert Kleta
- Department of Renal Medicine, University College London, London, UK.,Department of Paediatric Nephrology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Detlef Böckenhauer
- Department of Renal Medicine, University College London, London, UK.,Department of Paediatric Nephrology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Kelsey D J Jones
- Department of Paediatric Gastroenterology & Nutrition, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK. .,The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
22
|
Song S, Tu D, Meng C, Liu J, Wilson B, Wang Q, Shih YYI, Gao HM, Hong JS. Dysfunction of the noradrenergic system drives inflammation, α-synucleinopathy, and neuronal loss in mouse colon. Front Immunol 2023; 14:1083513. [PMID: 36845109 PMCID: PMC9950510 DOI: 10.3389/fimmu.2023.1083513] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
Clinical and pathological evidence revealed that α-synuclein (α-syn) pathology seen in PD patients starts in the gut and spreads via anatomically connected structures from the gut to the brain. Our previous study demonstrated that depletion of central norepinephrine (NE) disrupted brain immune homeostasis, producing a spatiotemporal order of neurodegeneration in the mouse brain. The purpose of this study was 1) to determine the role of peripheral noradrenergic system in the maintenance of gut immune homeostasis and in the pathogenesis of PD and 2) to investigate whether NE-depletion induced PD-like α-syn pathological changes starts from the gut. For these purposes, we investigated time-dependent changes of α-synucleinopathy and neuronal loss in the gut following a single injection of DSP-4 (a selective noradrenergic neurotoxin) to A53T-SNCA (human mutant α-syn) over-expression mice. We found DPS-4 significantly reduced the tissue level of NE and increased immune activities in gut, characterized by increased number of phagocytes and proinflammatory gene expression. Furthermore, a rapid-onset of α-syn pathology was observed in enteric neurons after 2 weeks and delayed dopaminergic neurodegeneration in the substantia nigra was detected after 3-5 months, associated with the appearance of constipation and impaired motor function, respectively. The increased α-syn pathology was only observed in large, but not in the small, intestine, which is similar to what was observed in PD patients. Mechanistic studies reveal that DSP-4-elicited upregulation of NADPH oxidase (NOX2) initially occurred only in immune cells during the acute intestinal inflammation stage, and then spread to enteric neurons and mucosal epithelial cells during the chronic inflammation stage. The upregulation of neuronal NOX2 correlated well with the extent of α-syn aggregation and subsequent enteric neuronal loss, suggesting that NOX2-generated reactive oxygen species play a key role in α-synucleinopathy. Moreover, inhibiting NOX2 by diphenyleneiodonium or restoring NE function by salmeterol (a β2-receptor agonist) significantly attenuated colon inflammation, α-syn aggregation/propagation, and enteric neurodegeneration in the colon and ameliorated subsequent behavioral deficits. Taken together, our model of PD shows a progressive pattern of pathological changes from the gut to the brain and suggests a potential role of the noradrenergic dysfunction in the pathogenesis of PD.
Collapse
Affiliation(s)
- Sheng Song
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
- Biomedical Research Imaging Center, University of North Caroline at Chapel Hill, Chapel Hill, NC, United States
| | - Dezhen Tu
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Institute for Brain Sciences, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of medicine, Nanjing University, Nanjing, China
| | - Chengbo Meng
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Jie Liu
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Belinda Wilson
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Qingshan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Yen-Yu Ian Shih
- Biomedical Research Imaging Center, University of North Caroline at Chapel Hill, Chapel Hill, NC, United States
| | - Hui-Ming Gao
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Institute for Brain Sciences, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of medicine, Nanjing University, Nanjing, China
| | - Jau-Shyong Hong
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| |
Collapse
|
23
|
Fernández-Lainez C, Aan de Stegge M, Silva-Lagos LA, López-Velázquez G, de Vos P. β(2 → 1)-β(2 → 6) branched graminan-type fructans and β(2 → 1) linear fructans impact mucus-related and endoplasmic reticulum stress-related genes in goblet cells and attenuate inflammatory responses in a fructan dependent fashion. Food Funct 2023; 14:1338-1348. [PMID: 36656019 DOI: 10.1039/d2fo02710k] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Dietary fibers such as fructans have beneficial effects on intestinal health but it is unknown whether they impact goblet cells (GCs). Here we studied the effects of inulin-type fructans (ITFs) and graminan-type fructans (GTFs) with different molecular weights on mucus- and endoplasmic reticulum (ER) stress-related genes in intestinal GCs. To that end, GCs were incubated in the presence of ITFs or GTFs, or ITFs and GTFs + TNFα or the N-glycosylation inhibitor tunicamycin (Tm). IL-8 production by GCs was studied as a marker of inflammation. Effects between ITFs and GTFs were compared. We found a beneficial impact of GTFs especially on the expression of RETNLB. GTF II protects from the TNFα-induced gene expression dysregulation of MUC2, TFF3, GAL3ST2, and CHST5. Also, all the studied fructans prevented Tm-induced dysregulation of GAL3ST2. Interestingly, only the short chain fructans ITF I and GTF I have anti-inflammatory properties on GCs. All the studied fructans except ITF I decreased the expression of the ER stress-related HSPA5 and XBP1. All these benefits were fructan-structure and chain length dependent. Our study contributes to a better understanding of chemical structure-dependent beneficial effects of ITFs and GTFs on gut barrier function, which could contribute to prevention of gut inflammatory disorders.
Collapse
Affiliation(s)
- Cynthia Fernández-Lainez
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands. .,Laboratorio de Errores innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Av. Iman 1, 04530, Ciudad de México, Mexico.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México. Edificio D, 1° Piso. Circuito de Posgrados, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Myrthe Aan de Stegge
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | - Luis Alfredo Silva-Lagos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | - Gabriel López-Velázquez
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, Av. Iman 1, 04530, Cuidad de México, Mexico.
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| |
Collapse
|
24
|
Sun B, Huang Y, Castro LFC, Yang S, Huang S, Jin W, Zhou H, Ijiri S, Luo Y, Gao J, Cao X. The chromosome-level genome and key genes associated with mud-dwelling behavior and adaptations of hypoxia and noxious environments in loach (Misgurnus anguillicaudatus). BMC Biol 2023; 21:18. [PMID: 36726103 PMCID: PMC9893644 DOI: 10.1186/s12915-023-01517-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 01/18/2023] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The loach (Misgurnus anguillicaudatus), the most widely distributed species of the family Cobitidae, displays a mud-dwelling behavior and intestinal air-breathing, inhabiting the muddy bottom of extensive freshwater habitats. However, lack of high-quality reference genome seriously limits the interpretation of the genetic basis of specialized adaptations of the loach to the adverse environments including but not limited to the extreme water temperature, hypoxic and noxious mud environment. RESULTS This study generated a 1.10-Gb high-quality, chromosome-anchored genome assembly, with a contig N50 of 3.83 Mb. Multiple comparative genomic analyses found that proto-oncogene c-Fos (fos), a regulator of bone development, is positively selected in loach. Knockout of fos (ID: Mis0086400.1) led to severe osteopetrosis and movement difficulties, combined with the comparison results of bone mineral density, supporting the hypothesis that fos is associated with loach mud-dwelling behavior. Based on genomic and transcriptomic analysis, we identified two key elements involved in the intestinal air-breathing of loach: a novel gene (ID: mis0158000.1) and heat shock protein beta-1 (hspb1). The flavin-containing monooxygenase 5 (fmo5) genes, central to xenobiotic metabolism, undergone expansion in loach and were identified as differentially expressed genes in a drug stress trial. A fmo5-/- (ID: Mis0185930.1) loach displayed liver and intestine injury, indicating the importance of this gene to the adaptation of the loach to the noxious mud. CONCLUSIONS Our work provides valuable insights into the genetic basis of biological adaptation to adverse environments.
Collapse
Affiliation(s)
- Bing Sun
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Postal address: No.1 Shizishan Stress, Hongshan District, Wuhan, 430070, Hubei Province, China
| | - Yuwei Huang
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Postal address: No.1 Shizishan Stress, Hongshan District, Wuhan, 430070, Hubei Province, China
| | - L Filipe C Castro
- Interdisciplinary Centre of Marine and Environmental Research of the University of Porto, 4450-208, Matosinhos, Portugal
- Department of Biology, University of Porto, 4450-208, Porto, Portugal
| | - Su Yang
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Postal address: No.1 Shizishan Stress, Hongshan District, Wuhan, 430070, Hubei Province, China
| | - Songqian Huang
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Bunkyo, Tokyo, 113-8657, Japan
| | - Wu Jin
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, Jiangsu, China
| | - He Zhou
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, 116023, China
| | - Shigeho Ijiri
- Division of Marine Life Sciences, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Hokkaido, 041-8611, Japan
| | - Yi Luo
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Postal address: No.1 Shizishan Stress, Hongshan District, Wuhan, 430070, Hubei Province, China
| | - Jian Gao
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Postal address: No.1 Shizishan Stress, Hongshan District, Wuhan, 430070, Hubei Province, China.
| | - Xiaojuan Cao
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Postal address: No.1 Shizishan Stress, Hongshan District, Wuhan, 430070, Hubei Province, China.
| |
Collapse
|
25
|
ERdj5 protects goblet cells from endoplasmic reticulum stress-mediated apoptosis under inflammatory conditions. Exp Mol Med 2023; 55:401-412. [PMID: 36759578 PMCID: PMC9981579 DOI: 10.1038/s12276-023-00945-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/21/2022] [Accepted: 12/06/2022] [Indexed: 02/11/2023] Open
Abstract
Endoplasmic reticulum stress is closely associated with the onset and progression of inflammatory bowel disease. ERdj5 is an endoplasmic reticulum-resident protein disulfide reductase that mediates the cleavage and degradation of misfolded proteins. Although ERdj5 expression is significantly higher in the colonic tissues of patients with inflammatory bowel disease than in healthy controls, its role in inflammatory bowel disease has not yet been reported. In the current study, we used ERdj5-knockout mice to investigate the potential roles of ERdj5 in inflammatory bowel disease. ERdj5 deficiency causes severe inflammation in mouse colitis models and weakens gut barrier function by increasing NF-κB-mediated inflammation. ERdj5 may not be indispensable for goblet cell function under steady-state conditions, but its deficiency induces goblet cell apoptosis under inflammatory conditions. Treatment of ERdj5-knockout mice with the chemical chaperone ursodeoxycholic acid ameliorated severe colitis by reducing endoplasmic reticulum stress. These findings highlight the important role of ERdj5 in preserving goblet cell viability and function by resolving endoplasmic reticulum stress.
Collapse
|
26
|
Cari L, Rosati L, Leoncini G, Lusenti E, Gentili M, Nocentini G, Riccardi C, Migliorati G, Ronchetti S. Association of GILZ with MUC2, TLR2, and TLR4 in Inflammatory Bowel Disease. Int J Mol Sci 2023; 24:ijms24032235. [PMID: 36768553 PMCID: PMC9917296 DOI: 10.3390/ijms24032235] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Ulcerative colitis (UC) and Crohn's Disease (CD) are chronic relapsing inflammatory diseases that are caused by genetic, environmental, and immune factors. Treatment strategies are currently based on symptomatic control by immunosuppression. The glucocorticoid-induced leucine zipper (GILZ), a mediator of several effects of glucocorticoids, was recently found to be secreted by goblet cells and play a role in inflammatory bowel disease (IBD). This study investigates which genes GILZ is associated with in its role in intestinal barrier functions. We examined datasets from the Gene Expression Omnibus (GEO) and ArrayExpress profiles of the gut of healthy subjects (HSs), as well as UC and CD patients. The human colonic epithelial HT29 cell line was used for in vitro validation experiments. GILZ was significantly correlated with MUC2, TLR2, and TLR4. In particular, an inverse correlation was found between the GILZ and MUC2 in HS and patients with IBD, mostly in those with an active disease. Further, direct pairwise correlations for GILZ/TLR2 and GILZ/TLR4 were found in HSs and UC patients, but not in CD patients. Overall, our results reveal the crosstalk at the transcription level between the GILZ, MUC2, and TLRs in the mucosal barrier through common pathways, and they open up new perspectives in terms of mucosal healing in IBD patients.
Collapse
Affiliation(s)
- Luigi Cari
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, 06132 Perugia, Italy
| | - Lucrezia Rosati
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, 06132 Perugia, Italy
| | - Giuseppe Leoncini
- First Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milano, Italy
| | - Eleonora Lusenti
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, 06132 Perugia, Italy
| | - Marco Gentili
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, 06132 Perugia, Italy
| | - Giuseppe Nocentini
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, 06132 Perugia, Italy
| | - Carlo Riccardi
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, 06132 Perugia, Italy
| | - Graziella Migliorati
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, 06132 Perugia, Italy
| | - Simona Ronchetti
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, 06132 Perugia, Italy
- Correspondence:
| |
Collapse
|
27
|
Tang Y, Zhou X, Cao T, Chen E, Li Y, Lei W, Hu Y, He B, Liu S. Endoplasmic Reticulum Stress and Oxidative Stress in Inflammatory Diseases. DNA Cell Biol 2022; 41:924-934. [PMID: 36356165 DOI: 10.1089/dna.2022.0353] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Yun Tang
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiangping Zhou
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ting Cao
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - En Chen
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yumeng Li
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wenbo Lei
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yibao Hu
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Bisha He
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shuangquan Liu
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
28
|
Tao Y, Qiao S, Lv C, Yun X, Yue M, Fang Y, Wei Z, Dai Y, Xia Y. Phytoestrogen arctigenin preserves the mucus barrier in inflammatory bowel diseases by inhibiting goblet cell apoptosis via the
ERβ
/
TRIM21
/
PHB1
pathway. Phytother Res 2022; 36:3248-3264. [DOI: 10.1002/ptr.7495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 01/17/2023]
Affiliation(s)
- Yu Tao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
- Jiangsu Province Hospital of Chinese Medicine The Affiliated Hospital of Nanjing University of Chinese Medicine Nanjing China
| | - Si‐miao Qiao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Chang‐jun Lv
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Xin‐ming Yun
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Meng‐fan Yue
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Yu‐lai Fang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Zhi‐feng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Yu‐feng Xia
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| |
Collapse
|
29
|
Livzan MA, Bicbavova GR, Romanyuk AE. Ulcerative colitis: focus on colonic mucosal resistance. BULLETIN OF SIBERIAN MEDICINE 2022; 21:121-132. [DOI: 10.20538/1682-0363-2022-1-121-132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
In recent decades, following cooperation between scientists in various specialties, new unique data on the pathogenesis of ulcerative colitis have been obtained. The role of an impaired immune response to antigens of gut microbiota in genetically predisposed individuals under the effect of certain environmental factors was proven. Assessing the interaction between the colonic mucosa and gut microbiota will help to understand the mechanisms of ulcerative colitis and develop new treatment strategies for the disease.This review presents modern views on the pathogenesis of ulcerative colitis with a focus on the imbalance between local protective and aggressive factors of the gastric and intestinal mucosa. The structure and role of the epithelial barrier both under normal conditions and in ulcerative colitis are considered in detail.The aim of this review was to summarize the data on resistance of the colonic mucosa and its damage in ulcerative colitis.
Collapse
|
30
|
Liang X, Xie J, Liu H, Zhao R, Zhang W, Wang H, Pan H, Zhou Y, Han W. STIM1 Deficiency In Intestinal Epithelium Attenuates Colonic Inflammation and Tumorigenesis by Reducing ER Stress of Goblet Cells. Cell Mol Gastroenterol Hepatol 2022; 14:193-217. [PMID: 35367664 PMCID: PMC9130113 DOI: 10.1016/j.jcmgh.2022.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS As an indispensable component of store-operated Ca2+ entry, stromal interaction molecule 1 (STIM1) is known to promote colorectal cancer and T-cell-mediated inflammatory diseases. However, whether the intestinal mucosal STIM1 is involved in inflammatory bowel diseases (IBDs) is unclear. This study aimed to investigate the role of intestinal epithelial STIM1 in IBD. METHODS Inflammatory and matched normal intestinal tissues were collected from IBD patients to investigate the expression of STIM1. Intestinal epithelium-specific STIM1 conditional knockout mice (STIM1ΔIEC) were generated and induced to develop colitis and colitis-associated colorectal cancer. The mucosal barrier, including the epithelial barrier and mucus barrier, was analyzed. The mechanisms by which STIM1 regulate goblet cell endoplasmic reticulum stress and apoptosis were assessed. RESULTS STIM1 could regulate intestinal epithelial homeostasis. STIM1 was augmented in the inflammatory intestinal tissues of IBD patients. In dextran sodium sulfate-induced colitis, STIM1 deficiency in intestinal epithelium reduced the loss of goblet cells through alleviating endoplasmic reticulum stress induced by disturbed Ca2+ homeostasis, resulting in the maintenance of the integrated mucus layer. These effects prevented commensal bacteria from contacting and stimulating the intestinal epithelium of STIM1ΔIEC mice and thereby rendered STIM1ΔIEC mice less susceptible to colitis and colitis-associated colorectal cancer. In addition, microbial diversity in dextran sodium sulfate-treated STIM1ΔIEC mice slightly shifted to an advantageous bacteria, which further protected the intestinal epithelium. CONCLUSIONS Our results establish STIM1 as a crucial regulator for the maintenance of the intestinal barrier during colitis and provide a potential target for IBD treatment.
Collapse
Affiliation(s)
- Xiaojing Liang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Jiansheng Xie
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Hao Liu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Rongjie Zhao
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Wei Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Haidong Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China,Correspondence Address correspondence to: Weidong Han, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qingchun Road, Hangzhou, Zhejiang 310016, China; fax: 86-571-86436673.
| |
Collapse
|
31
|
Wanner N, Barnhart J, Apostolakis N, Zlojutro V, Asosingh K. Using the Autofluorescence Finder on the Sony ID7000 TM Spectral Cell Analyzer to Identify and Unmix Multiple Highly Autofluorescent Murine Lung Populations. Front Bioeng Biotechnol 2022; 10:827987. [PMID: 35372303 PMCID: PMC8965042 DOI: 10.3389/fbioe.2022.827987] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Autofluorescence (AF) is a feature of all cell types, though some have more than others. In tissues with complex heterogeneous cellularity, AF is frequently a source of high background, masking faint fluorescent signals and reducing the available dynamic range of detectors for detecting fluorescence signals from markers of interest in a flow cytometry panel. Pulmonary flow cytometry presents unique challenges because lung cells are heterogeneous and contain varying amounts of high AF. The goal of this study was to demonstrate how a novel AF Finder tool on the Sony ID7000™ Spectral Cell Analyzer can be used to identify and screen multiple AF subsets in complex highly AF tissues like murine lungs. In lung single cell suspensions, the AF Finder tool identified four distinct AF spectra from six highly AF subsets. The subtraction of these distinct AF spectra resulted in a resolution increase by several log decades in several fluorescent channels. The major immune and lung tissue resident cells in a murine model of asthma were easily identified in a multi-color panel using AF subtraction. The findings demonstrate the practicality of the AF Finder tool, particularly when analyzing samples with multiple AF populations of varying intensities, in order to reduce fluorescence background and increase signal resolution in spectral flow cytometry.
Collapse
Affiliation(s)
- Nicholas Wanner
- Asosingh Lab, Department of Inflammation and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States
| | | | - Nicholas Apostolakis
- Asosingh Lab, Department of Inflammation and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States
| | - Violetta Zlojutro
- Asosingh Lab, Department of Inflammation and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States
| | - Kewal Asosingh
- Asosingh Lab, Department of Inflammation and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States
- Flow Cytometry Core, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States
| |
Collapse
|
32
|
Al Saedi A, Sharma S, Bani Hassan E, Chen L, Ghasem-Zadeh A, Hassanzadeganroudsari M, Gooi JH, Stavely R, Eri R, Miao D, Nurgali K, Duque G. Characterization of Skeletal Phenotype and Associated Mechanisms With Chronic Intestinal Inflammation in the Winnie Mouse Model of Spontaneous Chronic Colitis. Inflamm Bowel Dis 2022; 28:259-272. [PMID: 34347076 DOI: 10.1093/ibd/izab174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Osteoporosis is a common extraintestinal manifestation of inflammatory bowel disease (IBD). However, studies have been scarce, mainly because of the lack of an appropriate animal model of colitis-associated bone loss. In this study, we aimed to decipher skeletal manifestations in the Winnie mouse model of spontaneous chronic colitis, which carries a MUC2 gene mutation and closely replicates ulcerative colitis. In our study, Winnie mice, prior to the colitis onset at 6 weeks old and progression at 14 and 24 weeks old, were compared with age-matched C57BL/6 controls. We studied several possible mechanisms involved in colitis-associated bone loss. METHODS We assessed for bone quality (eg, microcomputed tomography [micro-CT], static and dynamic histomorphometry, 3-point bending, and ex vivo bone marrow analysis) and associated mechanisms (eg, electrochemical recordings for gut-derived serotonin levels, real-time polymerase chain reaction [qRT-PCR], double immunofluorescence microscopy, intestinal inflammation levels by lipocalin-2 assay, serum levels of calcium, phosphorus, and vitamin D) from Winnie (6-24 weeks) and age-matched C57BL6 mice. RESULTS Deterioration in trabecular and cortical bone microarchitecture, reductions in bone formation, mineral apposition rate, bone volume/total volume, osteoid volume/bone surface, and bone strength were observed in Winnie mice compared with controls. Decreased osteoblast and increased osteoclast numbers were prominent in Winnie mice compared with controls. Upregulation of 5-HTR1B gene and increased association of FOXO1 with ATF4 complex were identified as associated mechanisms concomitant to overt inflammation and high levels of gut-derived serotonin in 14-week and 24-week Winnie mice. CONCLUSIONS Skeletal phenotype of the Winnie mouse model of spontaneous chronic colitis closely represents manifestations of IBD-associated osteoporosis/osteopenia. The onset and progression of intestinal inflammation are associated with increased gut-derived serotonin level, increased bone resorption, and decreased bone formation.
Collapse
Affiliation(s)
- Ahmed Al Saedi
- Department of Medicine-Western Health, The University of Melbourne, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Department of Medicine-Western Health, Melbourne, VIC, Australia
| | - Shilpa Sharma
- Department of Medicine-Western Health, The University of Melbourne, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Department of Medicine-Western Health, Melbourne, VIC, Australia
| | - Ebrahim Bani Hassan
- Department of Medicine-Western Health, The University of Melbourne, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Department of Medicine-Western Health, Melbourne, VIC, Australia
| | - Lulu Chen
- Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Ali Ghasem-Zadeh
- Australian Institute for Musculoskeletal Science (AIMSS), Department of Medicine-Western Health, Melbourne, VIC, Australia
- Departments of Medicine and Endocrinology, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | | | - Jonathan H Gooi
- St. Vincent's Institute of Medical Research, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Rhian Stavely
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rajaraman Eri
- School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Dengshun Miao
- Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- Calcium Research Laboratory, McGill University Health Centre and Department of Medicine, McGill University, Montreal, QC, Canada
| | - Kulmira Nurgali
- Department of Medicine-Western Health, The University of Melbourne, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Department of Medicine-Western Health, Melbourne, VIC, Australia
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Gustavo Duque
- Department of Medicine-Western Health, The University of Melbourne, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Department of Medicine-Western Health, Melbourne, VIC, Australia
| |
Collapse
|
33
|
Zhu L, Miao B, Dymerska D, Kuswik M, Bueno-Martínez E, Sanoguera-Miralles L, Velasco EA, Paramasivam N, Schlesner M, Kumar A, Yuan Y, Lubinski J, Bandapalli OR, Hemminki K, Försti A. Germline Variants of CYBA and TRPM4 Predispose to Familial Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14030670. [PMID: 35158942 PMCID: PMC8833488 DOI: 10.3390/cancers14030670] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/17/2022] [Accepted: 01/26/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Whole-genome sequencing and bioinformatics analysis on unique colorectal cancer families revealed two attractive candidate predisposition genes, CYBA and TRPM4, each with a loss-of-function variant. Supported by our functional studies, we suggest that the two gene defects mechanistically involve intestinal barrier integrity through reactive oxygen species and mucus biology, which converges in chronic bowel inflammation, a known risk factor for colorectal cancer. Abstract Familial colorectal cancer (CRC) is only partially explained by known germline predisposing genes. We performed whole-genome sequencing in 15 Polish families of many affected individuals, without mutations in known CRC predisposing genes. We focused on loss-of-function variants and functionally characterized them. We identified a frameshift variant in the CYBA gene (c.246delC) in one family and a splice site variant in the TRPM4 gene (c.25–1 G > T) in another family. While both variants were absent or extremely rare in gene variant databases, we identified four additional Polish familial CRC cases and two healthy elderly individuals with the CYBA variant (odds ratio 2.46, 95% confidence interval 0.48–12.69). Both variants led to a premature stop codon and to a truncated protein. Functional characterization of the variants showed that knockdown of CYBA or TRPM4 depressed generation of reactive oxygen species (ROS) in LS174T and HT-29 cell lines. Knockdown of TRPM4 resulted in decreased MUC2 protein production. CYBA encodes a component in the NADPH oxidase system which generates ROS and controls, e.g., bacterial colonization in the gut. Germline CYBA variants are associated with early onset inflammatory bowel disease, supported with experimental evidence on loss of intestinal mucus barrier function due to ROS deficiency. TRPM4 encodes a calcium-activated ion channel, which, in a human colonic cancer cell line, controls calcium-mediated secretion of MUC2, a major component of intestinal mucus barrier. We suggest that the gene defects in CYBA and TRPM4 mechanistically involve intestinal barrier integrity through ROS and mucus biology, which converges in chronic bowel inflammation.
Collapse
Affiliation(s)
- Lizhen Zhu
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany; (L.Z.); (B.M.); (A.K.); (A.F.)
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China;
| | - Beiping Miao
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany; (L.Z.); (B.M.); (A.K.); (A.F.)
- Hopp Children’s Cancer Center (KiTZ), D-69120 Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), D-69120 Heidelberg, Germany
| | - Dagmara Dymerska
- Department of Genetics and Pathology, Hereditary Cancer Center, Pomeranian Medical University, Unii Lubelskiej 1, 71-252 Szczecin, Poland; (D.D.); (M.K.); (J.L.)
| | - Magdalena Kuswik
- Department of Genetics and Pathology, Hereditary Cancer Center, Pomeranian Medical University, Unii Lubelskiej 1, 71-252 Szczecin, Poland; (D.D.); (M.K.); (J.L.)
| | - Elena Bueno-Martínez
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biología y Genética Molecular (CSIC-UVa), 47003 Valladolid, Spain; (E.B.-M.); (L.S.-M.); (E.A.V.)
| | - Lara Sanoguera-Miralles
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biología y Genética Molecular (CSIC-UVa), 47003 Valladolid, Spain; (E.B.-M.); (L.S.-M.); (E.A.V.)
| | - Eladio A. Velasco
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biología y Genética Molecular (CSIC-UVa), 47003 Valladolid, Spain; (E.B.-M.); (L.S.-M.); (E.A.V.)
| | - Nagarajan Paramasivam
- Computational Oncology, Molecular Diagnostics Program, National Center for Tumor Diseases (NCT), D-69120 Heidelberg, Germany;
| | - Matthias Schlesner
- Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| | - Abhishek Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany; (L.Z.); (B.M.); (A.K.); (A.F.)
- Institute of Bioinformatics, International Technology Park, Bengaluru 560066, India
- Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Ying Yuan
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China;
| | - Jan Lubinski
- Department of Genetics and Pathology, Hereditary Cancer Center, Pomeranian Medical University, Unii Lubelskiej 1, 71-252 Szczecin, Poland; (D.D.); (M.K.); (J.L.)
| | - Obul Reddy Bandapalli
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany; (L.Z.); (B.M.); (A.K.); (A.F.)
- Hopp Children’s Cancer Center (KiTZ), D-69120 Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), D-69120 Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, D-69120 Heidelberg, Germany
- Correspondence: (O.R.B.); (K.H.)
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany; (L.Z.); (B.M.); (A.K.); (A.F.)
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, 30605 Pilsen, Czech Republic
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
- Correspondence: (O.R.B.); (K.H.)
| | - Asta Försti
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany; (L.Z.); (B.M.); (A.K.); (A.F.)
- Hopp Children’s Cancer Center (KiTZ), D-69120 Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), D-69120 Heidelberg, Germany
| |
Collapse
|
34
|
A Novel Pseudoalteromonas xiamenensis Marine Isolate as a Potential Probiotic: Anti-Inflammatory and Innate Immune Modulatory Effects against Thermal and Pathogenic Stresses. Mar Drugs 2021; 19:md19120707. [PMID: 34940706 PMCID: PMC8707914 DOI: 10.3390/md19120707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/11/2021] [Accepted: 12/12/2021] [Indexed: 11/16/2022] Open
Abstract
A marine bacterial strain was isolated from seawater and characterized for it beneficial probiotic effects using zebrafish as a model system. The strain was identified by morphological, physiological, biochemical, and phylogenetic analyses. The strain was most closely related to Pseudoalteromonas xiamenensis Y2, with 99.66% similarity; thus, we named it Pseudoalteromonas xiamenensis S1131. Improvement of host disease tolerance for the P. xiamenensis isolate was adapted in a zebrafish model using Edwardsiella piscicida challenge. The larvae were pre-exposed to P. xiamenensis prior to E. piscicida challenge, resulting in a 73.3% survival rate compared to a 46.6% survival for the control. The treated larvae tolerated elevated temperatures at 38 °C, with 85% survival, compared to 60% survival for the control. Assessment of immunomodulatory responses at the mRNA level demonstrated the suppression of pro-inflammatory markers tnfα and il6, and upregulation of heat shock protein hsp90 and mucin genes. The same effect was corroborated by immunoblot analysis, revealing significant inhibition of Tnfα and an enhanced expression of the Hsp90 protein. The antibacterial activity of P. xiamenensis may be related to mucin overexpression, which can suppress bacterial biofilm formation and enhance macrophage uptake. This phenomenon was evaluated using nonstimulated macrophage RAW264.7 cells. Further studies may be warranted to elucidate a complete profile of the probiotic effects, to expand the potential applications of the present P. xiamenensis isolate.
Collapse
|
35
|
MUC2 and related bacterial factors: Therapeutic targets for ulcerative colitis. EBioMedicine 2021; 74:103751. [PMID: 34902790 PMCID: PMC8671112 DOI: 10.1016/j.ebiom.2021.103751] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/21/2021] [Accepted: 11/30/2021] [Indexed: 12/26/2022] Open
Abstract
The mucin2 (MUC2) mucus barrier acts as the first barrier that prevents direct contact between intestinal bacteria and colonic epithelial cells. Bacterial factors related to the MUC2 mucus barrier play important roles in the response to changes in dietary patterns, MUC2 mucus barrier dysfunction, contact stimulation with colonic epithelial cells, and mucosal and submucosal inflammation during the occurrence and development of ulcerative colitis (UC). In this review, these underlying mechanisms are summarized and updated, and related interventions for treating UC, such as dietary adjustment, exogenous repair of the mucus barrier, microbiota transplantation and targeted elimination of pathogenic bacteria, are suggested. Such interventions are likely to induce and maintain a long and stable remission period and reduce or even avoid the recurrence of UC. A better mechanistic understanding of the MUC2 mucus barrier and its related bacterial factors may help researchers and clinicians to develop novel approaches for treating UC.
Collapse
|
36
|
Parrish A, Boudaud M, Kuehn A, Ollert M, Desai MS. Intestinal mucus barrier: a missing piece of the puzzle in food allergy. Trends Mol Med 2021; 28:36-50. [PMID: 34810087 DOI: 10.1016/j.molmed.2021.10.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 12/13/2022]
Abstract
The prevalence of food allergies has reached epidemic levels but the cause remains largely unknown. We discuss the clinical relevance of the gut mucosal barrier as a site for allergic sensitization to food. In this context, we focus on an important but overlooked part of the mucosal barrier in pathogenesis, the glycoprotein-rich mucus layer, and call attention to both beneficial and detrimental aspects of mucus-gut microbiome interactions. Studying the intricate links between the mucus barrier, the associated bacteria, and the mucosal immune system may advance our understanding of the mechanisms and inform prevention and treatment strategies in food allergy.
Collapse
Affiliation(s)
- Amy Parrish
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | - Marie Boudaud
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Annette Kuehn
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark.
| |
Collapse
|
37
|
Tiwari S, Begum S, Moreau F, Gorman H, Chadee K. Autophagy is required during high MUC2 mucin biosynthesis in colonic goblet cells to contend metabolic stress. Am J Physiol Gastrointest Liver Physiol 2021; 321:G489-G499. [PMID: 34494458 DOI: 10.1152/ajpgi.00221.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/02/2021] [Indexed: 01/31/2023]
Abstract
Goblet cells are specialized for the production and secretion of MUC2 glycoproteins that forms a thick layer covering the mucosal epithelium as a protective barrier against noxious substances and invading microbes. High MUC2 mucin biosynthesis induces endoplasmic reticulum (ER) stress and apoptosis in goblet cells during inflammatory and infectious diseases. Autophagy is an intracellular degradation process required for maintenance of intestinal homeostasis. In this study, we hypothesized that autophagy was triggered during high MUC2 mucin biosynthesis from colonic goblet cells to cope with metabolic stress. To interrogate this, we analyzed the autophagy process in high MUC2-producing human HT29-H and a clone HT29-L silenced for MUC2 expression by lentivirus-mediated shRNA, and WT and CRISPR/Cas9 MUC2 KO LS174T cells. Autophagy was constitutively increased in high MUC2-producing cells characterized by elevated pULK1S555 expression and increased numbers of autophagosomes as compared with MUC2 silenced or gene edited cells. Similarly, colonoids from Muc2+/+ but not Muc2-/- littermates differentiated into goblet cells showed increased autophagy. IL-22 treatment corrected misfolded MUC2 protein and alleviated the autophagy process in LS174T cells. This study highlights that autophagy plays an essential role in goblet cells to survive during high mucin biosynthesis by regulating cellular homeostasis.NEW & NOTEWORTHY It is unclear how colonic goblet cells survive by producing high output MUC2 mucin that triggers endoplasmic stress by misfolded MUC2 proteins. To cope with metabolic stress, we interrogated if autophagy played an essential role in regulating cellular homeostasis. Indeed, high MUC2 mucin biosynthesis dysregulated autophagy processes that was regulated by IL-22 to maintain gut barrier innate host defenses.
Collapse
Affiliation(s)
- Sameer Tiwari
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| | - Sharmin Begum
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| | - France Moreau
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| | - Hayley Gorman
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| | - Kris Chadee
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| |
Collapse
|
38
|
Chidambaram SB, Essa MM, Rathipriya AG, Bishir M, Ray B, Mahalakshmi AM, Tousif AH, Sakharkar MK, Kashyap RS, Friedland RP, Monaghan TM. Gut dysbiosis, defective autophagy and altered immune responses in neurodegenerative diseases: Tales of a vicious cycle. Pharmacol Ther 2021; 231:107988. [PMID: 34536490 DOI: 10.1016/j.pharmthera.2021.107988] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/16/2021] [Accepted: 08/25/2021] [Indexed: 02/08/2023]
Abstract
The human microbiota comprises trillions of symbiotic microorganisms and is involved in regulating gastrointestinal (GI), immune, nervous system and metabolic homeostasis. Recent observations suggest a bidirectional communication between the gut microbiota and the brain via immune, circulatory and neural pathways, termed the Gut-Brain Axis (GBA). Alterations in gut microbiota composition, such as seen with an increased number of pathobionts and a decreased number of symbionts, termed gut dysbiosis or microbial intestinal dysbiosis, plays a prominent role in the pathogenesis of central nervous system (CNS)-related disorders. Clinical reports confirm that GI symptoms often precede neurological symptoms several years before the development of neurodegenerative diseases (NDDs). Pathologically, gut dysbiosis disrupts the integrity of the intestinal barrier leading to ingress of pathobionts and toxic metabolites into the systemic circulation causing GBA dysregulation. Subsequently, chronic neuroinflammation via dysregulated immune activation triggers the accumulation of neurotoxic misfolded proteins in and around CNS cells resulting in neuronal death. Emerging evidence links gut dysbiosis to the aggravation and/or spread of proteinopathies from the peripheral nervous system to the CNS and defective autophagy-mediated proteinopathies. This review summarizes the current understanding of the role of gut microbiota in NDDs, and highlights a vicious cycle of gut dysbiosis, immune-mediated chronic neuroinflammation, impaired autophagy and proteinopathies, which contributes to the development of neurodegeneration in Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, amyotrophic lateral sclerosis and frontotemporal lobar degeneration. We also discuss novel therapeutic strategies targeting the modulation of gut dysbiosis through prebiotics, probiotics, synbiotics or dietary interventions, and faecal microbial transplantation (FMT) in the management of NDDs.
Collapse
Affiliation(s)
- Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India; Centre for Experimental Pharmacology and Toxicology (CPT), JSS Academy of Higher Education & Research, Mysuru 570015, KA, India.
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat 123, Oman; Ageing and Dementia Research Group, Sultan Qaboos University, Muscat 123, Oman; Biomedical Sciences Department, University of Pacific, Sacramento, CA, USA.
| | - A G Rathipriya
- Food and Brain Research Foundation, Chennai 600 094, Tamil Nadu, India
| | - Muhammed Bishir
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - Bipul Ray
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India; Centre for Experimental Pharmacology and Toxicology (CPT), JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - Arehally M Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - A H Tousif
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India; Centre for Experimental Pharmacology and Toxicology (CPT), JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - Meena K Sakharkar
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
| | - Rajpal Singh Kashyap
- Research Centre, Dr G. M. Taori Central India Institute of Medical Sciences (CIIMS), Nagpur, Maharashtra, India
| | - Robert P Friedland
- Department of Neurology, University of Louisville, Louisville, KY 40292, USA
| | - Tanya M Monaghan
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham NG7 2UH, UK; Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK.
| |
Collapse
|
39
|
Gundamaraju R, Chong WC. Consequence of distinctive expression of MUC2 in colorectal cancers: How much is actually bad? Biochim Biophys Acta Rev Cancer 2021; 1876:188579. [PMID: 34139275 DOI: 10.1016/j.bbcan.2021.188579] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 12/18/2022]
Abstract
Colorectal cancer (CRC) exhibits complex pathogenesis via compromised intestinal mucosal barrier. It is accepted that goblet cells secrete mucin which line the intestinal mucosal barrier and offer wide range protection and maintain the gut integrity. The principal mucin in the small and large intestine which is Mucin2 (MUC2) is predominantly expressed in the goblet cells which play a pivotal role in intestinal homeostasis. Its disruption is associated with diverse diseases and carcinomas. MUC2 has lately been identified as a principal marker in various mechanisms and secretory cell lineage. While MUC2 expression is regulated by various modulators, alterations in its expression are associated with immunomodulation, differences in tumor immunity and also regulation of microbiota. In the light of current literature, the present review explicates the regulation, functional mechanisms and essential role of MUC2 in colorectal cancer and aids in providing deep understanding of pathogenesis of the disease and also specifies the importance of the MUC2 in gaining more insights about the subtypes of colorectal cancer and how it can succour in approximating the prognosis and survival of the patients.
Collapse
Affiliation(s)
- Rohit Gundamaraju
- ER Stress and Gut Mucosal Immunology Laboratory, School of Health Sciences, University of Tasmania, Launceston, Tasmania 7248, Australia.
| | - Wai Chin Chong
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; Department of Molecular and Translational Science, School of Medicine, Nursing, and Health Science, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
40
|
Bai D, Sun T, Zhao J, Du J, Bu X, Cao W, Zhao Y, Lu N. Oroxylin A maintains the colonic mucus barrier to reduce disease susceptibility by reconstituting a dietary fiber-deprived gut microbiota. Cancer Lett 2021; 515:73-85. [PMID: 34052330 DOI: 10.1016/j.canlet.2021.05.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/16/2022]
Abstract
Dietary fiber intake helps to maintain gut homeostasis. Fiber deficiency causes commensals to utilize mucins as an energy source to destroy mucus layer, thus promoting susceptibility to inflammatory bowel disease. Here, we reported that oroxylin A, a natural flavonoid, ameliorated low-grade colonic inflammation caused by fiber deficiency, alleviated colitis, and further prevented colitis-associated colon cancer in mice. The anti-inflammatory effect of oroxylin A was due to its alteration of gut microbiota. We found that the levels of Eubacterium coprostanoligenes was significantly increased by oroxylin A and the colonized Eubacterium coprostanoligenes significantly protected against colitis and carcinogenesis in colon of mice. Together, our results in this study suggest that oroxylin A may reduce the susceptibility to intestinal diseases by increasing the level of Eubacterium coprostanoligenes which could provide a therapeutic alternation for the treatment of intestinal diseases.
Collapse
Affiliation(s)
- Dongsheng Bai
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Tifan Sun
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Jiawei Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Jiaying Du
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Xiumin Bu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Wangjia Cao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Yue Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China.
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
41
|
Ten Hove AS, Seppen J, de Jonge WJ. Neuronal innervation of the intestinal crypt. Am J Physiol Gastrointest Liver Physiol 2021; 320:G193-G205. [PMID: 33296267 DOI: 10.1152/ajpgi.00239.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mucosal damage is a key feature of inflammatory bowel diseases (IBD) and healing of the mucosa is an endpoint of IBD treatment that is often difficult to achieve. Autonomic neurons of the parasympathetic and sympathetic nervous system may influence intestinal epithelial cell growth and modulating epithelial innervation could for that reason serve as an interesting therapeutic option to improve mucosal healing. Understanding of the biological processes triggered by nonspecific and specific epithelial adrenergic and cholinergic receptor activation is of key importance. At present, with rising technological advances, bioelectronic neuromodulation as treatment modality has gained momentum. We discuss the current view on state-of-the-art innervation of the intestinal crypt and its impact on epithelial cell growth and differentiation. Furthermore, we outline bioelectronic technology and review its relevance to wound healing processes.
Collapse
Affiliation(s)
- Anne S Ten Hove
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Jurgen Seppen
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Wouter J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands.,Department of General, Visceral, Thoracic and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
42
|
Krolevets TS, Livzan MA, Mozgovoy SI. The Role of the Microbiome and Intestinal Mucosal Barrier in the Development and Progression of Non-Alcoholic Fatty Liver Disease. RUSSIAN JOURNAL OF GASTROENTEROLOGY, HEPATOLOGY, COLOPROCTOLOGY 2020; 30:42-48. [DOI: 10.22416/1382-4376-2020-30-5-42-48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Aim. To review available data on the role of the microbiome and intestinal mucosal barrier in the development and progression of non-alcoholic fatty liver disease (NAFLD).Key points. The role of the human microbiome in the development and progression of NAFLD is associated with its effects on the risk factors (obesity, insulin resistance, type 2 diabetes), permeability of the intestinal barrier and absorption of such substances as short-chain fatty acids, bile acids, choline and endogenous ethanol. Liver fibrosis constitutes the leading factor determining the prognosis of patients in NAFLD, including cases associated with cardiovascular complications. Changes in the microbiome composition were demonstrated for various degrees of fibrosis in NAFLD.Conclusion. The results of modern studies confirm the formation of a new concept in the pathophysiology of NAFLD, which encourages the development of new therapeutic strategies.
Collapse
|
43
|
|
44
|
Martinez-Carrasco R, Argüeso P, Fini ME. Dynasore protects ocular surface mucosal epithelia subjected to oxidative stress by maintaining UPR and calcium homeostasis. Free Radic Biol Med 2020; 160:57-66. [PMID: 32791188 PMCID: PMC7704702 DOI: 10.1016/j.freeradbiomed.2020.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/02/2020] [Accepted: 07/02/2020] [Indexed: 02/06/2023]
Abstract
The mucosal epithelia of the ocular surface protect against external threats to the eye. Using a model of human stratified corneal epithelial cells with mucosal differentiation, we previously demonstrated that a small molecule inhibitor of dynamin GTPases, dynasore, prevents damage to cells and their transcellular barriers when subjected to oxidative stress. Investigating mechanisms, we now report the novel finding that dynasore acts by maintaining Ca+2 homeostasis, thereby inhibiting the PERK branch of the unfolded protein response (UPR) that promotes cell death. Dynasore was found to protect mitochondria by preventing mitochondrial permeability transition pore opening (mPTP), but, unlike reports using other systems, this was not mediated by dynamin family member DRP1. Necrostatin-1, an inhibitor of RIPK1 and lytic forms of programmed cell death, also inhibited mPTP opening and further protected the plasma membrane barrier. Significantly, necrostatin-1 did not protect the mucosal barrier. Oxidative stress increased mRNA for sXBP1, a marker of the IRE1 branch of the UPR, and CHOP, a marker of the PERK branch. It also stimulated phosphorylation of eIF2α, the upstream regulator of CHOP, as well as an increase in intracellular Ca2+. Dynasore selectively inhibited the increase in PERK branch markers, and also prevented the increase intracellular Ca2+ in response to oxidative stress. The increase in PERK branch markers were also inhibited when cells were treated with the cell permeable Ca2+ chelator, BAPTA-AM. To our knowledge, this is the first time that dynasore has been shown to have an effect on the UPR and suggests therapeutic applications.
Collapse
Affiliation(s)
- Rafael Martinez-Carrasco
- New England Eye Center of Tufts Medical Center, Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA
| | - Pablo Argüeso
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - M Elizabeth Fini
- New England Eye Center of Tufts Medical Center, Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA; Program in Pharmacology and Drug Development, Tufts Graduate School of Biomedical Sciences, Tufts University, Boston, MA, USA.
| |
Collapse
|
45
|
Gao F, Fan H. Heat shock protein 5 and inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2020; 28:802-806. [DOI: 10.11569/wcjd.v28.i16.802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease is a kind of chronic recurrent intestinal inflammatory disease whose occurrence and development are affected by the integrity of the mucosal barrier. As the main component of the mucosal barrier, intestinal epithelial cells mainly include Paneth cells, goblet cells, etc. Heat shock protein 5 is a key factor for endoplasmic reticulum stress, and it affects the survival and apoptosis of intestinal epithelial cells mainly through endoplasmic reticulum stress pathways, and then participates in the process of inflammatory bowel disease.
Collapse
Affiliation(s)
- Fei Gao
- Department of Integrated Chinese and Western Medicine, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Heng Fan
- Department of Integrated Chinese and Western Medicine, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
46
|
Singh P, Sanchez-Fernandez LL, Ramiro-Cortijo D, Ochoa-Allemant P, Perides G, Liu Y, Medina-Morales E, Yakah W, Freedman SD, Martin CR. Maltodextrin-induced intestinal injury in a neonatal mouse model. Dis Model Mech 2020; 13:dmm044776. [PMID: 32753526 PMCID: PMC7473650 DOI: 10.1242/dmm.044776] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
Prematurity and enteral feedings are major risk factors for intestinal injury leading to necrotizing enterocolitis (NEC). An immature digestive system can lead to maldigestion of macronutrients and increased vulnerability to intestinal injury. The aim of this study was to test in neonatal mice the effect of maltodextrin, a complex carbohydrate, on the risk of intestinal injury. The goal was to develop a robust and highly reproducible murine model of intestinal injury that allows insight into the pathogenesis and therapeutic interventions of nutrient-driven intestinal injury. Five- to 6-day-old C57BL/6 mice were assigned to the following groups: dam fed (D); D+hypoxia+Klebsiella pneumoniae; maltodextrin-dominant human infant formula (M) only; M+hypoxia; and M+hypoxia+K. pneumoniae. The mice in all M groups were gavage fed five times a day for 4 days. Mice were exposed to hypoxia twice a day for 10 min prior to the first and last feedings, and K. pneumoniae was added to feedings as per group assignment. Mice in all M groups demonstrated reduced body weight, increased small intestinal dilatation and increased intestinal injury scores. Maltodextrin-dominant infant formula with hypoxia led to intestinal injury in neonatal mice accompanied by loss of villi, increased MUC2 production, altered expression of tight junction proteins, enhanced intestinal permeability, increased cell death and higher levels of intestinal inflammatory mediators. This robust and highly reproducible model allows for further interrogation of the effects of nutrients on pathogenic factors leading to intestinal injury and NEC in preterm infants.This article has an associated First Person interview with the first author of the paper.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Cytokines/metabolism
- Disease Models, Animal
- Enterocolitis, Necrotizing/chemically induced
- Enterocolitis, Necrotizing/metabolism
- Enterocolitis, Necrotizing/microbiology
- Enterocolitis, Necrotizing/pathology
- Goblet Cells/metabolism
- Goblet Cells/microbiology
- Goblet Cells/pathology
- Hypoxia/complications
- Inflammation Mediators/metabolism
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/microbiology
- Intestinal Mucosa/pathology
- Intestine, Small/metabolism
- Intestine, Small/microbiology
- Intestine, Small/pathology
- Klebsiella pneumoniae/pathogenicity
- Mice, Inbred C57BL
- Microvilli/pathology
- Mucin-2/metabolism
- Permeability
- Polysaccharides
- Tight Junction Proteins/metabolism
Collapse
Affiliation(s)
- Pratibha Singh
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Lady Leidy Sanchez-Fernandez
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - David Ramiro-Cortijo
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Pedro Ochoa-Allemant
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - George Perides
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Yan Liu
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Esli Medina-Morales
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - William Yakah
- Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Steven D Freedman
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Camilia R Martin
- Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
47
|
Li C, Wang X, Sun S, Liu S, Huan Y, Li R, Liu Q, Cao H, Zhou T, Lei L, Liu M, Shen Z. Effects of a ready-to-eat cereal formula powder on glucose metabolism, inflammation, and gut microbiota in diabetic db/db mice. Food Sci Nutr 2020; 8:4523-4533. [PMID: 32884732 PMCID: PMC7455957 DOI: 10.1002/fsn3.1761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 12/17/2022] Open
Abstract
The cereal formula powder, Zhengda Jingshan (ZDJS), comprises dietary fiber, multivitamins, fine protein, and various cereal ingredients. The present study evaluated the effects of ZDJS on glucose metabolism and explored the corresponding mechanisms in terms of modulating gut microbiota and the fecal metabolome. Type 2 diabetic db/db mice were given ZDJS (1 g/kg) orally twice daily for 55 days, after which glucose metabolism, inflammation, gut microbiota, and fecal metabolomics were assayed. Repeated administration of ZDJS was associated with a trend toward decreasing fasting blood glucose and a 0.12% decrease in hemoglobin A1c (HbA1c), as well as statistically significant increases in the insulin sensitivity index and decreases in serum levels of tumor necrosis factor (TNF-α) and ileum expression of mucin-2. ZDJS also ameliorated the compensatory enlargement of islets and decreased the ratio of the α-cell area to total islet area; however, this amelioration of impaired oral glucose tolerance became less pronounced as treatment continued. In addition, ZDJS remarkably decreased the abundance of phylum Proteobacteria and the phylum ratio of Firmicutes to Bacteroidetes, as well as altered the fecal metabolic profile. Taken together, our findings demonstrate that ZDJS improved glucose metabolism and reduced inflammation in type 2 diabetic db/db mice, which may be associated with a reshaping of the gut microbiome and fecal metabolome in db/db mice. Thus, our study suggests that ZDJS may represent a complementary therapy for patients with type 2 diabetes.
Collapse
Affiliation(s)
- Caina Li
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesKey Laboratory of Polymorphic Drugs of BeijingInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xing Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesKey Laboratory of Polymorphic Drugs of BeijingInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Sujuan Sun
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesKey Laboratory of Polymorphic Drugs of BeijingInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shuainan Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesKey Laboratory of Polymorphic Drugs of BeijingInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yi Huan
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesKey Laboratory of Polymorphic Drugs of BeijingInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Rongcui Li
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesKey Laboratory of Polymorphic Drugs of BeijingInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Quan Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesKey Laboratory of Polymorphic Drugs of BeijingInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Hui Cao
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesKey Laboratory of Polymorphic Drugs of BeijingInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Tian Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesKey Laboratory of Polymorphic Drugs of BeijingInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lei Lei
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesKey Laboratory of Polymorphic Drugs of BeijingInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Minzhi Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesKey Laboratory of Polymorphic Drugs of BeijingInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhufang Shen
- State Key Laboratory of Bioactive Substances and Functions of Natural MedicinesKey Laboratory of Polymorphic Drugs of BeijingInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
48
|
Dotsenko V, Oittinen M, Taavela J, Popp A, Peräaho M, Staff S, Sarin J, Leon F, Isola J, Mäki M, Viiri K. Genome-Wide Transcriptomic Analysis of Intestinal Mucosa in Celiac Disease Patients on a Gluten-Free Diet and Postgluten Challenge. Cell Mol Gastroenterol Hepatol 2020; 11:13-32. [PMID: 32745639 PMCID: PMC7593586 DOI: 10.1016/j.jcmgh.2020.07.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Gluten challenge studies are instrumental in understanding the pathophysiology of celiac disease. Our aims in this study were to reveal early gluten-induced transcriptomic changes in duodenal biopsies and to find tools for clinics. METHODS Duodenal biopsies were collected from 15 celiac disease patients on a strict long-term gluten-free diet (GFD) prior to and post a gluten challenge (PGC) and from 6 healthy control individuals (DC). Biopsy RNA was subjected to genome-wide 3' RNA-Seq. Sequencing data was used to determine the differences between the three groups and was compared to sequencing data from the public repositories. The biopsies underwent morphometric analyses. RESULTS In DC vs. GFD group comparisons, 167 differentially expressed genes were identified with 117 genes downregulated and 50 genes upregulated. In PGC vs. GFD group comparisons, 417 differentially expressed genes were identified with 195 genes downregulated and 222 genes upregulated. Celiac disease patients on a GFD were not "healthy". In particular, genes encoding proteins for transporting small molecules were expressed less. In addition to the activation of immune response genes, a gluten challenge induced hyperactive intestinal wnt-signaling and consequent immature crypt gene expression resulting in less differentiated epithelium. Biopsy gene expression in response to a gluten challenge correlated with the extent of the histological damage. Regression models using only four gene transcripts described 97.2% of the mucosal morphology and 98.0% of the inflammatory changes observed. CONCLUSIONS Our gluten challenge trial design provided an opportunity to study the transition from health to disease. The results show that even on a strict GFD, despite being deemed healthy, patients reveal patterns of ongoing disease. Here, a transcriptomic regression model estimating the extent of gluten-induced duodenal mucosal injury is presented.
Collapse
Affiliation(s)
- Valeriia Dotsenko
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University Tampere, Finland
| | - Mikko Oittinen
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University Tampere, Finland
| | - Juha Taavela
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University Tampere, Finland,Department of Internal Medicine, Central Finland Central Hospital, Jyväskylä, Finland
| | - Alina Popp
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University Tampere, Finland,Pediatric Department, Carol Davila University of Medicine and Pharmacy, Alessandrescu-Rusescu National Institute for Mother and Child Health, Bucharest, Romania
| | - Markku Peräaho
- Department of Internal Medicine, Central Finland Central Hospital, Jyväskylä, Finland
| | - Synnöve Staff
- Department of Obstetrics and Gynecology and Tays Cancer Centre, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University, Tampere, Finland
| | - Jani Sarin
- Laboratory of Cancer Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland,Jilab Inc, Tampere, Finland
| | | | - Jorma Isola
- Laboratory of Cancer Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland,Jilab Inc, Tampere, Finland
| | - Markku Mäki
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University Tampere, Finland
| | - Keijo Viiri
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University Tampere, Finland,Correspondence Address correspondence to: Keijo Viiri, PhD, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Arvo Ylpön katu 34, Tampere, FIN-33520, Finland; fax: +35833641369.
| |
Collapse
|
49
|
Potential role of ovomucin and its peptides in modulation of intestinal health: A review. Int J Biol Macromol 2020; 162:385-393. [PMID: 32569696 PMCID: PMC7305749 DOI: 10.1016/j.ijbiomac.2020.06.148] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/11/2020] [Accepted: 06/16/2020] [Indexed: 01/06/2023]
Abstract
Intestinal dysfunction, which may cause a series of metabolic diseases, has become a worldwide health problem. In the past few years, studies have shown that consumption of poultry eggs has the potential to prevent a variety of metabolic diseases, and increasing attention has been directed to the bioactive proteins and their peptides in poultry eggs. This review mainly focused on the biological activities of an important egg-derived protein named ovomucin. Ovomucin and its derivatives have good anti-inflammatory, antioxidant, immunity-regulating and other biological functions. These activities may affect the physical, biological and immune barriers associated with intestinal health. This paper reviewed the structure and the structure-activity relationship of ovomucin,the potential role of ovomucin and its derivatives in modulation of intestinal health are also summarized. Finally, the potential applications of ovomucin and its peptides as functional food components to prevent and assist in the pretreatment of intestinal health problems are prospected.
Collapse
|
50
|
Li CN, Wang X, Lei L, Liu MZ, Li RC, Sun SJ, Liu SN, Huan Y, Zhou T, Liu Q, Cao H, Bai GL, Han YW, Shen ZF. Berberine combined with stachyose induces better glycometabolism than berberine alone through modulating gut microbiota and fecal metabolomics in diabetic mice. Phytother Res 2019; 34:1166-1174. [PMID: 31833107 PMCID: PMC7216932 DOI: 10.1002/ptr.6588] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/17/2019] [Accepted: 11/24/2019] [Indexed: 01/19/2023]
Abstract
Berberine (BBR), a small alkaloid, is used as a hypoglycemic agent in China. Stachyose (Sta), a Rehmannia glutinosa oligosaccharide, acts as a prebiotic. This study aimed to evaluate whether BBR combined with Sta produced better glycometabolism than BBR alone, and explored the effects on gut microbiota and metabolomics. Type‐2 diabetic db/db mice were administered BBR (100 mg/kg), Sta (200 mg/kg), or both by gavage once daily. Glucose metabolism, the balance of α‐ and β‐cells, and mucin‐2 expression were ameliorated by combined treatment of BBR and Sta, with stronger effects than upon treatment with BBR alone. The microbial diversity and richness were altered after combined treatment and after treatment with BBR alone. The abundance of Akkermansia muciniphila was increased by combined treatment compared to treatment with BBR alone, while the levels of the metabolite all‐trans‐heptaprenyl diphosphate were decreased and the levels of fumaric acid were increased, which both showed a strong correlation with A. muciniphila. In summary, BBR combined with Sta produced better glycometabolism than BBR alone through modulating gut microbiota and fecal metabolomics, and may aid in the development of a novel pharmaceutical strategy for treating Type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Cai-Na Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xing Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lei Lei
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Min-Zhi Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Rong-Cui Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Su-Juan Sun
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shuai-Nan Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yi Huan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Tian Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Quan Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Hui Cao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Guo-Liang Bai
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yu-Wei Han
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhu-Fang Shen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Key laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|