1
|
Fang L, Peng H, Tan Z, Deng N, Peng X. The Role of Gut Microbiota on Intestinal Fibrosis in Inflammatory Bowel Disease and Traditional Chinese Medicine Intervention. J Inflamm Res 2025; 18:5951-5967. [PMID: 40357383 PMCID: PMC12067688 DOI: 10.2147/jir.s504827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, relapsing inflammatory disorder of the intestine, frequently complicated by intestinal fibrosis. As fibrosis progresses, it can result in luminal stricture and compromised intestinal function, significantly diminishing patients' quality of life. Emerging evidence suggests that gut microbiota and their metabolites contribute to the pathogenesis of IBD-associated intestinal fibrosis by influencing inflammation and modulating immune responses. This review systematically explores the mechanistic link between gut microbiota and intestinal fibrosis in IBD and evaluates the therapeutic potential of traditional Chinese medicine (TCM) interventions. Relevant studies were retrieved from PubMed, Web of Science, Embase, Scopus, CNKI, Wanfang, and VIP databases. Findings indicate that TCM, including Chinese herbal prescriptions and bioactive constituents, can modulate gut microbiota composition and microbial metabolites, ultimately alleviating intestinal fibrosis through anti-inflammatory, immunemodulatory, and anti-fibrotic mechanisms. These insights highlight the potential of TCM as a promising strategy for targeting gut microbiota in the management of IBD-associated fibrosis.
Collapse
Affiliation(s)
- Leyao Fang
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Huiyi Peng
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Zhoujin Tan
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Na Deng
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Xinxin Peng
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| |
Collapse
|
2
|
Zhang Y, Zhuang H, Chen K, Zhao Y, Wang D, Ran T, Zou D. Intestinal fibrosis associated with inflammatory bowel disease: Known and unknown. Chin Med J (Engl) 2025; 138:883-893. [PMID: 40012095 PMCID: PMC12037091 DOI: 10.1097/cm9.0000000000003545] [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: 10/29/2024] [Indexed: 02/28/2025] Open
Abstract
ABSTRACT Intestinal fibrosis is a major complication of inflammatory bowel disease (IBD), leading to a high incidence of surgical interventions and significant disability. Despite its clinical relevance, no targeted pharmacological therapies are currently available. This review aims to explore the underlying mechanisms driving intestinal fibrosis and address unresolved scientific questions, offering insights into potential future therapeutic strategies. We conducted a literature review using data from PubMed up to October 2024, focusing on studies related to IBD and fibrosis. Intestinal fibrosis results from a complex network involving stromal cells, immune cells, epithelial cells, and the gut microbiota. Chronic inflammation, driven by factors such as dysbiosis, epithelial injury, and immune activation, leads to the production of cytokines like interleukin (IL)-1β, IL-17, and transforming growth factor (TGF)-β. These mediators activate various stromal cell populations, including fibroblasts, pericytes, and smooth muscle cells. The activated stromal cells secrete excessive extracellular matrix components, thereby promoting fibrosis. Additionally, stromal cells influence the immune microenvironment through cytokine production. Future research would focus on elucidating the temporal and spatial relationships between immune cell-driven inflammation and stromal cell-mediated fibrosis. Additionally, investigations are needed to clarify the differentiation origins of excessive extracellular matrix-producing cells, particularly fibroblast activation protein (FAP) + fibroblasts, in the context of intestinal fibrosis. In conclusion, aberrant stromal cell activation, triggered by upstream immune signals, is a key mechanism underlying intestinal fibrosis. Further investigations into immune-stromal cell interactions and stromal cell activation are essential for the development of therapeutic strategies to prevent, alleviate, and potentially reverse fibrosis.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Haiming Zhuang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kai Chen
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yizhou Zhao
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Danshu Wang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Taojing Ran
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
3
|
Chauhan G, Rieder F. The Pathogenesis of Inflammatory Bowel Diseases. Surg Clin North Am 2025; 105:201-215. [PMID: 40015812 PMCID: PMC11868724 DOI: 10.1016/j.suc.2024.10.008] [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] [Indexed: 03/01/2025]
Abstract
Inflammatory bowel diseases (IBDs) are relapsing, remitting inflammatory diseases of the intestinal tract. Familial aggregation and genome-wide association studies revealed susceptibility variants that point toward a combination of innate immune and adaptive immune dysregulation that in concert with environmental factors, such as our microbiome, can initiate and perpetuate inflammation. Innate immune perturbations include functional abnormalities in the intestinal barrier, endoplasmic reticulum stress, and abnormal recognition of microbes. Adaptive immune changes include dysregulation of cytokines, regulatory T cells, and leukocyte migration. IBD is linked with an abnormal wound-healing response leading to fibrosis. This article summarizes key pathogenic mechanisms in the pathogenesis of IBDs.
Collapse
Affiliation(s)
- Gaurav Chauhan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA; Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases Institute; Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| |
Collapse
|
4
|
Zhang L, Liu Q, Yang X, Su C, Ding H, Hu J, Han W, Wu J, Zhang M, Zuo L, Mei Q. Mechanosensitive Ion Channel PIEZO1 as a Key Regulator of Intestinal Fibrosis in Crohn's Disease. Inflamm Bowel Dis 2025:izaf041. [PMID: 40053528 DOI: 10.1093/ibd/izaf041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Indexed: 03/09/2025]
Abstract
BACKGROUND We aimed to elucidate the function of the mechanosensitive ion channel PIEZO1 in intestinal fibrosis, which is invariably associated with Crohn's disease (CD) and often results in strictures and obstructions, requiring surgical intervention. Notably, PIEZO1 is strongly expressed in fibrotic tissues and linked with fibrotic progression. METHODS Intestinal tissues were procured from 28 patients diagnosed with CD and 8 healthy control subjects. Histological and immunofluorescence assays verified that PIEZO1 is substantially overexpressed in fibrotic intestinal tissues and is involved in epithelial‒mesenchymal transition (EMT). Further gene knockout experiments and transcriptome sequencing elucidated the specific role of PIEZO1 in the pathogenesis of intestinal fibrosis in CD. We generated mice with Piezo1 deletion specifically in intestinal epithelial cells (Piezo1f/f Vilcre) to validate in vivo that inhibiting Piezo1 function attenuates or reverses intestinal fibrosis associated with CD. RESULTS PIEZO1 expression was strongly increased in the fibrotic small intestine of CD patients, thereby promoting EMT and exacerbating intestinal fibrosis. In vivo investigations revealed that the conditional suppression of Piezo1 in intestinal epithelial cells significantly mitigated intestinal fibrosis in dextran sulphate sodium (DSS)- and 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced chronic colitis model mice. In vitro examinations revealed that Piezo1 expression in intestinal epithelial cells preserved the stability of HIF-1α, induced EMT to stimulate the expression of fibrosis-associated molecules, and promoted fibrosis. CONCLUSION PIEZO1 plays a pivotal role in the regulation of intestinal fibrosis by maintaining the levels of HIF-1α, thereby promoting EMT. Therapeutic strategies targeting PIEZO1 could be used to prevent intestinal fibrosis in CD patients.
Collapse
Affiliation(s)
- Luyao Zhang
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qiuyuan Liu
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaodong Yang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chang Su
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hao Ding
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Hu
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Han
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Juan Wu
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Manli Zhang
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Zuo
- College of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Qiao Mei
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
5
|
Chen Y, Liu J, Zhong S, Zhang T, Yuan J, Zhang J, Chen Y, Liang J, Chen Y, Hou S, Huang H, Gao J. Monotropein inhibits epithelial-mesenchymal transition in chronic colitis via the mTOR/P70S6K pathway. Front Pharmacol 2025; 16:1536091. [PMID: 40041493 PMCID: PMC11876156 DOI: 10.3389/fphar.2025.1536091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/21/2025] [Indexed: 03/06/2025] Open
Abstract
Introduction Patients with chronic colitis are at risk of developing intestinal fibrosis through epithelial-mesenchymal transition (EMT). Monotropein (MON) is the main active ingredient in the traditional Chinese medicine Morinda officinalis How. It has been reported that monotropein can improve ulcerative colitis, but the mechanism remains unclear. However, whether monotropein can improve chronic colitis-associated intestinal fibrosis remains unknown. The study aimed to investigate the effect of monotropein on EMT in chronic colitis and its underlying mechanism. Methods The mice chronic colitis model was induced by dextran sodium sulfate (DSS). Cytokines were detected by ELISA. Concentrations of fluorescein isothiocyanate dextran (FITC-Dextran) in serum were detected using a fluorescein microplate analyzer. Intestinal tight junction proteins were detected by immunofluorescence. EMT marker proteins were detected by immunohistochemistry. Transforming growth factor-β1 (TGF-β1) was used to induce EMT in IEC-6 cells. Western blot, real-time quantitative PCR, and immunofluorescence were used to test the inhibitory effect of monotropein on the development of EMT and explore its mechanism. Results Results showed that monotropein significantly improved colonic injury and inhibited the expression of colonic tissue EMT marker protein. In addition, molecular docking and molecular dynamics (MD) simulation, cellular thermal shift assay (CETSA), and drug affinity responsive target stability (DARTS) assay validated monotropein targeting of mTOR. Monotropein inhibited TGF-β1-induced EMT in IEC-6 cells, inhibited the phosphorylation of mTOR and its downstream proteins, and increased the autophagy activity in chronic colitis mice and IEC-6 cells. Discussion The study indicates that monotropein inhibits the development of EMT in DSS-induced chronic colitis mice and TGF-β1-induced IEC-6 cells. Its inhibitory effect on EMT is associated with the mTOR/P70S6K pathway.
Collapse
Affiliation(s)
- Yuanfan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- College of Education, Guangzhou Huali Science and Technology Vocational College, Guangzhou, Guangdong, China
- College of Education, University of Visayas, Cebu, Philippines
| | - Jiaying Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shaowen Zhong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Tianwu Zhang
- Pu’er Hospital of Traditional Chinese Medicine, Puer, Kunming, Yunnan, China
| | - Jin Yuan
- Pu’er Hospital of Traditional Chinese Medicine, Puer, Kunming, Yunnan, China
| | - Jing Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ying Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jian Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yonger Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- School of Basic Medical Sciences, State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shaozhen Hou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Haiyang Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Development Planning Department, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jie Gao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Nanì MF, Pagano E, De Cicco P, Lucariello G, Cattaneo F, Tropeano FP, Cicia D, Amico R, Raucci F, Ercolano G, Maione F, Rinaldi MM, Esposito F, Ammendola R, Luglio G, Capasso R, Makriyannis A, Petrosino S, Borrelli F, Romano B, Izzo AA. Pharmacological Inhibition of N-Acylethanolamine Acid Amidase (NAAA) Mitigates Intestinal Fibrosis Through Modulation of Macrophage Activity. J Crohns Colitis 2025; 19:jjae132. [PMID: 39211986 PMCID: PMC11836880 DOI: 10.1093/ecco-jcc/jjae132] [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: 12/21/2023] [Revised: 08/10/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND AIMS Intestinal fibrosis, a frequent complication of inflammatory bowel disease, is characterized by stricture formation with no pharmacological treatment to date. N-acylethanolamine acid amidase (NAAA) is responsible for the hydrolysis of acylethanolamides (AEs, eg, palmitoylethanolamide and oleoylethanolamide). Here, we investigated NAAA and AE signaling in gut fibrosis. METHODS NAAA and AE signaling were evaluated in human intestinal specimens from patients with stenotic Crohn's disease (CD). Gut fibrosis was induced by 2,4,6-trinitrobenzenesulfonic acid, monitored by colonoscopy, and assessed by qRT-PCR, histological analyses, and confocal microscopy. Immune cells in mesenteric lymph nodes were analyzed by FACS. Colonic fibroblasts were cultured in conditioned media derived from polarized or non-polarized bone marrow-derived macrophages (BMDMs). IL-23 signaling was evaluated by qRT-PCR, ELISA, FACS, and western blot in BMDMs and in lamina propria CX3CR1+ cells. RESULTS In ileocolonic human CD strictures, increased transcript expression of NAAA was observed with a decrease in its substrates oleoylethanolamide and palmitoylethanolamide. NAAA inhibition reduced intestinal fibrosis in vivo, as indicated by a decrease in inflammatory parameters, collagen deposition, and fibrosis-related genes, including those involved in epithelial-to-mesenchymal transition. More in-depth studies revealed modulation of the immune response related to IL-23 following NAAA inhibition. The antifibrotic actions of NAAA inhibition are mediated by Mφ and M2 macrophages that indirectly affect fibroblast collagenogenesis. NAAA inhibitor AM9053 normalized IL-23 signaling in BMDMs and in lamina propria CX3CR1+ cells. CONCLUSIONS Our findings provide new insights into the pathophysiological mechanism of intestinal fibrosis and identify NAAA as a promising target for the development of therapeutic treatments to alleviate CD-related fibrosis.
Collapse
Affiliation(s)
- Maria Francesca Nanì
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Ester Pagano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Paola De Cicco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Giuseppe Lucariello
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Fabio Cattaneo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Francesca Paola Tropeano
- Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Donatella Cicia
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Rebecca Amico
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Federica Raucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Giuseppe Ercolano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Maria Michela Rinaldi
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Fabiana Esposito
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Rosario Ammendola
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Gaetano Luglio
- Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
| | - Alexandros Makriyannis
- Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Stefania Petrosino
- Institute of Biomolecular Chemistry, National Research Council, Pozzuoli, Italy
- Epitech Group SpA, Saccolongo, Italy
| | - Francesca Borrelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Barbara Romano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Angelo A Izzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
7
|
Tavares de Sousa H, Ferreira M, Gullo I, Rocha AM, Pedro A, Leitão D, Oliveira C, Carneiro F, Magro F. Fibrosis-related Transcriptome Unveils a Distinctive Remodelling Matrix Pattern in Penetrating Ileal Crohn's Disease. J Crohns Colitis 2024; 18:1741-1752. [PMID: 38700484 DOI: 10.1093/ecco-jcc/jjae064] [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: 12/30/2023] [Revised: 04/02/2024] [Accepted: 05/01/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND AND AIMS Stricturing [B2] and penetrating [B3] ileal Crohn's disease have been reported to present similar levels of histopathological transmural fibrosis. This study aimed to compare the fibrosis-related transcriptomic profiles of penetrating and stricturing ileal Crohn's disease. METHODS Using Nanostring technology and comparative bioinformatics, we analysed the expression of 787 fibrosis-related genes in 36 ileal surgical specimens, 12 B2 and 24 B3, the latter including 12 cases with associated stricture[s] [B3s] and 12 without [B3o]. Quality control of extracted RNA was performed according to Nanostring parameters and principal component analysis for the distribution analysis. For the selection of the differentially expressed genes, a p-adjusted <0.05 and fold change ≤-1.5 or ≥1.5 were adopted. Quantitative polymerase chain reaction (qPCR) and immunohistochemistry analyses were used to validate selected differentially expressed genes. RESULTS We included 34 patients with B2 and B3 phenotypes, balanced for age at diagnosis, age at surgery, gender, Crohn's disease localisation, perianal disease, and therapy. Inflammation and fibrosis histopathological scoring were similar in all cases. B2 and B3 groups showed a very good clustering regarding 30 significantly differentially expressed genes, all being remarkably upregulated in B3. More than half of these genes were involved in Crohn's disease fibrogenesis, and eight differentially expressed genes were so in other organs. The most significantly active biological processes and pathways in penetrating disease were response to TGFβ and matrix organisation and degradation, as validated by immunohistochemistry. CONCLUSIONS Despite the histopathological similarities in fibrosis between stricturing and penetrating ileal Crohn's disease, their fibrosis-related transcriptomic profiles are distinct. Penetrating disease exhibits a distinctive transcriptomic landscape related to enhanced matrix remodelling.
Collapse
Affiliation(s)
- Helena Tavares de Sousa
- Gastroenterology Department, Algarve University Hospital Center [CHUA], Portimão, Portugal
- ABC-Algarve Biomedical Center, University of Algarve, Faro, Portugal
| | - Marta Ferreira
- Computer Science Department, Faculty of Sciences, University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto [IPATIMUP], Porto, Portugal
- Instituto de Investigação e Inovação em Saúde [i3S], University of Porto, Porto, Portugal
| | - Irene Gullo
- Institute of Molecular Pathology and Immunology, University of Porto [IPATIMUP], Porto, Portugal
- Instituto de Investigação e Inovação em Saúde [i3S], University of Porto, Porto, Portugal
- Department of Pathology, Centro Hospitalar de São João, Porto, Portugal
- Department of Pathology, Faculty of Medicine of the University of Porto [FMUP], Porto, Portugal
| | - Ana Mafalda Rocha
- Institute of Molecular Pathology and Immunology, University of Porto [IPATIMUP], Porto, Portugal
- Instituto de Investigação e Inovação em Saúde [i3S], University of Porto, Porto, Portugal
| | - Ana Pedro
- Instituto de Investigação e Inovação em Saúde [i3S], University of Porto, Porto, Portugal
| | - Dina Leitão
- Department of Pathology, Faculty of Medicine of the University of Porto [FMUP], Porto, Portugal
| | - Carla Oliveira
- Institute of Molecular Pathology and Immunology, University of Porto [IPATIMUP], Porto, Portugal
- Instituto de Investigação e Inovação em Saúde [i3S], University of Porto, Porto, Portugal
- Department of Pathology, Centro Hospitalar de São João, Porto, Portugal
| | - Fátima Carneiro
- Institute of Molecular Pathology and Immunology, University of Porto [IPATIMUP], Porto, Portugal
- Instituto de Investigação e Inovação em Saúde [i3S], University of Porto, Porto, Portugal
- Department of Pathology, Centro Hospitalar de São João, Porto, Portugal
- Department of Pathology, Faculty of Medicine of the University of Porto [FMUP], Porto, Portugal
| | - Fernando Magro
- Unit of Pharmacology and Therapeutics, Department of Biomedicine, Faculty of Medicine of the University of Porto [FMUP], Portugal
- Department of Gastroenterology, São João University Hospital Center, Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
8
|
Neupane YR, Yogananda TM, Rompicharla SVK, Selaru FM, Ensign LM. Emerging therapeutics for the management of intestinal fibrosis and strictures. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:107-139. [PMID: 39521597 DOI: 10.1016/bs.apha.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Chronic intestinal inflammation in patients with inflammatory bowel disease (IBD) can lead to the development of fibrosis and the formation of strictures. Endoscopic balloon dilation and surgical resection are currently the only available treatments for fibrotic strictures. However, both strategies are associated with potential complications and high rates of stricture recurrence, necessitating additional procedures and/or multiple surgical resections. IBD therapeutic modalities aimed at inflammation, including anti-inflammatory agents, such as corticosteroids, biologics and small molecules, have shown limited efficacy in altering the natural history of strictures, ameliorating fibrosis progression, or preventing recurrences. New and innovative therapeutic approaches targeted at fibrosis are urgently needed. Herein, we provide an overview of emerging therapeutics, including novel drug delivery systems, for the management of intestinal fibrosis and strictures.
Collapse
Affiliation(s)
- Yub Raj Neupane
- Center for Nanomedicine at the Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Thanuja Marasarakottige Yogananda
- Center for Nanomedicine at the Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sri Vishnu Kiran Rompicharla
- Center for Nanomedicine at the Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Florin M Selaru
- Division of Gastroenterology and Hepatology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Oncology, Sidney Kimmel Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; The Institute for Nanobiotechnology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Laura M Ensign
- Center for Nanomedicine at the Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Oncology, Sidney Kimmel Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Departments of Gynecology and Obstetrics, Pharmacology and Molecular Sciences, and Medicine (Infectious Diseases), The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Chemical and Biomolecular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
9
|
Bi GW, Wu ZG, Li Y, Wang JB, Yao ZW, Yang XY, Yu YB. Intestinal flora and inflammatory bowel disease: Causal relationships and predictive models. Heliyon 2024; 10:e38101. [PMID: 39381207 PMCID: PMC11458943 DOI: 10.1016/j.heliyon.2024.e38101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is significantly influenced by intestinal flora. Understanding the genetic and microbiotic interplay is crucial for IBD prediction and treatment. METHODS We used Mendelian randomization (MR), transcriptomic analysis, and machine learning techniques, integrating data from the MiBioGen Consortium and various GWAS datasets. SNPs associated with intestinal flora were mapped to genes, with LASSO regression refining gene selection. Differentially expressed genes (DEGs) and immune infiltration patterns were identified through transcriptomic analysis. Six machine learning models were used for predictive modeling. FINDINGS MR analysis identified 25 gut microbiota classifications causally related to IBD. SNP mapping and gene expression analysis highlighted 24 significant genes. Drug target MR and colocalization validated these genes' causal relationships with IBD. Key pathways identified included the PI3K-Akt signaling pathway and epithelial-mesenchymal transition. Immune infiltration analysis revealed distinct patterns between high and low LASSO score groups. Machine learning models demonstrated high predictive value, with soft voting enhancing reliability. INTERPRETATION By integrating MR, transcriptomic analysis, and sophisticated machine learning approaches, this study elucidates the causal relationships between intestinal flora and IBD. The application of machine learning not only enhanced predictive modeling but also offered new insights into IBD pathogenesis, highlighted potential therapeutic targets, and established a robust framework for predicting IBD onset.
Collapse
Affiliation(s)
- Guan-Wei Bi
- First Clinical College, Shandong University, Jinan, Shandong Province, PR China
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
| | - Zhen-Guo Wu
- First Clinical College, Shandong University, Jinan, Shandong Province, PR China
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
| | - Yu Li
- First Clinical College, Shandong University, Jinan, Shandong Province, PR China
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
| | - Jin-Bei Wang
- First Clinical College, Shandong University, Jinan, Shandong Province, PR China
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
| | - Zhi-Wen Yao
- First Clinical College, Shandong University, Jinan, Shandong Province, PR China
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
| | - Xiao-Yun Yang
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
| | - Yan-Bo Yu
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
- Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital, Shandong University, Jinan, Shandong Province, PR China
| |
Collapse
|
10
|
Artone S, Ciafarone A, Augello FR, Lombardi F, Cifone MG, Palumbo P, Cinque B, Latella G. Evaluation of the Antifibrotic Effects of Drugs Commonly Used in Inflammatory Intestinal Diseases on In Vitro Intestinal Cellular Models. Int J Mol Sci 2024; 25:8862. [PMID: 39201548 PMCID: PMC11354868 DOI: 10.3390/ijms25168862] [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: 07/10/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
The mechanism underlying intestinal fibrosis, the main complication of inflammatory bowel disease (IBD), is not yet fully understood, and there is no therapy to prevent or reverse fibrosis. We evaluated, in in vitro cellular models, the ability of different classes of drugs currently used in IBD to counteract two pivotal processes of intestinal fibrosis, the differentiation of intestinal fibroblasts to activated myofibroblasts using CCD-18Co cells, and the epithelial-to-mesenchymal transition (EMT) of intestinal epithelial cells using Caco-2 cells (IEC), both being processes induced by transforming growth factor-β1 (TGF-β1). The drugs tested included mesalamine, azathioprine, methotrexate, prednisone, methylprednisolone, budesonide, infliximab, and adalimumab. The expression of fibrosis and EMT markers (collagen-I, α-SMA, pSmad2/3, occludin) was assessed by Western blot analysis and by immunofluorescence. Of the drugs used, only prednisone, methylprednisolone, budesonide, and adalimumab were able to antagonize the pro-fibrotic effects induced by TGF-β1 on CCD-18Co cells, reducing the fibrosis marker expression. Methylprednisolone, budesonide, and adalimumab were also able to significantly counteract the TGF-β1-induced EMT process on Caco-2 IEC by increasing occludin and decreasing α-SMA expression. This is the first study that evaluates, using in vitro cellular models, the direct antifibrotic effects of drugs currently used in IBD, highlighting which drugs have potential antifibrotic effects.
Collapse
Affiliation(s)
- Serena Artone
- Department of Life, Health & Environmental Sciences, University of L’Aquila, Via Pompeo Spennati, Building Rita Levi Montalcini, Coppito, 67100 L’Aquila, Italy; (S.A.); (A.C.); (F.R.A.); (F.L.); (M.G.C.); (P.P.); (B.C.)
- PhD School in Medicine and Public Health, Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Alessia Ciafarone
- Department of Life, Health & Environmental Sciences, University of L’Aquila, Via Pompeo Spennati, Building Rita Levi Montalcini, Coppito, 67100 L’Aquila, Italy; (S.A.); (A.C.); (F.R.A.); (F.L.); (M.G.C.); (P.P.); (B.C.)
- PhD School in Health & Environmental Sciences, Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Francesca Rosaria Augello
- Department of Life, Health & Environmental Sciences, University of L’Aquila, Via Pompeo Spennati, Building Rita Levi Montalcini, Coppito, 67100 L’Aquila, Italy; (S.A.); (A.C.); (F.R.A.); (F.L.); (M.G.C.); (P.P.); (B.C.)
| | - Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L’Aquila, Via Pompeo Spennati, Building Rita Levi Montalcini, Coppito, 67100 L’Aquila, Italy; (S.A.); (A.C.); (F.R.A.); (F.L.); (M.G.C.); (P.P.); (B.C.)
| | - Maria Grazia Cifone
- Department of Life, Health & Environmental Sciences, University of L’Aquila, Via Pompeo Spennati, Building Rita Levi Montalcini, Coppito, 67100 L’Aquila, Italy; (S.A.); (A.C.); (F.R.A.); (F.L.); (M.G.C.); (P.P.); (B.C.)
| | - Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L’Aquila, Via Pompeo Spennati, Building Rita Levi Montalcini, Coppito, 67100 L’Aquila, Italy; (S.A.); (A.C.); (F.R.A.); (F.L.); (M.G.C.); (P.P.); (B.C.)
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L’Aquila, Via Pompeo Spennati, Building Rita Levi Montalcini, Coppito, 67100 L’Aquila, Italy; (S.A.); (A.C.); (F.R.A.); (F.L.); (M.G.C.); (P.P.); (B.C.)
| | - Giovanni Latella
- Department of Life, Health & Environmental Sciences, University of L’Aquila, Via Pompeo Spennati, Building Rita Levi Montalcini, Coppito, 67100 L’Aquila, Italy; (S.A.); (A.C.); (F.R.A.); (F.L.); (M.G.C.); (P.P.); (B.C.)
- Unit of Gastroenterology, Hepatology, and Nutrition, Department of Life, Health & Environmental Sciences, University of L’Aquila, Via Pompeo Spennati, Building Rita Levi Montalcini, Coppito, 67100 L’Aquila, Italy
| |
Collapse
|
11
|
Kim NY, Kim MO, Shin S, Kwon WS, Kim B, Lee JY, In Lee S. Effect of atractylenolide III on zearalenone-induced Snail1-mediated epithelial-mesenchymal transition in porcine intestinal epithelium. J Anim Sci Biotechnol 2024; 15:80. [PMID: 38845033 PMCID: PMC11157892 DOI: 10.1186/s40104-024-01038-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: 01/10/2024] [Accepted: 04/18/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND The intestinal epithelium performs essential physiological functions, such as nutrient absorption, and acts as a barrier to prevent the entry of harmful substances. Mycotoxins are prevalent contaminants found in animal feed that exert harmful effects on the health of livestock. Zearalenone (ZEA) is produced by the Fusarium genus and induces gastrointestinal dysfunction and disrupts the health and immune system of animals. Here, we evaluated the molecular mechanisms that regulate the effects of ZEA on the porcine intestinal epithelium. RESULTS Treatment of IPEC-J2 cells with ZEA decreased the expression of E-cadherin and increased the expression of Snai1 and Vimentin, which induced Snail1-mediated epithelial-to-mesenchymal transition (EMT). In addition, ZEA induces Snail-mediated EMT through the activation of TGF-β signaling. The treatment of IPEC-J2 cells with atractylenolide III, which were exposed to ZEA, alleviated EMT. CONCLUSIONS Our findings provide insights into the molecular mechanisms of ZEA toxicity in porcine intestinal epithelial cells and ways to mitigate it.
Collapse
Affiliation(s)
- Na Yeon Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeong-sangbuk-do, 37224, Republic of Korea
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeong-sangbuk-do, 37224, Republic of Korea
- Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Gyeongsangbuk-do, 37224, Republic of Korea
| | - Sangsu Shin
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeong-sangbuk-do, 37224, Republic of Korea
- Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Gyeongsangbuk-do, 37224, Republic of Korea
| | - Woo-Sung Kwon
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeong-sangbuk-do, 37224, Republic of Korea
- Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Gyeongsangbuk-do, 37224, Republic of Korea
| | - Bomi Kim
- National Institute for Korean Medicine Development, Gyeongsan, 38540, Republic of Korea
| | - Joon Yeop Lee
- National Institute for Korean Medicine Development, Gyeongsan, 38540, Republic of Korea
| | - Sang In Lee
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeong-sangbuk-do, 37224, Republic of Korea.
- Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Gyeongsangbuk-do, 37224, Republic of Korea.
| |
Collapse
|
12
|
Mignini I, Blasi V, Termite F, Esposto G, Borriello R, Laterza L, Scaldaferri F, Ainora ME, Gasbarrini A, Zocco MA. Fibrostenosing Crohn's Disease: Pathogenetic Mechanisms and New Therapeutic Horizons. Int J Mol Sci 2024; 25:6326. [PMID: 38928032 PMCID: PMC11204249 DOI: 10.3390/ijms25126326] [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: 04/30/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Bowel strictures are well recognized as one of the most severe complications in Crohn's disease, with variable impacts on the prognosis and often needing surgical or endoscopic treatment. Distinguishing inflammatory strictures from fibrotic ones is of primary importance due to the different therapeutic approaches required. Indeed, to better understand the pathogenesis of fibrosis, it is crucial to investigate molecular processes involving genetic factors, cytokines, alteration of the intestinal barrier, and epithelial and endothelial damage, leading to an increase in extracellular matrix synthesis, which ultimately ends in fibrosis. In such a complex mechanism, the gut microbiota also seems to play a role. A better comprehension of molecular processes underlying bowel fibrosis, in addition to radiological and histopathological findings, has led to the identification of high-risk patients for personalized follow-up and testing of new therapies, primarily in preclinical models, targeting specific pathways involving Transforming Growth Factor-β, interleukins, extracellular matrix balance, and gut microbiota. Our review aims to summarize current evidence about molecular factors involved in intestinal fibrosis' pathogenesis, paving the way for potential diagnostic biomarkers or anti-fibrotic treatments for stricturing Crohn's disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Maria Assunta Zocco
- CEMAD Digestive Diseases Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (I.M.); (V.B.); (G.E.); (R.B.); (L.L.); (F.S.); (M.E.A.); (A.G.)
| |
Collapse
|
13
|
Li Z, Chen Z, Zhang R, Lin J, Huang S, Shi K, Shen X, Xiang Z, Wang X, Huang L, Zheng Q, Liu X, Tan J, Chen M, Li Z, Mao R, Zhang X, Wang Y, Song X, Li X. Comparative analysis of [ 18F]F-FAPI PET/CT, [ 18F]F-FDG PET/CT and magnetization transfer MR imaging to detect intestinal fibrosis in Crohn's disease: A prospective animal model and human cohort study. Eur J Nucl Med Mol Imaging 2024; 51:1856-1868. [PMID: 38355741 DOI: 10.1007/s00259-024-06644-7] [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: 12/08/2023] [Accepted: 02/05/2024] [Indexed: 02/16/2024]
Abstract
PURPOSE Accurately and early detection of intestinal fibrosis in Crohn's disease (CD) is crucial for clinical management yet remains an unmet need. Fibroblast activation protein inhibitor (FAPI) PET/CT has emerged as a promising tool to assess fibrosis. We aimed to investigate the diagnostic capability of [18F]F-FAPI PET/CT in detecting intestinal fibrosis and compared it with[18F]F-FDG PET/CT and magnetization transfer MR imaging (MTI). METHODS Twenty-two rats underwent TNBS treatment to simulate fibrosis development, followed by three quantitative imaging sessions within one week. Mean and maximum standardized uptake values (SUVmean and SUVmax) were calculated on[18F]F-FAPI and [18F]F-FDG PET/CT, along with normalized magnetization transfer ratio on MTI. Intestinal fibrosis was assessed pathologically, with MTI serving as imaging standard for fibrosis. The diagnostic efficacy of imaging parameters in fibrosis was compared using pathological and imaging standards. Ten patients with 34 bowel strictures were prospectively recruited to validate their diagnostic performance, using the identical imaging protocol. RESULTS In CD patients, the accuracy of FAPI uptake (both AUCs = 0.87, both P ≤ 0.01) in distinguishing non-to-mild from moderate-to-severe fibrosis was higher than FDG uptake (both AUCs = 0.82, P ≤ 0.01) and comparable to MTI (AUCs = 0.90, P ≤ 0.001). In rats, FAPI uptake responded earlier to fibrosis development than FDG and MTI; consistently, during early phase, FAPI uptake showed a stronger correlation (SUVmean: R = 0.69) with pathological fibrosis than FDG (SUVmean: R = 0.17) and MTI (R = 0.52). CONCLUSION The diagnostic efficacy of [18F]F-FAPI PET/CT in detecting CD fibrosis is superior to [18F]F-FDG PET/CT and comparable to MTI, exhibiting great potential for early detection of intestinal fibrosis.
Collapse
Affiliation(s)
- Zhoulei Li
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Zhihui Chen
- Department of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
- Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-Sen University, 3 Foziling Road, Nanning, 530000, People's Republic of China
| | - Ruonan Zhang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Jinjiang Lin
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Siyun Huang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Kuangyu Shi
- Department of Nuclear Medicine, University of Bern, Freiburgstrasse 18, CH-3010, Bern, Switzerland
| | - Xiaodi Shen
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Zijun Xiang
- Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Xinyue Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Lili Huang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Qingzhu Zheng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Xubin Liu
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Jinyu Tan
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Ziping Li
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China
| | - Xiangsong Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China.
| | - Yangdi Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China.
| | - Xinming Song
- Department of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China.
| | - Xuehua Li
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
14
|
Yang HW, Chun-Yu Ho D, Liao HY, Liao YW, Fang CY, Ng MY, Yu CC, Lin FC. Resveratrol inhibits arecoline-induced fibrotic properties of buccal mucosal fibroblasts via miR-200a activation. J Dent Sci 2024; 19:1028-1035. [PMID: 38618058 PMCID: PMC11010603 DOI: 10.1016/j.jds.2023.06.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/26/2023] [Indexed: 04/16/2024] Open
Abstract
Background/purpose Oral submucous fibrosis (OSF) is a precancerous lesion in the oral cavity, commonly results from the Areca nut chewing habit. Arecoline, the main component of Areca nut, is known to stimulate the activation of myofibroblasts, which can lead to abnormal collagen I deposition. Meanwhile, Resveratrol is a non-flavonoid phenolic substance that can be naturally obtained from various berries and foods. Given that resveratrol has significant anti-fibrosis traits in other organs, but little is known about its effect on OSF, this study aimed to investigate the therapeutic impact of resveratrol on OSF and its underlying mechanism. Materials and methods The cytotoxicity of resveratrol was tested using normal buccal mucosal fibroblasts (BMFs). Myofibroblast phenotypes such as collagen contractile, enhanced migration, and wound healing capacities in dose-dependently resveratrol-treated fBMFs were examined. Results Current results showed that arecoline induced cell migration and contractile activity in BMFs as well as upregulated the expressions of α-SMA, type I collagen, and ZEB1 markers. Resveratrol intervention, on the other hand, was shown to inhibit arecoline-induced myofibroblast activation and reduce myofibroblast hallmarks and EMT markers. Additionally, resveratrol was also demonstrated to restore the downregulated miR-200a in the arecoline-stimulated cells. Conclusion In a nutshell, these findings implicate that resveratrol may have an inhibitory influence on arecoline-induced fibrosis via the regulation of miR-200a. Hence, resveratrol may be used as a therapeutic strategy for OSF intervention.
Collapse
Affiliation(s)
- Hui-Wen Yang
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Dennis Chun-Yu Ho
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Heng-Yi Liao
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Wen Liao
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chih-Yuan Fang
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Min Yee Ng
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Cheng-Chia Yu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Fu-Chen Lin
- Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan
| |
Collapse
|
15
|
Hausmann M, Seuwen K, de Vallière C, Busch M, Ruiz PA, Rogler G. Role of pH-sensing receptors in colitis. Pflugers Arch 2024; 476:611-622. [PMID: 38514581 PMCID: PMC11006753 DOI: 10.1007/s00424-024-02943-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/23/2024]
Abstract
Low pH in the gut is associated with severe inflammation, fibrosis, and colorectal cancer (CRC) and is a hallmark of active inflammatory bowel disease (IBD). Subsequently, pH-sensing mechanisms are of interest for the understanding of IBD pathophysiology. Tissue hypoxia and acidosis-two contributing factors to disease pathophysiology-are linked to IBD, and understanding their interplay is highly relevant for the development of new therapeutic options. One member of the proton-sensing G protein-coupled receptor (GPCR) family, GPR65 (T-cell death-associated gene 8, TDAG8), was identified as a susceptibility gene for IBD in a large genome-wide association study. In response to acidic extracellular pH, GPR65 induces an anti-inflammatory response, whereas the two other proton-sensing receptors, GPR4 and GPR68 (ovarian cancer G protein-coupled receptor 1, OGR1), mediate pro-inflammatory responses. Here, we review the current knowledge on the role of these proton-sensing receptors in IBD and IBD-associated fibrosis and cancer, as well as colitis-associated cancer (CAC). We also describe emerging small molecule modulators of these receptors as therapeutic opportunities for the treatment of IBD.
Collapse
Affiliation(s)
- Martin Hausmann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland.
| | - Klaus Seuwen
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Cheryl de Vallière
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Moana Busch
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Pedro A Ruiz
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| |
Collapse
|
16
|
Kadirvelu L, Sivaramalingam SS, Jothivel D, Chithiraiselvan DD, Karaiyagowder Govindarajan D, Kandaswamy K. A review on antimicrobial strategies in mitigating biofilm-associated infections on medical implants. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 6:100231. [PMID: 38510214 PMCID: PMC10951465 DOI: 10.1016/j.crmicr.2024.100231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024] Open
Abstract
Biomedical implants are crucial in providing support and functionality to patients with missing or defective body parts. However, implants carry an inherent risk of bacterial infections that are biofilm-associated and lead to significant complications. These infections often result in implant failure, requiring replacement by surgical restoration. Given these complications, it is crucial to study the biofilm formation mechanism on various biomedical implants that will help prevent implant failures. Therefore, this comprehensive review explores various types of implants (e.g., dental implant, orthopedic implant, tracheal stent, breast implant, central venous catheter, cochlear implant, urinary catheter, intraocular lens, and heart valve) and medical devices (hemodialyzer and pacemaker) in use. In addition, the mechanism of biofilm formation on those implants, and their pathogenesis were discussed. Furthermore, this article critically reviews various approaches in combating implant-associated infections, with a special emphasis on novel non-antibiotic alternatives to mitigate biofilm infections.
Collapse
Affiliation(s)
- Lohita Kadirvelu
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| | - Sowmiya Sri Sivaramalingam
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| | - Deepsikha Jothivel
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| | - Dhivia Dharshika Chithiraiselvan
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| | | | - Kumaravel Kandaswamy
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| |
Collapse
|
17
|
Xiao W, Hu C, Ni Y, Wang J, Jiao K, Zhou M, Li Z. 27-Hydroxycholesterol activates the GSK-3β/β-catenin signaling pathway resulting in intestinal fibrosis by inducing oxidative stress: effect of dietary interventions. Inflamm Res 2024; 73:289-304. [PMID: 38184500 DOI: 10.1007/s00011-023-01835-8] [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: 07/15/2023] [Revised: 11/30/2023] [Accepted: 12/09/2023] [Indexed: 01/08/2024] Open
Abstract
OBJECTIVE Intestinal fibrosis, a common and serious complication of inflammatory bowel disease (IBD), results from chronic inflammation. A high-cholesterol diet may be a risk factor for IBD and 27-hydroxylcholesterol (27HC) is the main human cholesterol metabolite. This study investigated whether 27HC can induce intestinal fibrosis. METHODS The effects of cholesterol and 27HC on intestinal fibrosis were assessed in zebrafish and human intestinal epithelial Caco-2 cells. RESULTS Cholesterol and 27HC induced intestinal inflammation and collagen deposition, inhibited E-cadherin (E-ca) expression in the intestinal epithelium, and promoted nuclear translocation of β-catenin in zebrafish. Cholesterol and 27HC up-regulated expression of COL-1, α-SMA, CTGF, TIMP1, N-cadherin, vimentin, glycogen synthesis kinase-3β (GSK-3β) and β-catenin, but inhibited E-ca, in Caco-2 cells. The expression of these proteins was inhibited by CYP27A1 knockdown and β-catenin knockdown. 27HC-induced nuclear translocation of β-catenin occurs in Caco-2 cells. p38, ERK, and AKT activate β-catenin and thereby participate in 27HC-induced epithelia-mesenchymal transition (EMT) and fibrosis. 27HC-increased oxidative stress and the fibrosis and EMT markers, the nuclear translocation of β-catenin, and the up-regulation of p-cell kinase proteins promoted by 27HC were inhibited by N-acetyl-L-cysteine (NAC). Folic acid (FA), resveratrol (RES), and NAC all ameliorated the 27HC-induced effects in Caco-2 cells and zebrafish. CONCLUSION A high-cholesterol diet caused intestinal fibrosis in zebrafish, mediated by a major cholesterol metabolite, 27HC. 27HC increased oxidative stress and activated p38, ERK, AKT, and β-catenin, leading to EMT of epithelial cells and intestinal fibrosis. FA and RES both ameliorated intestinal fibrosis by restraining 27HC-induced β-catenin activation.
Collapse
Affiliation(s)
- Wei Xiao
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chunyan Hu
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yifan Ni
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jie Wang
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Kailin Jiao
- Department of Nutrition, The Second Affiliated Hospital, Air Force Medical University, Xi'an, China.
| | - Ming Zhou
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Zhong Li
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
18
|
Niu W, Liu Q, Huo X, Luo Y, Zhang X. TL1A promotes metastasis and EMT process of colorectal cancer. Heliyon 2024; 10:e24392. [PMID: 38312710 PMCID: PMC10835226 DOI: 10.1016/j.heliyon.2024.e24392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 12/15/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
BACKGROUND Metastasis is the major problem of colorectal cancer (CRC) and is correlated with the high mortality. Tumor necrosis factor-like cytokine 1A (TL1A) is a novel regulatory factor for inflammatory diseases. This work aimed to investigate the role of TL1A in CRC metastasis. METHOD AOM/DSS-induced mouse model, xenograft tumor model and metastasis murine model were established to mimic the colitis-associated CRC and investigate CRC growth and metastasis in vivo. Colon tissues were assessed by hematoxylin/eosin (HE) staining and immunohistochemistry (IHC). CRC cell metastasis in vivo was observed using in vivo imaging system (IVIS). Cell viability and proliferation were examined using cell counting kit 8 (CCK-8) and EdU experiments. The expression of tumor growth factor β (TGFβ) and metastatic biomarkers were detected using western blotting experiment. The in vitro cell metastasis was measured by Transwell. RESULTS Knockdown of TL1A notably suppressed the generation of colonic tumors in azoxymethane/dextran sodium sulfate (AOM/DSS) model, suppressed in vivo CRC cell growth, as well as lung and liver metastasis. The inflammation response and inflammatory cell infiltration in tumor sites were decreased by TL1A depletion. The in vitro CRC cell growth and metastasis was also suppressed by shTL1A, along with altered expression of epithelial mesenchymal transition (EMT) biomarkers. TL1A depletion suppressed the level of the TGF-β1 receptor (TβRI) and phosphorylation of Smad3 in CRC cells. Stimulation with TGF-β recovered the CRC cell migration and invasion that suppressed by shTL1A. CONCLUSION Our work implicated TL1A as a promoter of CRC generation and metastasis and defines TGF-β/Smad3 signaling as mediator of TL1A-regualated CRC cell metastasis.
Collapse
Affiliation(s)
- Weiwei Niu
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| | - Qian Liu
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| | - Xiaoxia Huo
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| | - Yuxin Luo
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| | - Xiaolan Zhang
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| |
Collapse
|
19
|
Gorreja F, Bendix M, Rush STA, Maasfeh L, Savolainen O, Dige A, Agnholt J, Öhman L, Magnusson MK. Fecal Supernatants from Patients with Crohn's Disease Induce Inflammatory Alterations in M2 Macrophages and Fibroblasts. Cells 2023; 13:60. [PMID: 38201264 PMCID: PMC10777926 DOI: 10.3390/cells13010060] [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: 11/22/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Intestinal macrophages and fibroblasts act as microenvironmental sentinels mediating inflammation and disease progression in Crohn's disease (CD). We aimed to establish the effects of fecal supernatants (FSs) from patients with CD on macrophage and fibroblast phenotype and function. FS were obtained by ultracentrifugation, and the metabolites were analyzed. Monocyte-derived M2 macrophages and fibroblasts were conditioned with FS, and secreted proteins, surface proteins and gene expression were analyzed. M2 macrophage efferocytosis was evaluated. Patients with CD (n = 15) had a skewed fecal metabolite profile compared to healthy subjects (HS, n = 10). FS from CD patients (CD-FS) induced an anti-inflammatory response in M2 macrophages with higher expression of IL-10, IL1RA and CD206 as compared to healthy FS (HS-FS) while the efferocytotic capacity was unaltered. CD-FS did not affect extracellular matrix production from fibroblasts, but increased expression of the pro-inflammatory proteins IL-6 and MCP-1. Conditioned media from M2 macrophages treated with CD-FS modulated gene expression in fibroblasts for TGFβ superfamily members and reduced IL-4 expression compared to HS-FS. We show that M2 macrophages and fibroblasts react abnormally to the fecal microenvironment of CD patients, resulting in altered protein expression related to inflammation but not fibrosis. This implies that the gut microbiota and its metabolites have an important role in the generation and/or perpetuation of inflammation in CD.
Collapse
Affiliation(s)
- Frida Gorreja
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (F.G.); (S.T.A.R.); (L.M.); (L.Ö.)
| | - Mia Bendix
- Medical Department, Randers Regional Hospital, 8930 Randers, Denmark;
| | - Stephen T. A. Rush
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (F.G.); (S.T.A.R.); (L.M.); (L.Ö.)
| | - Lujain Maasfeh
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (F.G.); (S.T.A.R.); (L.M.); (L.Ö.)
| | - Otto Savolainen
- Chalmers Mass Spectrometry Infrastructure, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden;
| | - Anders Dige
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, 8200 Aarhus, Denmark; (A.D.); (J.A.)
| | - Jorgen Agnholt
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, 8200 Aarhus, Denmark; (A.D.); (J.A.)
| | - Lena Öhman
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (F.G.); (S.T.A.R.); (L.M.); (L.Ö.)
| | - Maria K. Magnusson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (F.G.); (S.T.A.R.); (L.M.); (L.Ö.)
| |
Collapse
|
20
|
Kim SW, Lee JY, Lee HC, Ahn JB, Kim JH, Park IS, Cheon JH, Kim DH. Downregulation of Heat Shock Protein 72 Contributes to Fibrostenosis in Crohn's Disease. Gut Liver 2023; 17:905-915. [PMID: 36814356 PMCID: PMC10651382 DOI: 10.5009/gnl220308] [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: 07/18/2022] [Revised: 10/21/2022] [Accepted: 11/29/2022] [Indexed: 02/24/2023] Open
Abstract
Background/Aims Crohn's disease (CD) with recurrent inflammation can cause intestinal fibrostenosis due to dysregulated deposition of extracellular matrix. However, little is known about the pathogenesis of fibrostenosis. Here, we performed a differential proteomic analysis between normal, inflamed, and fibrostenotic specimens of patients with CD and investigated the roles of the candidate proteins in myofibroblast activation and fibrosis. Methods We performed two-dimensional difference gel electrophoresis and identified candidate proteins using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and orbitrap liquid chromatography-mass spectrometry. We also verified the levels of candidate proteins in clinical specimens and examined their effects on 18Co myofibroblasts and Caco-2 intestinal epithelial cells. Results We identified five of 30 proteins (HSP72, HSPA5, KRT8, PEPCK-M, and FABP6) differentially expressed in fibrostenotic CD. Among these proteins, the knockdown of heat shock protein 72 (HSP72) promoted the activation and wound healing of myofibroblasts. Moreover, knockdown of HSP72 induced the epithelial-mesenchymal transition of intestinal epithelial cells by reducing E-cadherin and inducing fibronectin and α-smooth muscle actin, which contribute to fibrosis. Conclusions HSP72 is an important mediator that regulates myofibroblasts and epithelial-mesenchymal transition in fibrosis of CD, suggesting that HSP72 can serve as a target for antifibrotic therapy.
Collapse
Affiliation(s)
- Seung Won Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Seoul, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jae-Young Lee
- Department of Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Han Cheol Lee
- Department of Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Bum Ahn
- Department of Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyung Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Seoul, Korea
| | - I Seul Park
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Seoul, Korea
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Seoul, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Duk Hwan Kim
- Digestive Disease Center, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| |
Collapse
|
21
|
Fazilaty H, Basler K. Reactivation of embryonic genetic programs in tissue regeneration and disease. Nat Genet 2023; 55:1792-1806. [PMID: 37904052 DOI: 10.1038/s41588-023-01526-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 09/11/2023] [Indexed: 11/01/2023]
Abstract
Embryonic genetic programs are reactivated in response to various types of tissue damage, providing cell plasticity for tissue regeneration or disease progression. In acute conditions, these programs remedy the damage and then halt to allow a return to homeostasis. In chronic situations, including inflammatory diseases, fibrosis and cancer, prolonged activation of embryonic programs leads to disease progression and tissue deterioration. Induction of progenitor identity and cell plasticity, for example, epithelial-mesenchymal plasticity, are critical outcomes of reactivated embryonic programs. In this Review, we describe molecular players governing reactivated embryonic genetic programs, their role during disease progression, their similarities and differences and lineage reversion in pathology and discuss associated therapeutics and drug-resistance mechanisms across many organs. We also discuss the diversity of reactivated programs in different disease contexts. A comprehensive overview of commonalities between development and disease will provide better understanding of the biology and therapeutic strategies.
Collapse
Affiliation(s)
- Hassan Fazilaty
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland.
| | - Konrad Basler
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| |
Collapse
|
22
|
Li J, Xu JZ, Dou B, Huang TF, Chen J, Wang TM, Ouyang HJ. Circ_0001666 upregulation promotes intestinal epithelial cell fibrosis in pediatric Crohn's disease via the SRSF1/BMP7 axis. Kaohsiung J Med Sci 2023; 39:966-977. [PMID: 37530654 DOI: 10.1002/kjm2.12734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 08/03/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) is closely associated with Crohn's disease (CD) related intestinal fibrosis, a condition whose prevalence is increasing annually among children. Recently, the CD marker gene microarray screening revealed an upregulation of circ_0001666 in the colon tissues of CD patients, but its underlying mechanisms remain unclear. In this study, we explored the molecular mechanism of circ_0001666 in regulating EMT-mediated fibrosis in CD in vitro. The levels of circ_0001666 and EMT-associated proteins were assessed in CD clinical samples, and a CD cell model was established using TGF-β1 to induce human intestinal epithelial cells (HIECs). Additionally, the expression levels of genes and proteins related to EMT and fibrosis were analyzed by quantitative real-time PCR and western blot, cell migration, and invasion were assessed via wound healing assay and transwell, respectively, and RNA pull-down and RNA immunoprecipitation assays were performed to verify the relationship between SRSF1 and BMP7 or circ_0001666. Circ_0001666 was overexpressed in the intestinal mucosal tissues of CD patients and was positively correlated with EMT. Silencing circ_0001666 inhibited the migration, invasion, EMT, and fibrosis of HIECs induced by TGF-β1. Mechanistically, circ_0001666 regulated BMP7 expression by interacting with SRSF1. Furthermore, the effects of inhibiting circ_0001666 on HIECs could be partially reversed by overexpressing SRSF1 or silencing BMP7. Collectively, circ_0001666 regulates TGF-β1-induced HIEC migration, invasion, EMT, and fibrosis. Circ_0001666 also promoted EMT-mediated fibrosis by interacting with SRSF1 to accelerate BMP7 mRNA decay. These findings provide new insights into the pathogenesis of CD and suggest that circ_0001666 might be a potential therapeutic target for CD.
Collapse
Affiliation(s)
- Jun Li
- Department of Pediatrics, Changsha Maternal and Child Health Hospital, Changsha, Hunan Province, China
| | - Ji-Zhi Xu
- Department of Pediatrics, Changsha Maternal and Child Health Hospital, Changsha, Hunan Province, China
| | - Bo Dou
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Teng-Fei Huang
- Department of Pediatrics, Changsha Maternal and Child Health Hospital, Changsha, Hunan Province, China
| | - Jie Chen
- Department of Pediatrics, Changsha Maternal and Child Health Hospital, Changsha, Hunan Province, China
| | - Tuan-Mei Wang
- Department of Pediatrics, Changsha Maternal and Child Health Hospital, Changsha, Hunan Province, China
| | - Hong-Juan Ouyang
- Department of Digestive Nutrition, Hunan Children's Hospital, Changsha, Hunan Province, China
| |
Collapse
|
23
|
Macias-Ceja DC, Mendoza-Ballesteros MT, Ortega-Albiach M, Barrachina MD, Ortiz-Masià D. Role of the epithelial barrier in intestinal fibrosis associated with inflammatory bowel disease: relevance of the epithelial-to mesenchymal transition. Front Cell Dev Biol 2023; 11:1258843. [PMID: 37822869 PMCID: PMC10562728 DOI: 10.3389/fcell.2023.1258843] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/14/2023] [Indexed: 10/13/2023] Open
Abstract
In inflammatory bowel disease (IBD), chronic inflammation in the gastrointestinal tract can lead to tissue damage and remodelling, which can ultimately result in fibrosis. Prolonged injury and inflammation can trigger the activation of fibroblasts and extracellular matrix (ECM) components. As fibrosis progresses, the tissue becomes increasingly stiff and less functional, which can lead to complications such as intestinal strictures, obstructive symptoms, and eventually, organ dysfunction. Epithelial cells play a key role in fibrosis, as they secrete cytokines and growth factors that promote fibroblast activation and ECM deposition. Additionally, epithelial cells can undergo a process called epithelial-mesenchymal transition, in which they acquire a more mesenchymal-like phenotype and contribute directly to fibroblast activation and ECM deposition. Overall, the interactions between epithelial cells, immune cells, and fibroblasts play a critical role in the development and progression of fibrosis in IBD. Understanding these complex interactions may provide new targets for therapeutic interventions to prevent or treat fibrosis in IBD. In this review, we have collected and discussed the recent literature highlighting the contribution of epithelial cells to the pathogenesis of the fibrotic complications of IBD, including evidence of EMT, the epigenetic control of the EMT, the potential influence of the intestinal microbiome in EMT, and the possible therapeutic strategies to target EMT. Finally we discuss the pro-fibrotic interactions epithelial-immune cells and epithelial-fibroblasts cells.
Collapse
Affiliation(s)
- Dulce C. Macias-Ceja
- Departamento de Farmacología and CIBEREHD, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | | | | | - M. Dolores Barrachina
- Departamento de Farmacología and CIBEREHD, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Dolores Ortiz-Masià
- Departamento de Farmacología and CIBEREHD, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
24
|
Gunavathy N, Balaji R, Kumaravel V. Association of TCF7L2 Variants in Type 2 Diabetes Mellitus with Hypertriglyceridemia - A Case-Control Study. Indian J Endocrinol Metab 2023; 27:346-350. [PMID: 37867984 PMCID: PMC10586556 DOI: 10.4103/ijem.ijem_35_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/03/2023] [Accepted: 05/21/2023] [Indexed: 10/24/2023] Open
Abstract
Background Type 2 Diabetes Mellitus (T2DM) is a chronic metabolic condition involving various genetic and environmental factors leading to impaired insulin secretion, resulting in hyperglycemia. The transcription factor 7-like 2 (TCF7L2) gene is an element of the Wnt signaling pathway that plays an important role in glucose and lipid metabolism. The aim of this study is to evaluate the association of TCF7L2 rs7903146 and rs12255372 polymorphisms in T2DM with hypertriglyceridemia. Methods We investigated the effect of rs7903146 and rs12255372 on T2DM with high triglyceride (TG) levels in 60 patients and 20 controls. The anthropometric measurements and biochemical tests were assessed. Peripheral blood samples were collected, and genomic DNA was extracted. The genotyping of TCF7L2 polymorphisms was carried out using polymerase chain reaction (PCR)-based direct sequencing and allele-specific PCR methods. The T2DM patients and controls were compared by means of the t-test, Chi-square test, odds ratio (OR), and 95% confidence interval (CI) using Epi Info v7. Results The HbA1c was found to be 9.7 ± 2.1 and 5.4 ± 0.5% in patients and controls, respectively. The average TG levels (P < 0.005) in patients were 205.2 ± 145.7 and 106.4 ± 27.4mg/dl in controls. Significant evidence of association was found in T2DM patients having high TG levels with rs7903146 CT/TT (OR: 4.89; P = 0.0105) and rs12255372 GT/TT (OR: 5.23; P = 0.0101) genotypes when compared to controls. Conclusion The results of this study show that TCF7L2 rs7903146 CT/TT and rs12255372 GT/TT genotypes are significantly associated with the risk of hypertriglyceridemia in individuals with T2DM among the studied population.
Collapse
Affiliation(s)
- Nagarajan Gunavathy
- Department of Molecular Genetics, Alpha Health Foundation, Madurai, Tamil Nadu, India
| | - Ramanathan Balaji
- Department of Molecular Genetics, Alpha Health Foundation, Madurai, Tamil Nadu, India
| | - Velayutham Kumaravel
- Department of Molecular Genetics, Alpha Health Foundation, Madurai, Tamil Nadu, India
| |
Collapse
|
25
|
Gao Y, Lu LJ, Zhang ZZ, Yang X, Du J, Wen K, Huang H, Wang XP, Sun XL. Xue-jie-San prevents the early development of colitis-associated intestinal fibrosis by blocking Notch1 and FGL1 signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2023; 315:116678. [PMID: 37263315 DOI: 10.1016/j.jep.2023.116678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/07/2023] [Accepted: 05/21/2023] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xue-Jie-San (XJS), as a traditional Chinese herb prescription, has satisfactory effects on improving clinical symptoms and facilitating the healing of intestinal ulcers in patients with Crohn's disease (CD). This motivates the application of XJS on CD-associated complications. AIM OF THE STUDY Intestinal fibrosis is a debilitating complication of CD. Currently, there is no effective medication available for preventing or reversing CD-related intestinal fibrosis. This study aimed to assess the efficacy and underlying mechanisms of XJS in the treatment of colitis-associated intestinal fibrosis. MATERIALS AND METHODS A rat model of CD-related intestinal fibrosis was induced by 2,4,6-trinitrobenzene sulfonic acid administration and treated with XJS. The pathological changes of intestinal fibrosis were evaluated using Masson staining. Collagen deposition and epithelial-to-mesenchymal transition (EMT) were verified by immunohistochemical staining and western blot analysis. Endothelial-to-mesenchymal transition (EndoMT) was assessed with immunofluorescence and immunohistochemical staining as well as western blot analysis. Transmission electron microscopy was utilized to observe autophagosomes. The levels of autophagy-related proteins were detected via immunofluorescence staining and western blot. Finally, the mTOR/ULK1 signaling pathway regulated by Notch1 or FGL1 was analyzed by western blot. RESULTS The results found that XJS ameliorated intestinal fibrosis through reducing the deposition of collagens such as Collagen 1 and Collagen 3. XJS inhibited the EMT process by increasing E-cadherin levels and decreasing the expressions of N-cadherin, Vimentin and Snail, which played a crucial role in collagen secretion and intestinal fibrosis. In addition, XJS also repressed the EndoMT process as reflected by the upregulation of CD31 and VE-cadherin levels and the downregulation of FSP1 and α-SMA expressions. Autophagy was activated following XJS treatment via suppression of the mTOR/ULK1 signaling pathway. Furthermore, XJS acted as an inhibitor of Notch1 and FGL1 signals, both of which regulated the mTOR signaling. CONCLUSIONS Our findings validated that XJS prevented the early development of CD-related intestinal fibrosis by blocking the Notch1 and FGL1 signaling pathways to activate autophagy and thereby inhibit EMT and EndoMT.
Collapse
Affiliation(s)
- Ying Gao
- Department of Colorectal Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, China.
| | - Li-Juan Lu
- Department of Gynaecology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, China.
| | - Zhao-Zheng Zhang
- Department of Colorectal Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, China.
| | - Xiao Yang
- Department of Colorectal Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, China.
| | - Jun Du
- Department of Colorectal Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, China.
| | - Ke Wen
- Department of Colorectal Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, China.
| | - Hua Huang
- Department of Anorectal Surgery, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, 215500, China.
| | - Xiao-Peng Wang
- Department of Colorectal Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, China.
| | - Xue-Liang Sun
- Department of Colorectal Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, China.
| |
Collapse
|
26
|
Burr AHP, Ji J, Ozler K, Mentrup HL, Eskiocak O, Yueh B, Cumberland R, Menk AV, Rittenhouse N, Marshall CW, Chiaranunt P, Zhang X, Mullinax L, Overacre-Delgoffe A, Cooper VS, Poholek AC, Delgoffe GM, Mollen KP, Beyaz S, Hand TW. Excess Dietary Sugar Alters Colonocyte Metabolism and Impairs the Proliferative Response to Damage. Cell Mol Gastroenterol Hepatol 2023; 16:287-316. [PMID: 37172822 PMCID: PMC10394273 DOI: 10.1016/j.jcmgh.2023.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND & AIMS The colonic epithelium requires continuous renewal by crypt resident intestinal stem cells (ISCs) and transit-amplifying (TA) cells to maintain barrier integrity, especially after inflammatory damage. The diet of high-income countries contains increasing amounts of sugar, such as sucrose. ISCs and TA cells are sensitive to dietary metabolites, but whether excess sugar affects their function directly is unknown. METHODS Here, we used a combination of 3-dimensional colonoids and a mouse model of colon damage/repair (dextran sodium sulfate colitis) to show the direct effect of sugar on the transcriptional, metabolic, and regenerative functions of crypt ISCs and TA cells. RESULTS We show that high-sugar conditions directly limit murine and human colonoid development, which is associated with a reduction in the expression of proliferative genes, adenosine triphosphate levels, and the accumulation of pyruvate. Treatment of colonoids with dichloroacetate, which forces pyruvate into the tricarboxylic acid cycle, restored their growth. In concert, dextran sodium sulfate treatment of mice fed a high-sugar diet led to massive irreparable damage that was independent of the colonic microbiota and its metabolites. Analyses on crypt cells from high-sucrose-fed mice showed a reduction in the expression of ISC genes, impeded proliferative potential, and increased glycolytic potential without a commensurate increase in aerobic respiration. CONCLUSIONS Taken together, our results indicate that short-term, excess dietary sucrose can directly modulate intestinal crypt cell metabolism and inhibit ISC/TA cell regenerative proliferation. This knowledge may inform diets that better support the treatment of acute intestinal injury.
Collapse
Affiliation(s)
- Ansen H P Burr
- Richard King Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Junyi Ji
- School of Medicine, Tsinghua University, Beijing, China
| | - Kadir Ozler
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Heather L Mentrup
- Department of Surgery, University of Pittsburgh School of Medicine. University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Onur Eskiocak
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Brian Yueh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Rachel Cumberland
- Tumor Microenvironment Center, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Ashley V Menk
- Tumor Microenvironment Center, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Natalie Rittenhouse
- Richard King Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Chris W Marshall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Pailin Chiaranunt
- Richard King Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiaoyi Zhang
- Richard King Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Gastroenterology, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center Children's Hospital
| | - Lauren Mullinax
- Richard King Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Gastroenterology, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center Children's Hospital
| | - Abigail Overacre-Delgoffe
- Richard King Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Vaughn S Cooper
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Amanda C Poholek
- Richard King Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Greg M Delgoffe
- Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania; Tumor Microenvironment Center, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Kevin P Mollen
- Department of Surgery, University of Pittsburgh School of Medicine. University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Timothy W Hand
- Richard King Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
27
|
Solitano V, Dal Buono A, Gabbiadini R, Wozny M, Repici A, Spinelli A, Vetrano S, Armuzzi A. Fibro-Stenosing Crohn's Disease: What Is New and What Is Next? J Clin Med 2023; 12:jcm12093052. [PMID: 37176493 PMCID: PMC10179180 DOI: 10.3390/jcm12093052] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/03/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Fibro-stenosing Crohn's disease (CD) is a common disease presentation that leads to impaired quality of life and often requires endoscopic treatments or surgery. From a pathobiology perspective, the conventional view that intestinal fibro-stenosis is an irreversible condition has been disproved. Currently, there are no existing imaging techniques that can accurately quantify the amount of fibrosis within a stricture, and managing patients is challenging, requiring a multidisciplinary team. Novel therapies targeting different molecular components of the fibrotic pathways are increasing regarding other diseases outside the gut. However, a large gap between clinical need and the lack of anti-fibrotic agents in CD remains. This paper reviews the current state of pathobiology behind fibro-stenosing CD, provides an updated diagnostic and therapeutic approach, and finally, focuses on clinical trial endpoints and possible targets of anti-fibrotic therapies.
Collapse
Affiliation(s)
- Virginia Solitano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
- Division of Gastroenterology, Department of Medicine, Western University, London, ON N6A 4V2, Canada
| | - Arianna Dal Buono
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Roberto Gabbiadini
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Marek Wozny
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Alessandro Repici
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
- Department of Endoscopy, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Antonino Spinelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
- Division of Colon and Rectal Surgery, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Stefania Vetrano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Alessandro Armuzzi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| |
Collapse
|
28
|
Rizzo G, Rubbino F, Elangovan S, Sammarco G, Lovisa S, Restelli S, Pineda Chavez SE, Massimino L, Lamparelli L, Paulis M, Maroli A, Roda G, Shalaby M, Carvello M, Foppa C, Drummond SP, Spaggiari P, Ungaro F, Spinelli A, Malesci A, Repici A, Day AJ, Armuzzi A, Danese S, Vetrano S. Dysfunctional Extracellular Matrix Remodeling Supports Perianal Fistulizing Crohn's Disease by a Mechanoregulated Activation of the Epithelial-to-Mesenchymal Transition. Cell Mol Gastroenterol Hepatol 2022; 15:741-764. [PMID: 36521659 PMCID: PMC9898761 DOI: 10.1016/j.jcmgh.2022.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND AIMS Perianal fistula represents one of the most disabling manifestations of Crohn's disease (CD) due to complete destruction of the affected mucosa, which is replaced by granulation tissue and associated with changes in tissue organization. To date, the molecular mechanisms underlying perianal fistula formation are not well defined. Here, we dissected the tissue changes in the fistula area and addressed whether a dysregulation of extracellular matrix (ECM) homeostasis can support fistula formation. METHODS Surgical specimens from perianal fistula tissue and the surrounding region of fistulizing CD were analyzed histologically and by RNA sequencing. Genes significantly modulated were validated by real-time polymerase chain reaction, Western blot, and immunofluorescence assays. The effect of the protein product of TNF-stimulated gene-6 (TSG-6) on cell morphology, phenotype, and ECM organization was investigated with endogenous lentivirus-induced overexpression of TSG-6 in Caco-2 cells and with exogenous addition of recombinant human TSG-6 protein to primary fibroblasts from region surrounding fistula. Proliferative and migratory assays were performed. RESULTS A markedly different organization of ECM was found across fistula and surrounding fistula regions with an increased expression of integrins and matrix metalloproteinases and hyaluronan (HA) staining in the fistula, associated with increased newly synthesized collagen fibers and mechanosensitive proteins. Among dysregulated genes associated with ECM, TNFAI6 (gene encoding for TSG-6) was as significantly upregulated in the fistula compared with area surrounding fistula, where it promoted the pathological formation of complexes between heavy chains from inter-alpha-inhibitor and HA responsible for the formation of a crosslinked ECM. There was a positive correlation between TNFAI6 expression and expression of mechanosensitive genes in fistula tissue. The overexpression of TSG-6 in Caco-2 cells promoted migration, epithelial-mesenchymal transition, transcription factor SNAI1, and HA synthase (HAs) levels, while in fibroblasts, isolated from the area surrounding the fistula, it promoted an activated phenotype. Moreover, the enrichment of an HA scaffold with recombinant human TSG-6 protein promoted collagen release and increase of SNAI1, ITGA4, ITGA42B, and PTK2B genes, the latter being involved in the transduction of responses to mechanical stimuli. CONCLUSIONS By mediating changes in the ECM organization, TSG-6 triggers the epithelial-mesenchymal transition transcription factor SNAI1 through the activation of mechanosensitive proteins. These data point to regulators of ECM as new potential targets for the treatment of CD perianal fistula.
Collapse
Affiliation(s)
- Giulia Rizzo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Federica Rubbino
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | | | - Giusy Sammarco
- IBD Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Sara Lovisa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy; IBD Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Silvia Restelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | | | - Luca Massimino
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy; Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Luigi Lamparelli
- IBD Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Marianna Paulis
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Institute of Genetic and Biomedical Research, UOS Milan, National Research Council of Italy, Milan, Italy
| | - Annalisa Maroli
- Colon and Rectal Surgery Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Giulia Roda
- IBD Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Mohammad Shalaby
- IBD Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Michele Carvello
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy; Colon and Rectal Surgery Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Caterina Foppa
- Colon and Rectal Surgery Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Sheona P Drummond
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Paola Spaggiari
- Department of Pathology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Federica Ungaro
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy; Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, Milan, Italy; Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Antonino Spinelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy; Colon and Rectal Surgery Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Alberto Malesci
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy; Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Alessandro Repici
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy; Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Clinical and Research Center, Rozzano, Italy
| | - Anthony J Day
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Alessandro Armuzzi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy; IBD Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Silvio Danese
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy; Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, Milan, Italy; Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Stefania Vetrano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy; IBD Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy.
| |
Collapse
|
29
|
Liu J, Gong W, Liu P, Li Y, Jiang H, Wu X, Zhao Y, Ren J. Macrophages-microenvironment crosstalk in fibrostenotic inflammatory bowel disease: from basic mechanisms to clinical applications. Expert Opin Ther Targets 2022; 26:1011-1026. [PMID: 36573664 DOI: 10.1080/14728222.2022.2161889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Intestinal fibrosis is a common complication of Inflammatory Bowel Disease (IBD) with no available drugs. The current therapeutic principle is surgical intervention as the core. Intestinal macrophages contribute to both the progression of inflammation and fibrosis. Understanding the role of macrophages in the intestinal microenvironment could bring new hope for fibrosis prevention or even reversal. AREAS COVERED This article reviewed the most relevant reports on macrophage in the field of intestinal fibrosis. The authors discussed current opinions about how intestinal macrophages function and interact with surrounding mediators during inflammation resolution and fibrostenotic IBD. Based on biological mechanisms findings, authors summarized related clinical trial outcomes. EXPERT OPINION The plasticity of intestinal macrophages allows them to undergo dramatic alterations in their phenotypes or functions when exposed to gastrointestinal environmental stimuli. They exhibit distinct metabolic characteristics, secrete various cytokines, express unique surface markers, and transmit different signals. Nevertheless, the specific mechanism through which the intestinal macrophages contribute to intestinal fibrosis remains unclear. It should further elucidate a novel therapeutic approach by targeting macrophages, especially distinct mechanisms in specific subgroups of macrophages involved in the progression of fibrogenesis in IBD.
Collapse
Affiliation(s)
- Juanhan Liu
- Department of General Surgery, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, 210002, Nanjing, P. R. China
| | - Wenbin Gong
- Department of General Surgery, Southeast University, 210096, Nanjing, P. R. China
| | - Peizhao Liu
- Department of General Surgery, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, 210002, Nanjing, P. R. China
| | - Yangguang Li
- Department of General Surgery, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, 210002, Nanjing, P. R. China
| | - Haiyang Jiang
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, 210019, Nanjing, P. R. China
| | - Xiuwen Wu
- Department of General Surgery, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, 210002, Nanjing, P. R. China
| | - Yun Zhao
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, 210019, Nanjing, P. R. China
| | - Jianan Ren
- Department of General Surgery, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, 210002, Nanjing, P. R. China
| |
Collapse
|
30
|
Yao H, Tang G. Macrophages in intestinal fibrosis and regression. Cell Immunol 2022; 381:104614. [PMID: 36182587 DOI: 10.1016/j.cellimm.2022.104614] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 11/03/2022]
Abstract
Intestinal macrophages are heterogenous cell populations with different developmental ontogeny and tissue anatomy. The concerted actions of intestinal macrophage subsets are critical to maintaining tissue homeostasis. However, the dysregulation of macrophages following tissue injury or chronic inflammation could also lead to intestinal fibrosis, with few treatment options in the clinic. In this review, we will characterize the features of intestinal macrophages in light of the latest advances in lineage tracing and single-cell sequencing technology. The roles of macrophages in distinct stages of intestinal fibrosis would be also elaborated. Finally, based on the reciprocal interaction between macrophages and intestinal fibrosis, we will propose the potential macrophage targeting anti-intestinal fibrosis therapies.
Collapse
Affiliation(s)
- Hui Yao
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China; National Center for Stomatology, Shanghai 200011, China; National Clinical Research Center for Oral Diseases, Shanghai 200011, China; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Guoyao Tang
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China; National Center for Stomatology, Shanghai 200011, China; National Clinical Research Center for Oral Diseases, Shanghai 200011, China; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China.
| |
Collapse
|
31
|
Wang Z, Zhou H, Cheng F, Zhang Z, Long S. MiR-21 regulates epithelial-mesenchymal transition in intestinal fibrosis of Crohn's disease by targeting PTEN/mTOR. Dig Liver Dis 2022; 54:1358-1366. [PMID: 35504804 DOI: 10.1016/j.dld.2022.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 04/08/2022] [Accepted: 04/13/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Previous studies suggested miR-21 regulated epithelial-mesenchymal transition (EMT) and fibrosis in organs. The aim of this study was to explore the role and mechanism of miR-21 in EMT process of CD(Crohn's disease)-associated intestinal fibrosis. METHODS Tissue biopsies from fibrotic and nonfibrotic intestine of CD patients, and non-CD patients were obtained; chronic intestinal fibrosis model established by TNBS was treated with antagonist of miR-21; human intestinal epithelial cell, NCM460, were transfected with miR-21 mimics or inhibitor. The expressions of PTEN and mTOR, EMT-related markers and severity of colitis and fibrosis were examined. RESULTS Compared to the controls, miR-21 was significantly upregulated in the intestinal tissues from CD patients with fibro stenosis, followed by decreased PTEN expression, increased EMT markers, and mTOR expression, and imbalanced ratio of MMP9(matrix metalloproteinase 9)/TIMP1(tissue inhibitor of metalloproteinase 1). MiR-21 downregulated the expression of PTEN and upregulated mTOR signal in NCM460 cell. Also, knocking miR-21 down reduced EMT in vitro. Inhibiting miR-21 with antagonists reversed TNBS-induced intestinal fibrosis in vivo, through suppressing EMT and balancing MMPs/TIMPs. CONCLUSION We identified the involvement of miR-21 in EMT during intestinal fibrosis via targeting PTEN and mTOR, and miR-21 inhibition relieved intestinal fibrosis by regulating extracellular matrix (ECM) remodeling . Our results indicated miR-21 as a potential new target for the treatment of fibrosis complication in CD.
Collapse
Affiliation(s)
- Zhizhi Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University and China Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi 330006, China
| | - Huihui Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University and China Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi 330006, China
| | - Fei Cheng
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University and China Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi 330006, China
| | - Zhendong Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University and China Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi 330006, China
| | - Shunhua Long
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University and China Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
32
|
Naghdalipour M, Moradi N, Fadaei R, Rezghi Barez S, Sayyahfar S, Mokhtare M, Fard TK, Fallah S, Esteghamati A. Alteration of miR-21, miR-433 and miR-590 tissue expression related to the TGF-β signaling pathway in ulcerative colitis patients. Arch Physiol Biochem 2022; 128:1170-1174. [PMID: 32412349 DOI: 10.1080/13813455.2020.1762656] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Ulcerative colitis (UC) is an inflammatory disease, and studies have suggested a role for TGF-β signalling pathway in the pathogenesis of UC. In the present study, we evaluated expression of TGF-β signalling genes and their regulatory microRNAs in patients with UC and control subjects. The expression of TGF-β1, SMAD2, SMAD3, miR-21, miR-101, miR-433, and miR-590 were evaluated using real-time PCR in biopsy samples of the patients and controls. Results showed increased expression of TGF-β1 and SMAD3 in the patients compared to controls. In addition, miR-21 and miR-433 were found to be higher in the patients compared to controls; however, miR-590 was found to be lower. Moreover, miR-433 was demonstrated to have positive correlation with SMAD3 and TGF-β while miR-21 was positively correlated with TGF-β1. MiR-590 was negatively correlated with SMAD2 and SMAD3. Results of the present study suggested a role for TGF-β signalling pathway related microRNAs in pathogenesis of UC.
Collapse
Affiliation(s)
- Mehri Naghdalipour
- Research Center of Pediatric Infectious Diseases, Institute of Immunology and Infectious Diseases, Iran University of medical sciences, Tehran, Iran
| | - Nariman Moradi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Reza Fadaei
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shekufe Rezghi Barez
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shirin Sayyahfar
- Research Center of Pediatric Infectious Diseases, Institute of Immunology and Infectious Diseases, Iran University of medical sciences, Tehran, Iran
| | - Marjan Mokhtare
- Colorectal Research Center, Iran university of medical sciences, Tehran, Iran
| | - Toktam Kazemi Fard
- Department of Clinical Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soudabeh Fallah
- Department of Clinical Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Aboulreza Esteghamati
- Research Center of Pediatric Infectious Diseases, Institute of Immunology and Infectious Diseases, Iran University of medical sciences, Tehran, Iran
| |
Collapse
|
33
|
Lovinfosse P, Hustinx R. The role of PET imaging in inflammatory bowel diseases: state-of-the-art review. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2022; 66:206-217. [PMID: 35708600 DOI: 10.23736/s1824-4785.22.03467-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Inflammatory bowel diseases (IBD), i.e. Crohn disease and ulcerative colitis, are autoimmune processes of undetermined origin characterized by the chronic inflammation of the digestive tract. There is no single gold-standard to diagnose IBD which is therefore carried out through the combination of endoscopy, biopsy, radiological and biological investigations; and the development of non-invasive technique allowing the assessment and monitoring of these diseases is necessary. In this state-of-the-art review of the literature, we present the results of PET imaging studies for the diagnosis and staging of IBD (suspected or known), response evaluation to treatment and evaluation of one the main complication, i.e. strictures; explain the reasons why this examination is currently not considered in the IBD guidelines, e.g. radiation exposure, lack of standardization and not validated performances; and finally discuss the perspectives that could possibly allow it to find a place in the future, e.g. digital PET-CT, dynamic PET images acquisition, new radiopharmaceuticals, use of radiomics and use of artificial intelligence for automatically characterize and quantify digestive [18F]FDG uptake.
Collapse
Affiliation(s)
- Pierre Lovinfosse
- Division of Nuclear Medicine and Oncological Imaging, University Hospital CHU of Liège, Liège, Belgium -
- GIGA-CRC in vivo Imaging, University of Liège, Liège, Belgium -
| | - Roland Hustinx
- Division of Nuclear Medicine and Oncological Imaging, University Hospital CHU of Liège, Liège, Belgium
- GIGA-CRC in vivo Imaging, University of Liège, Liège, Belgium
| |
Collapse
|
34
|
Vieujean S, Caron B, Haghnejad V, Jouzeau JY, Netter P, Heba AC, Ndiaye NC, Moulin D, Barreto G, Danese S, Peyrin-Biroulet L. Impact of the Exposome on the Epigenome in Inflammatory Bowel Disease Patients and Animal Models. Int J Mol Sci 2022; 23:7611. [PMID: 35886959 PMCID: PMC9321337 DOI: 10.3390/ijms23147611] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel diseases (IBD) are chronic inflammatory disorders of the gastrointestinal tract that encompass two main phenotypes, namely Crohn's disease and ulcerative colitis. These conditions occur in genetically predisposed individuals in response to environmental factors. Epigenetics, acting by DNA methylation, post-translational histones modifications or by non-coding RNAs, could explain how the exposome (or all environmental influences over the life course, from conception to death) could influence the gene expression to contribute to intestinal inflammation. We performed a scoping search using Medline to identify all the elements of the exposome that may play a role in intestinal inflammation through epigenetic modifications, as well as the underlying mechanisms. The environmental factors epigenetically influencing the occurrence of intestinal inflammation are the maternal lifestyle (mainly diet, the occurrence of infection during pregnancy and smoking); breastfeeding; microbiota; diet (including a low-fiber diet, high-fat diet and deficiency in micronutrients); smoking habits, vitamin D and drugs (e.g., IBD treatments, antibiotics and probiotics). Influenced by both microbiota and diet, short-chain fatty acids are gut microbiota-derived metabolites resulting from the anaerobic fermentation of non-digestible dietary fibers, playing an epigenetically mediated role in the integrity of the epithelial barrier and in the defense against invading microorganisms. Although the impact of some environmental factors has been identified, the exposome-induced epimutations in IBD remain a largely underexplored field. How these environmental exposures induce epigenetic modifications (in terms of duration, frequency and the timing at which they occur) and how other environmental factors associated with IBD modulate epigenetics deserve to be further investigated.
Collapse
Affiliation(s)
- Sophie Vieujean
- Hepato-Gastroenterology and Digestive Oncology, University Hospital CHU of Liège, 4000 Liege, Belgium;
| | - Bénédicte Caron
- Department of Gastroenterology NGERE (INSERM U1256), Nancy University Hospital, University of Lorraine, Vandœuvre-lès-Nancy, F-54052 Nancy, France; (B.C.); (V.H.)
| | - Vincent Haghnejad
- Department of Gastroenterology NGERE (INSERM U1256), Nancy University Hospital, University of Lorraine, Vandœuvre-lès-Nancy, F-54052 Nancy, France; (B.C.); (V.H.)
| | - Jean-Yves Jouzeau
- CNRS (French National Centre for Scientific Research), Laboratoire IMoPA, Université de Lorraine, UMR 7365, F-54000 Nancy, France; (J.-Y.J.); (P.N.); (D.M.); (G.B.)
| | - Patrick Netter
- CNRS (French National Centre for Scientific Research), Laboratoire IMoPA, Université de Lorraine, UMR 7365, F-54000 Nancy, France; (J.-Y.J.); (P.N.); (D.M.); (G.B.)
| | - Anne-Charlotte Heba
- NGERE (Nutrition-Genetics and Exposure to Environmental Risks), National Institute of Health and Medical Research, University of Lorraine, F-54000 Nancy, France; (A.-C.H.); (N.C.N.)
| | - Ndeye Coumba Ndiaye
- NGERE (Nutrition-Genetics and Exposure to Environmental Risks), National Institute of Health and Medical Research, University of Lorraine, F-54000 Nancy, France; (A.-C.H.); (N.C.N.)
| | - David Moulin
- CNRS (French National Centre for Scientific Research), Laboratoire IMoPA, Université de Lorraine, UMR 7365, F-54000 Nancy, France; (J.-Y.J.); (P.N.); (D.M.); (G.B.)
| | - Guillermo Barreto
- CNRS (French National Centre for Scientific Research), Laboratoire IMoPA, Université de Lorraine, UMR 7365, F-54000 Nancy, France; (J.-Y.J.); (P.N.); (D.M.); (G.B.)
- Lung Cancer Epigenetics, Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Universidad de la Salud del Estado de Puebla, Puebla 72000, Mexico
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele and University Vita-Salute San Raffaele, 20132 Milan, Italy;
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology NGERE (INSERM U1256), Nancy University Hospital, University of Lorraine, Vandœuvre-lès-Nancy, F-54052 Nancy, France; (B.C.); (V.H.)
| |
Collapse
|
35
|
Lu Y, Guan T, Xu S, Chen YE, Shen Q, Zhu S, Liu Y, Liang J, Hou S. Asperuloside inhibited epithelial-mesenchymal transition in colitis associated cancer via activation of vitamin D receptor. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154070. [PMID: 35523114 DOI: 10.1016/j.phymed.2022.154070] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/13/2022] [Accepted: 03/19/2022] [Indexed: 05/10/2023]
Abstract
BACKGROUND Asperuloside is a natural compound extracted from various herbs with several bioactivities. Its effects on anti-inflammation and anti-tumor indicated that asperuloside might prevent colorectal cancer developing from inflammatory bowel diseases (IBD). But there were few reports about the efficacy and mechanism of asperuloside on improving colorectal cancer. It has been reported that vitamin D receptor (VDR) could regulate the expression of SMAD3. In previous study, asperuloside could significantly improve the expression of VDR and reduced Smad3 mRNA in IEC-6 cell. PURPOSE The present study was aimed to investigate the potential mechanism of asperuloside on inhibiting epithelial-mesenchymal transition (EMT) in colitis associated cancer. STUDY DESIGN First, in LPS-injured IEC-6 cell, asperuloside inhibited phosphorylated p65 (p-p65) level, improved VDR expression and reduced Smad3 mRNA. Second, we wonder the relationship between VDR signaling and nucleus factor-kappaB (NF-κB) signaling during asperuloside on reducing Smad3 mRNA. And then, the effect of asperuloside on inhibiting EMT development through VDR/Smad3 was investigated. Finally, we testified the effect of asperuloside on protecting against colitis associated cancer (CAC) by inhibiting EMT development through VDR/Smad3. METHODS Pyrrolidinedithiocarbamate ammonium (PDTC) was used for established NF-κB-inhibited IEC-6 cell. This cell was applied for investigating the relationship between NF-κB and VDR of asperuloside on inhibiting Smad3. VDR-inhibited cell was established by small interfering RNA (siRNA) of VDR and was employed to investigate the role of VDR for asperuloside on decreasing Smad3. Transforming growth factor β1 (TGFβ1) was used for inducing EMT/fibrosis in IEC-6 cell. TGFβ1-stimulated cell was used for testifying the effect of asperuloside on inhibiting EMT development. AOM/DSS-induced CAC was established to investigate the effect of asperuloside on suppressing cancer development. RESULTS Asperuloside inhibited the level of p-p65 which was up-regulated by LPS. Asperuloside could up-regulate VDR signaling and reduce Smad3 mRNA in NF-κB-knockdown IEC-6 cells. Asperuloside failed to reduce Smad3 mRNA due to VDR knockdown, which implied that asperuloside might down-regulate Smad3 mRNA dependently on activation of VDR signaling and independently on inhibiting NF-κB signaling. Asperuloside exhibited significant prevention of EMT development in TGFβ1-induced IEC-6 cell (EMT cell) and mice CAC. Asperuloside reduced the transform of epithelial phenotype into motile mesenchymal phenotype in EMT cell along with decreasing levels of EMT markers by inhibiting Smad3 mRNA via activation of VDR. In mice with CAC, expression of VDR in colon was improved by asperuloside. Symptoms of colitis, tumor number and tumor size were significantly inhibited by asperuloside. Suppressed EMT development was determined by reduced α-SMA expression and decreased mRNAs of several EMT markers. CONCLUSION Asperuloside might prevent CAC through inhibiting EMT development via regulation of VDR/Smad3 pathway.
Collapse
Affiliation(s)
- Yingyu Lu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ting Guan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuoxi Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong-Er Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Shen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shumin Zhu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jian Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shaozhen Hou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
36
|
Estrada HQ, Patel S, Rabizadeh S, Casero D, Targan SR, Barrett RJ. Development of a Personalized Intestinal Fibrosis Model Using Human Intestinal Organoids Derived From Induced Pluripotent Stem Cells. Inflamm Bowel Dis 2022; 28:667-679. [PMID: 34918082 PMCID: PMC9074870 DOI: 10.1093/ibd/izab292] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Intestinal fibrosis is a serious complication of Crohn's disease. Numerous cell types including intestinal epithelial and mesenchymal cells are implicated in this process, yet studies are hampered by the lack of personalized in vitro models. Human intestinal organoids (HIOs) derived from induced pluripotent stem cells (iPSCs) contain these cell types, and our goal was to determine the feasibility of utilizing these to develop a personalized intestinal fibrosis model. METHODS iPSCs from 2 control individuals and 2 very early onset inflammatory bowel disease patients with stricturing complications were obtained and directed to form HIOs. Purified populations of epithelial and mesenchymal cells were derived from HIOs, and both types were treated with the profibrogenic cytokine transforming growth factor β (TGFβ). Quantitative polymerase chain reaction and RNA sequencing analysis were used to assay their responses. RESULTS In iPSC-derived mesenchymal cells, there was a significant increase in the expression of profibrotic genes (Col1a1, Col5a1, and TIMP1) in response to TGFβ. RNA sequencing analysis identified further profibrotic genes and demonstrated differential responses to this cytokine in each of the 4 lines. Increases in profibrotic gene expression (Col1a1, FN, TIMP1) along with genes associated with epithelial-mesenchymal transition (vimentin and N-cadherin) were observed in TGFβ -treated epithelial cells. CONCLUSIONS We demonstrate the feasibility of utilizing iPSC-HIO technology to model intestinal fibrotic responses in vitro. This now permits the generation of near unlimited quantities of patient-specific cells that could be used to reveal cell- and environmental-specific mechanisms underpinning intestinal fibrosis.
Collapse
Affiliation(s)
- Hannah Q Estrada
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shachi Patel
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shervin Rabizadeh
- Division of Pediatric Gastroenterology, Cedars-Sinai Medical Center, Los Angeles, CA, USAand
| | - David Casero
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephan R Targan
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Robert J Barrett
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
37
|
Inhibition of epithelial SHH signaling exerts a dual protective effect against inflammation and epithelial–mesenchymal transition in inflammatory bowel disease. Toxicol In Vitro 2022; 82:105382. [DOI: 10.1016/j.tiv.2022.105382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/29/2022] [Accepted: 05/09/2022] [Indexed: 11/19/2022]
|
38
|
Wang Y, Huang B, Jin T, Ocansey DKW, Jiang J, Mao F. Intestinal Fibrosis in Inflammatory Bowel Disease and the Prospects of Mesenchymal Stem Cell Therapy. Front Immunol 2022; 13:835005. [PMID: 35370998 PMCID: PMC8971815 DOI: 10.3389/fimmu.2022.835005] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
Intestinal fibrosis is an important complication of inflammatory bowel disease (IBD). In the course of the development of fibrosis, certain parts of the intestine become narrowed, significantly destroying the structure and function of the intestine and affecting the quality of life of patients. Chronic inflammation is an important initiating factor of fibrosis. Unfortunately, the existing anti-inflammatory drugs cannot effectively prevent and alleviate fibrosis, and there is no effective anti-fibrotic drug, which makes surgical treatment the mainstream treatment for intestinal fibrosis and stenosis. Mesenchymal stem cells (MSCs) are capable of tissue regeneration and repair through their self-differentiation, secretion of cytokines, and secretion of extracellular vesicles. MSCs have been shown to play an important therapeutic role in the fibrosis of many organs. However, the role of MSC in intestinal fibrosis largely remained unexplored. This review summarizes the mechanism of intestinal fibrosis, including the role of immune cells, TGF-β, and the gut microbiome and metabolites. Available treatment options for fibrosis, particularly, MSCs are also discussed.
Collapse
Affiliation(s)
- Yifei Wang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Bin Huang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- General Surgery Department, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
| | - Tao Jin
- Department of Gastrointestinal and Endoscopy, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
- Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
- *Correspondence: Jiajia Jiang, ; Fei Mao,
| | - Fei Mao
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
- *Correspondence: Jiajia Jiang, ; Fei Mao,
| |
Collapse
|
39
|
Suau R, Pardina E, Domènech E, Lorén V, Manyé J. The Complex Relationship Between Microbiota, Immune Response and Creeping Fat in Crohn's Disease. J Crohns Colitis 2022; 16:472-489. [PMID: 34528668 DOI: 10.1093/ecco-jcc/jjab159] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the last decade, there has been growing interest in the pathological involvement of hypertrophic mesenteric fat attached to the serosa of the inflamed intestinal segments involved in Crohn's disease [CD], known as creeping fat. In spite of its protective nature, creeping fat harbours an aberrant inflammatory activity which, in an already inflamed intestine, may explain why creeping fat is associated with a greater severity of CD. The transmural inflammation of CD facilitates the interaction of mesenteric fat with translocated intestinal microorganisms, contributing to activation of the immune response. This may be not the only way in which microorganisms alter the homeostasis of this fatty tissue: intestinal dysbiosis may also impair xenobiotic metabolism. All these CD-related alterations have a functional impact on nuclear receptors such as the farnesoid X receptor or the peroxisome proliferator-activated receptor γ, which are implicated in regulation of the immune response, adipogenesis and the maintenance of barrier function, as well as on creeping fat production of inflammatory-associated cells such as adipokines. The dysfunction of creeping fat worsens the inflammatory course of CD and may favour intestinal fibrosis and fistulizing complications. However, our current knowledge of the pathophysiology and pathogenic role of creeping fat is controversial and a better understanding might provide new therapeutic targets for CD. Here we aim to review and update the key cellular and molecular alterations involved in this inflammatory process that link the pathological components of CD with the development of creeping fat.
Collapse
Affiliation(s)
- Roger Suau
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| | - Eva Pardina
- Biochemistry and Molecular Biomedicine Department, University of Barcelona, Barcelona (Catalonia), Spain
| | - Eugeni Domènech
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain.,Gastroenterology Department, 'Germans Trias i Pujol' University Hospital, Badalona (Catalonia), Spain
| | - Violeta Lorén
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| | - Josep Manyé
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| |
Collapse
|
40
|
Caparrós E, Wiest R, Scharl M, Rogler G, Gutiérrez Casbas A, Yilmaz B, Wawrzyniak M, Francés R. Dysbiotic microbiota interactions in Crohn's disease. Gut Microbes 2021; 13:1949096. [PMID: 34313550 PMCID: PMC8320851 DOI: 10.1080/19490976.2021.1949096] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Crohn's disease (CD) is a major form of inflammatory bowel disease characterized by transmural inflammation along the alimentary tract. Changes in the microbial composition and reduction in species diversity are recognized as pivotal hallmarks in disease dynamics, challenging the gut barrier function and shaping a pathological immune response in genetically influenced subjects. The purpose of this review is to delve into the modification of the gut microbiota cluster network during CD progression and to discuss how this shift compromises the gut barrier integrity, granting the translocation of microbes and their products. We then complete the scope of the review by retracing gut microbiota dysbiosis interactions with the main pathophysiologic factors of CD, starting from the host's genetic background to the immune inflammatory and fibrotic processes, providing a standpoint on the lifestyle/exogenous factors and the potential benefits of targeting a specific gut microbiota.
Collapse
Affiliation(s)
- Esther Caparrós
- Dpto Medicina Clínica, Universidad Miguel Hernández, San Juan De Alicante, Spain,Iis Isabial, Hospital General Universitario De Alicante, Alicante, Spain
| | - Reiner Wiest
- Department for Biomedical Research, Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Ana Gutiérrez Casbas
- Iis Isabial, Hospital General Universitario De Alicante, Alicante, Spain,CIBERehd, Instituto De Salud Carlos III, Madrid, Spain
| | - Bahtiyar Yilmaz
- Department for Biomedical Research, Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Marcin Wawrzyniak
- Department of Gastroenterology and Hepatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Rubén Francés
- Dpto Medicina Clínica, Universidad Miguel Hernández, San Juan De Alicante, Spain,Iis Isabial, Hospital General Universitario De Alicante, Alicante, Spain,CIBERehd, Instituto De Salud Carlos III, Madrid, Spain,CONTACT Rubén Francés Hepatic and Intestinal Immunobiology Group. Departamento De Medicina Clínica, Universidad Miguel Hernández De Elche. Carretera Alicante-Valencia, Km 8,703550San Juan De Alicante
| |
Collapse
|
41
|
Schmoyer CJ, Saidman J, Bohl JL, Bierly CL, Kuemmerle JF, Bickston SJ. The Pathogenesis and Clinical Management of Stricturing Crohn Disease. Inflamm Bowel Dis 2021; 27:1839-1852. [PMID: 33693860 DOI: 10.1093/ibd/izab038] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Indexed: 02/07/2023]
Abstract
Stricturing of the gastrointestinal tract is a common complication in Crohn disease and is a significant cause of morbidity and mortality among this population. The inflammatory process initiates fibrosis, leading to aberrant wound healing and excess deposition of extracellular matrix proteins. Our understanding of this process has grown and encompasses cellular mechanisms, epigenetic modifications, and inherent genetic predisposition toward fibrosis. Although medications can improve inflammation, there is still no drug to attenuate scar formation. As such, management of stricturing disease requires a multidisciplinary and individualized approach including medical management, therapeutic endoscopy, and surgery. This review details the current understanding regarding the pathogenesis, detection, and management of stricturing Crohn disease.
Collapse
Affiliation(s)
- Christopher J Schmoyer
- Virginia Commonwealth University, Division of Gastroenterology and Hepatology, Richmond, Virginia, USA
| | - Jakob Saidman
- Virginia Commonwealth University, Division of Gastroenterology and Hepatology, Richmond, Virginia, USA
| | - Jaime L Bohl
- Virginia Commonwealth University, Division of Colorectal Surgery, Richmond, Virginia, USA
| | - Claire L Bierly
- Virginia Commonwealth University, Division of Gastroenterology and Hepatology, Richmond, Virginia, USA
| | - John F Kuemmerle
- Virginia Commonwealth University, Division of Gastroenterology and Hepatology, Richmond, Virginia, USA.,Virginia Commonwealth University, Department of Physiology and Biophysics, Richmond, Virginia, USA
| | - Stephen J Bickston
- Virginia Commonwealth University, Division of Gastroenterology and Hepatology, Richmond, Virginia, USA
| |
Collapse
|
42
|
Corsi F, Sorrentino L, Albasini S, Colombo F, Cigognini M, Massari A, Morasso C, Mazzucchelli S, Piccotti F, Ardizzone S, Sampietro GM, Truffi M. Circulating Fibroblast Activation Protein as Potential Biomarker in Patients With Inflammatory Bowel Disease. Front Med (Lausanne) 2021; 8:725726. [PMID: 34621763 PMCID: PMC8490650 DOI: 10.3389/fmed.2021.725726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
A major concern in the management of Inflammatory Bowel Disease (IBD) is the absence of accurate and specific biomarkers to drive diagnosis and monitor disease status timely and non-invasively. Fibroblast activation protein (FAP) represents a hallmark of IBD bowel strictures, being overexpressed in stenotic intestinal myofibroblasts. The present study aimed at evaluating the potential of circulating FAP (cFAP) as an accessible blood biomarker of IBD. Quantitative determination of cFAP was performed by enzyme-linked immunosorbent assay on plasma samples prospectively collected from patients with IBD and control subjects. A discrimination model was established on a training set of 50% patients and validated on independent samples. Results showed that cFAP concentration was reduced in patients with IBD when compared to controls (p < 0.0001). Age, sex, smoking, disease location and behavior, disease duration and therapy were not associated with cFAP. The sensitivity and specificity of cFAP in discriminating IBD from controls were 70 and 84%, respectively, based on the optimal cutoff (57.6 ng mL−1, AUC = 0.78). Predictions on the test set had 57% sensitivity, 65% specificity, and 61% accuracy. There was no strong correlation between cFAP and routine inflammatory markers in the patients' population. A subgroup analysis was performed on patients with Crohn's disease undergoing surgery and revealed that cFAP correlates with endoscopic mucosal healing. In conclusion, cFAP deserves attention as a promising blood biomarker to triage patients with suspected IBD. Moreover, it might function as a biomarker of post-operative remission in patients with Crohn's disease.
Collapse
Affiliation(s)
- Fabio Corsi
- Breast Unit, Surgery Department, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy.,Department of Biomedical and Clinical Sciences "L. Sacco", Universitá di Milano, Milan, Italy
| | - Luca Sorrentino
- Department of Biomedical and Clinical Sciences "L. Sacco", Universitá di Milano, Milan, Italy
| | - Sara Albasini
- Breast Unit, Surgery Department, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Francesco Colombo
- Division of General Surgery, ASST Fatebenefratelli Sacco, Luigi Sacco University Hospital, Milan, Italy
| | - Maria Cigognini
- Division of General Surgery, ASST Rhodense, Rho Memorial Hospital, Milan, Italy
| | - Alessandro Massari
- Division of Gastroenterology, ASST Fatebenefratelli Sacco, Luigi Sacco University Hospital, Milan, Italy
| | - Carlo Morasso
- Nanomedicine and Molecular Imaging Lab, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Serena Mazzucchelli
- Department of Biomedical and Clinical Sciences "L. Sacco", Universitá di Milano, Milan, Italy
| | - Francesca Piccotti
- Nanomedicine and Molecular Imaging Lab, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Sandro Ardizzone
- Department of Biomedical and Clinical Sciences "L. Sacco", Universitá di Milano, Milan, Italy.,Division of Gastroenterology, ASST Fatebenefratelli Sacco, Luigi Sacco University Hospital, Milan, Italy
| | | | - Marta Truffi
- Nanomedicine and Molecular Imaging Lab, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| |
Collapse
|
43
|
Kessler L, Ferdinandus J, Hirmas N, Zarrad F, Nader M, Kersting D, Weber M, Kazek S, Sraieb M, Hamacher R, Lueckerath K, Umutlu L, Fendler WP, Rischpler C. Pitfalls and common findings in 68Ga-FAPI-PET - A pictorial analysis. J Nucl Med 2021; 63:890-896. [PMID: 34620730 DOI: 10.2967/jnumed.121.262808] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/20/2021] [Indexed: 11/16/2022] Open
Abstract
Introduction: Fibroblast activation protein inhibitor positron emission tomography (FAPI-PET) is a new tool in the diagnostic workup of cancer. With growing volume of applications pitfalls and common findings need to be considered for Ga-68-FAPI-PET image interpretation. The aim of this study was to summarize common findings and report pitfalls in Ga-68-FAPI-PET. Methods and materials: 91 patients underwent whole-body PET/CT with either FAPI-04 (N = 25) or FAPI-46 (N = 66). Findings were rated in a consensus session of two experienced readers. Pitfalls and common findings were defined as focal or localized uptake above background and categorized as unspecific / non-malignant and grouped into degenerative, muscular, scarring / wound-healing, uterine, mammary glands and head-and-neck findings. Frequency of findings was reported on a per-patient and per-group basis and SUVmax, SUVmean and SUVpeak was measured. Results: Non-tumor specific tracer uptake was found in 81.3 % of patients. The most frequent finding was tracer uptake in degenerative lesions (51.6%) with mean SUVmax 7.7 ± 2.9 and head-and-neck (45.1%) findings. Except for salivary glands, the uptake values did not differ between 10 and 60 min p.i. in most findings. Uterine uptake was found in most women (66.7%) with mean SUVmax 12.2 ± 7.3 and uptake correlated negatively with age (SUVmax r = -0.6, p<0.01; SUVpeak r = -0.57, p<0.01; SUVmean r = -0.58, p<0.01). Conclusion: Pitfalls include non-tumor specific Ga-68-FAPI uptake in degenerative lesions, muscle, head-and-neck, scarring, mammary glands or uterus. Here we summarize findings to inform readers at centers introducing Ga-68-FAPI-PET to avoid common mistakes.
Collapse
Affiliation(s)
- Lukas Kessler
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Justin Ferdinandus
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Nader Hirmas
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Fadi Zarrad
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Michael Nader
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Germany
| | - David Kersting
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Manuel Weber
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Sandra Kazek
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Miriam Sraieb
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Rainer Hamacher
- Department of Medical Oncology, University Hospital Essen, University of Duisburg-Essen
| | - Katharina Lueckerath
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Lale Umutlu
- Institute of Diagnostic and Interventional Radiology, University of Duisburg-Essen
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Christoph Rischpler
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Germany
| |
Collapse
|
44
|
Pulakazhi Venu VK, Alston L, Iftinca M, Tsai YC, Stephens M, Warriyar K V V, Rehal S, Hudson G, Szczepanski H, von der Weid PY, Altier C, Hirota SA. Nr4A1 modulates inflammation-associated intestinal fibrosis and dampens fibrogenic signaling in myofibroblasts. Am J Physiol Gastrointest Liver Physiol 2021; 321:G280-G297. [PMID: 34288735 DOI: 10.1152/ajpgi.00338.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal fibrosis is a common complication of the inflammatory bowel diseases (IBDs), contributing to tissue stiffening and luminal narrowing. Human nuclear receptor 4A 1 (NR4A1) was previously reported to regulate mesenchymal cell function and dampen fibrogenic signaling. NR4A1 gene variants are associated with IBD risk, and it has been shown to regulate intestinal inflammation. Here, we tested the hypothesis that NR4A1 acts as a negative regulator of intestinal fibrosis through regulating myofibroblast function. Using the SAMP1/YitFc mouse, we tested whether two pharmacological agents known to enhance NR4A1 signaling, cytosporone B (Csn-B) or 6-mercaptopurine (6-MP), could reduce fibrosis. We also used the dextran sulfate sodium (DSS) model of colitis and assessed the magnitude of colonic fibrosis in mouse nuclear receptor 4A 1 (Nr4a1-/-) and their wild-type littermates (Nr4a1+/+). Lastly, intestinal myofibroblasts isolated from Nr4a1-/- and Nr4a1+/+ mice or primary human intestinal myofibroblasts were stimulated with transforming growth factor-β1 (TGF-β1), in the presence or absence of Csn-B or 6-MP, and proliferation and ECM gene expression assessed. Csn-B or 6-MP treatment significantly reduced ileal thickness, collagen, and overall ECM content in SAMP1/YitFc mice. This was associated with a reduction in proliferative markers within the mesenchymal compartment. Nr4a1-/- mice exposed to DSS exhibited increased colonic thickening and ECM content. Nr4a1-/- myofibroblasts displayed enhanced TGF-β1-induced proliferation. Furthermore, Csn-B or 6-MP treatment was antiproliferative in Nr4a1+/+ but not Nr4a1-/- cells. Lastly, activating NR4A1 in human myofibroblasts reduced TGF-β1-induced collagen deposition and fibrosis-related gene expression. Our data suggest that NR4A1 can attenuate fibrotic processes in intestinal myofibroblasts and could provide a valuable clinical target to treat inflammation-associated intestinal fibrosis.NEW & NOTEWORTHY Fibrosis and increased muscle thickening contribute to stricture formation and intestinal obstruction, a complication that occurs in 30%-50% of patients with CD within 10 yr of disease onset. More than 50% of those who undergo surgery to remove the obstructed bowel will experience stricture recurrence. To date, there are no drug-based approaches approved to treat intestinal strictures. In the current submission, we identify NR4A1 as a novel target to treat inflammation-associated intestinal fibrosis.
Collapse
Affiliation(s)
- Vivek Krishna Pulakazhi Venu
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Disease, University of Calgary, Calgary, Alberta, Canada
| | - Laurie Alston
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Disease, University of Calgary, Calgary, Alberta, Canada
| | - Mircea Iftinca
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Disease, University of Calgary, Calgary, Alberta, Canada
| | - Yi-Cheng Tsai
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Disease, University of Calgary, Calgary, Alberta, Canada
| | - Matthew Stephens
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Disease, University of Calgary, Calgary, Alberta, Canada
| | - Vineetha Warriyar K V
- Faculty of Kinesiology, Sport Injury Prevention Research Centre, University of Calgary, Calgary, Alberta, Canada
| | - Sonia Rehal
- Department of Advanced Diagnostics, University Health Network, Toronto, Ontario, Canada
| | - Grace Hudson
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Disease, University of Calgary, Calgary, Alberta, Canada
| | - Holly Szczepanski
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Disease, University of Calgary, Calgary, Alberta, Canada
| | - Pierre-Yves von der Weid
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Disease, University of Calgary, Calgary, Alberta, Canada
| | - Christophe Altier
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Disease, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Simon A Hirota
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Department of Immunology, Microbiology & Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Disease, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
45
|
Lu S, Zhu K, Guo Y, Wang E, Huang J. Evaluation of animal models of Crohn's disease with anal fistula (Review). Exp Ther Med 2021; 22:974. [PMID: 34335916 PMCID: PMC8290422 DOI: 10.3892/etm.2021.10406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/09/2021] [Indexed: 12/28/2022] Open
Abstract
Anal fistula is a common and serious complication of Crohn's disease (CD). A sufficiently suitable animal model that may be used to simulate this disease is yet to be established. The aim of the present review was to summarize the different characteristics and experimental methods of commonly used animal models of CD with anal fistula. Electronic databases were searched for studies reporting on the use of this type of animal model. A total of 234 related articles were retrieved, of which six articles met the inclusion criteria; these were used as references for the present review article. The characteristics of the animal models, the advantages and disadvantages of the modeling methods and the similarities with patients with CD and anal fistula were summarized and analyzed. The evidence suggests that a sufficiently suitable animal preclinical model requires to be established.
Collapse
Affiliation(s)
- Shuangshuang Lu
- Department of Internal Medicine, School of Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Gastrointestinal Center, Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Keyuan Zhu
- Department of Internal Medicine, School of Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Gastrointestinal Center, Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Yongxin Guo
- Department of Internal Medicine, School of Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Gastrointestinal Center, Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Enjing Wang
- Gastrointestinal Center, Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China.,Department of Internal Medicine, School of Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Jin Huang
- Department of Internal Medicine, School of Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Gastrointestinal Center, Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
46
|
Wenxiu J, Mingyue Y, Fei H, Yuxin L, Mengyao W, Chenyang L, Jia S, Hong Z, Shih DQ, Targan SR, Xiaolan Z. Effect and Mechanism of TL1A Expression on Epithelial-Mesenchymal Transition during Chronic Colitis-Related Intestinal Fibrosis. Mediators Inflamm 2021; 2021:5927064. [PMID: 34257516 PMCID: PMC8253633 DOI: 10.1155/2021/5927064] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 03/30/2021] [Accepted: 05/28/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND AIMS Recent evidences reveal that epithelial to mesenchymal transition (EMT) exacerbates the process of intestinal fibrosis. Tumor necrosis factor-like ligand 1A (TL1A) is a member of the tumor necrosis family (TNF), which can take part in the development of colonic inflammation and fibrosis by regulating immune response or inflammatory factors. The purpose of this study was to elucidate the possible contribution of TL1A in onset and progression of intestinal inflammation and fibrosis through EMT. METHODS Colonic specimens were obtained from patients with inflammatory bowel disease (IBD) and control individuals. The expression levels of TL1A and EMT-related markers in intestinal tissues were evaluated. Furthermore, the human colorectal adenocarcinoma cell line, HT-29, was stimulated with TL1A, anti-TL1A antibody, or BMP-7 to assess EMT process. In addition, transgenic mice expressing high levels of TL1A in lymphoid cells were used to further investigate the mechanism of TL1A in intestinal fibrosis. RESULTS High levels of TL1A expression were detected in the intestinal specimens of patients with ulcerative colitis and Crohn's disease and were negatively associated with the expression of an epithelial marker (E-cadherin), while it was positively associated with the expression of interstitial markers (FSP1 and α-SMA). Transgenic mice with high expression of TL1A were more sensitive to dextran sodium sulfate and exhibited severe intestinal inflammation and fibrosis. Additionally, the TGF-β1/Smad3 pathway may be involved in TL1A-induced EMT, and the expression of IL-13 and EMT-related transcriptional molecules (e.g., ZEB1 and Snail1) was increased in the intestinal specimens of the transgenic mice. Furthermore, TL1A-induced EMT can be influenced by anti-TL1A antibody or BMP-7 in vitro. CONCLUSIONS TL1A participates in the formation and process of EMT in intestinal fibrosis. This new knowledge enables us to better understand the pathogenesis of intestinal fibrosis and identify new therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Jia Wenxiu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, No. 80 Huanghe Road, Yuhua District, Shijiazhuang, Hebei, China
| | - Yang Mingyue
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, No. 80 Huanghe Road, Yuhua District, Shijiazhuang, Hebei, China
| | - Han Fei
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, No. 80 Huanghe Road, Yuhua District, Shijiazhuang, Hebei, China
| | - Luo Yuxin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, No. 80 Huanghe Road, Yuhua District, Shijiazhuang, Hebei, China
| | - Wu Mengyao
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, No. 80 Huanghe Road, Yuhua District, Shijiazhuang, Hebei, China
| | - Li Chenyang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, No. 80 Huanghe Road, Yuhua District, Shijiazhuang, Hebei, China
| | - Song Jia
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, No. 80 Huanghe Road, Yuhua District, Shijiazhuang, Hebei, China
| | - Zhang Hong
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, No. 80 Huanghe Road, Yuhua District, Shijiazhuang, Hebei, China
| | - David Q. Shih
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephan R. Targan
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Zhang Xiaolan
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, No. 80 Huanghe Road, Yuhua District, Shijiazhuang, Hebei, China
| |
Collapse
|
47
|
Lopes TCM, Almeida GG, Souza IA, Borges DC, de Lima WG, Prazeres PHDM, Birbrair A, Arantes RME, Mosser DM, Goncalves R. High-Density-Immune-Complex Regulatory Macrophages Promote Recovery of Experimental Colitis in Mice. Inflammation 2021; 44:1069-1082. [PMID: 33394188 DOI: 10.1007/s10753-020-01403-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/29/2020] [Accepted: 12/13/2020] [Indexed: 12/12/2022]
Abstract
Macrophages not only play a fundamental role in the pathogenesis of inflammatory bowel disease (IBD), but they also play a major role in preserving intestinal homeostasis. In this work, we evaluated the role of macrophages in IBD and investigated whether the functional reprogramming of macrophages to a very specific phenotype could decrease disease pathogenesis. Thus, macrophages were stimulated in the presence of high-density immune complexes which strongly upregulate their production of IL-10 and downregulate pro-inflammatory cytokines. The transfer of these high-density-immune-complex regulatory macrophages into mice with colitis was examined as a potential therapy proposal to control the disease. Animals subjected to colitis induction received these high-density-immune-complex regulatory macrophages, and then the Disease Activity Index (DAI), and macroscopic and microscopic lesions were measured. The treated group showed a dramatic improvement in all parameters analyzed, with no difference with the control group. The colon was macroscopically normal in appearance and size, and microscopically colon architecture was preserved. The immunofluorescence migration assay showed that these cells migrated to the inflamed intestine, being able to locally produce the cytokine IL-10, which could explain the dramatic improvement in the clinical and pathological condition of the animals. Thus, our results demonstrate that the polarization of macrophages to a high IL-10 producer profile after stimulation with high-density immune complexes was decisive in controlling experimental colitis, and that macrophages are a potential therapeutic target to be explored in the control of colitis.
Collapse
Affiliation(s)
- Tamara Cristina Moreira Lopes
- Departamento de Patologia Geral-Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | | | - Izabela Aparecida Souza
- Departamento de Patologia Geral-Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Diego Costa Borges
- Departamento de Bioquímica e Imunologia-Instituto de Ciências Biológicas, UFMG, Belo Horizonte, MG, Brazil
| | - Wanderson Geraldo de Lima
- Departamento de Ciências Biológicas-Instituto de Ciências Biológicas e Exatas, Universidade Federal de Ouro Preto (UFOP), Ouro Preto, MG, Brazil
| | - Pedro Henrique Dias Moura Prazeres
- Departamento de Patologia Geral-Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Alexander Birbrair
- Departamento de Patologia Geral-Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Rosa Maria Esteves Arantes
- Departamento de Patologia Geral-Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - David M Mosser
- Laboratory of Macrophage and Host Defense - Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Ricardo Goncalves
- Departamento de Patologia Geral-Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| |
Collapse
|
48
|
Talamona F, Truffi M, Caldarone AA, Ricciardi A, Corsi F, Pellegrini G, Morasso C, Taglietti A. Stable and scalable SERS tags conjugated with neutravidin for the detection of fibroblast activation protein (FAP) in primary fibroblasts. NANOTECHNOLOGY 2021; 32:295703. [PMID: 33831854 DOI: 10.1088/1361-6528/abf5fd] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/08/2021] [Indexed: 06/12/2023]
Abstract
SERS tags are a class of nanoparticles with great potential in advanced imaging experiments. The preparation of SERS tags however is complex, as they suffer from the high variability of the SERS signals observed even at the slightest sign of aggregation. Here, we developed a method for the preparation of SERS tags based on the use of gold nanostars conjugated with neutravidin. The SERS tags here obtained are extremely stable in all biological buffers commonly employed and can be prepared at a relatively large scale in very mild conditions. The obtained SERS tags have been used to monitor the expression of fibroblast activation protein alpha (FAP) on the membrane of primary fibroblasts obtained from patients affected by Crohn's disease. The SERS tags allowed the unambiguous identification of FAP on the surface of cells thus suggesting the feasibility of semi-quantitative analysis of the target protein. Moreover, the use of the neutravidin-biotin system allows to apply the SERS tags for any other marker detection, for example, different cancer cell types, simply by changing the biotinylated antibody chosen in the analysis.
Collapse
Affiliation(s)
- Federica Talamona
- Department of Chemistry, University of Pavia, Viale Taramelli 12, I-27100 Pavia, Italy
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, I-27100 Pavia, Italy
| | - Marta Truffi
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, I-27100 Pavia, Italy
| | | | | | - Fabio Corsi
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, I-27100 Pavia, Italy
- Department of Biomedical and Clinical Sciences 'Luigi Sacco', Università degli Studi di Milano, Milano, Italy
| | - Giovanni Pellegrini
- Department of Physics, University of Pavia, Via Bassi 6, I-27100 Pavia, Italy
| | - Carlo Morasso
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, I-27100 Pavia, Italy
| | - Angelo Taglietti
- Department of Chemistry, University of Pavia, Viale Taramelli 12, I-27100 Pavia, Italy
| |
Collapse
|
49
|
Vieujean S, Hu S, Bequet E, Salee C, Massot C, Bletard N, Pierre N, Quesada Calvo F, Baiwir D, Mazzucchelli G, De Pauw E, Coimbra Marques C, Delvenne P, Rieder F, Louis E, Meuwis MA. Potential Role of Epithelial Endoplasmic Reticulum Stress and Anterior Gradient Protein 2 Homologue in Crohn's Disease Fibrosis. J Crohns Colitis 2021; 15:1737-1750. [PMID: 33822017 PMCID: PMC8861373 DOI: 10.1093/ecco-jcc/jjab061] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Intestinal fibrosis is a common complication of Crohn's disease [CD]. It is characterised by an accumulation of fibroblasts differentiating into myofibroblasts secreting excessive extracellular matrix. The potential role of the intestinal epithelium in this fibrotic process remains poorly defined. METHODS We performed a pilot proteomic study comparing the proteome of surface epithelium, isolated by laser-capture microdissection, in normal and fibrotic zones of resected ileal CD strictures [13 zones collected in five patients]. Proteins of interests were validated by immunohistochemistry [IHC] in ileal and colonic samples of stricturing CD [n = 44], pure inflammatory CD [n = 29], and control [n = 40] subjects. The pro-fibrotic role of one selected epithelial protein was investigated through in-vitro experiments using HT-29 epithelial cells and a CCD-18Co fibroblast to myofibroblast differentiation model. RESULTS Proteomic study revealed an endoplasmic reticulum [ER] stress proteins increase in the epithelium of CD ileal fibrotic strictures, including anterior gradient protein 2 homologue [AGR2] and binding-immunoglobulin protein [BiP]. This was confirmed by IHC. In HT-29 cells, tunicamycin-induced ER stress triggered AGR2 intracellular expression and its secretion. Supernatant of these HT-29 cells, pre-conditioned by tunicamycin, led to a myofibroblastic differentiation when applied on CCD-18Co fibroblasts. By using recombinant protein and blocking agent for AGR2, we demonstrated that the secretion of this protein by epithelial cells can play a role in the myofibroblastic differentiation. CONCLUSIONS The development of CD fibrotic strictures could involve epithelial ER stress and particularly the secretion of AGR2.
Collapse
Affiliation(s)
- Sophie Vieujean
- Corresponding author: Sophie Vieujean, MD, Laboratory of Translational Gastroenterology, University of Liège, GIGA-Research, +2, B34, Avenue de l’hôpital 1, 4000 Liège, Belgium. Tel.: +32-4-3667256; fax: +32-4-3667889; mail:
| | | | - Emeline Bequet
- Laboratory of Translational Gastroenterology, University of Liège, Liège, Belgium,Division of Hepato-Gastroenterology, Department of Paediatrics, University Hospital of Liège, Liège, Belgium
| | - Catherine Salee
- Laboratory of Translational Gastroenterology, University of Liège, Liège, Belgium
| | - Charlotte Massot
- Laboratory of Translational Gastroenterology, University of Liège, Liège, Belgium
| | - Noëlla Bletard
- Pathological Anatomy and Cytology, University Hospital CHU of Liège, Liège, Belgium
| | - Nicolas Pierre
- Laboratory of Translational Gastroenterology, University of Liège, Liège, Belgium
| | | | | | - Gabriel Mazzucchelli
- MolSys Research Unit, Laboratory of Mass Spectrometry, University of Liège, Liège, Belgium
| | - Edwin De Pauw
- MolSys Research Unit, Laboratory of Mass Spectrometry, University of Liège, Liège, Belgium
| | | | - Philippe Delvenne
- Pathological Anatomy and Cytology, University Hospital CHU of Liège, Liège, Belgium
| | - Florian Rieder
- Gastroenterology, Hepatology & Nutrition, Cleveland Clinic, Cleveland, OH, USA
| | | | | |
Collapse
|
50
|
Hwang JS, Ma DJ, Choi J, Shin YJ. COL8A2 Regulates the Fate of Corneal Endothelial Cells. Invest Ophthalmol Vis Sci 2021; 61:26. [PMID: 32931574 PMCID: PMC7500139 DOI: 10.1167/iovs.61.11.26] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose To investigate the effect of COL8A2 repression on corneal endothelial cells (CECs) in vitro and in vivo. Methods Cultured human CECs (hCECs) were transfected with COL8A2 siRNA (siCOL8A2), and the cell viability and proliferation rate were measured. The expression of cell proliferation–associated molecules was evaluated by Western blotting and real-time reverse transcription PCR. Cell shape, Wingless-INT (WNT) signaling, and mitochondrial oxidative stress were also measured. For in vivo experiments, siCOL8A2 was transfected into rat CECs (rCECs), and corneal opacity and corneal endothelium were evaluated. Results After transfection with siCOL8A2, COL8A2 expression was reduced (80%). Cell viability, cell proliferation rate, cyclin D1 expression, and the number of cells in the S-phase were reduced in siCOL8A2-treated cells. The cell attained a fibroblast-like shape, and SNAI1, pSMAD2, and β-catenin expression, along with mitochondrial mass and oxidative stress levels, were altered. Corneal opacity increased, and the CECs were changed in rats in the siCOL8A2 group. Conclusions COL8A2 is required to maintain normal wound healing and CEC function.
Collapse
Affiliation(s)
- Jin Sun Hwang
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Dae Joong Ma
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Jinju Choi
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Young Joo Shin
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|