1
|
Morgan M, Loel B, Lang R, Reilly C, Balouch F, Burgess C, Thapar N, Lewindon P. The use of oral antibiotic cocktail in medically refractory paediatric acute severe colitis: a case series. Eur J Gastroenterol Hepatol 2025; 37:594-599. [PMID: 39976007 DOI: 10.1097/meg.0000000000002950] [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] [Indexed: 02/21/2025]
Abstract
OBJECTIVE Paediatric acute severe colitis (ASC) is a life-threatening gastroenterological emergency and a predictor of poor long-term inflammatory bowel disease outcomes. We report our experience with oral antibiotic combination therapy as rescue therapy for children with ASC failing to respond to conventional medical therapy (CMT). METHODS We analysed data of children admitted with ASC between January 2020 and January 2023 who failed steroids and infliximab and received the oral antibiotic combination therapy (vancomycin, amoxicillin, metronidazole and doxycycline). Treatments and responses including Paediatric Ulcerative Colitis Activity Index (PUCAI), biochemical markers, intestinal ultrasound (IUS) and colectomy rates (acute and deferred) were collated. RESULTS Oral antibiotic combination therapy was prescribed in 12 episodes of ASC in 11 children following failure of CMT. Improvements were seen in PUCAI (mean difference = -27.86, 95% confidence interval = -43.43 to -12.28, P < 0.001), albumin (mean: 29.5-33.6) and CRP (mean: 12-4), as well as in IUS (bowel wall thickening, extent of involvement or vascularity in five of seven). Five of 11 children were colectomy free at a maximum follow-up of 24 months. Three children had acute colectomy during index admission and three underwent deferred colectomy at a mean of 4 months. CONCLUSION Oral antibiotic combination therapy shows promise in deferring and, in some cases, averting the need for acute colectomy in medically refractory ASC. This notable finding warrants confirmation in larger studies.
Collapse
Affiliation(s)
- Manal Morgan
- Department of Paediatric Gastroenterology, Hepatology and Liver Transplant
| | - Benjamin Loel
- General Paediatric Department, Queensland Children's Hospital, Brisbane, Queensland, Australia
| | - Ray Lang
- Department of Paediatric Gastroenterology, Hepatology and Liver Transplant
| | - Claire Reilly
- Department of Paediatric Gastroenterology, Hepatology and Liver Transplant
| | - Fariha Balouch
- Department of Paediatric Gastroenterology, Hepatology and Liver Transplant
| | - Chris Burgess
- Department of Paediatric Gastroenterology, Hepatology and Liver Transplant
| | - Nikhil Thapar
- Department of Paediatric Gastroenterology, Hepatology and Liver Transplant
| | - Peter Lewindon
- Department of Paediatric Gastroenterology, Hepatology and Liver Transplant
| |
Collapse
|
2
|
Petracco G, Faimann I, Reichmann F. Inflammatory bowel disease and neuropsychiatric disorders: Mechanisms and emerging therapeutics targeting the microbiota-gut-brain axis. Pharmacol Ther 2025; 269:108831. [PMID: 40023320 DOI: 10.1016/j.pharmthera.2025.108831] [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: 09/23/2024] [Revised: 02/03/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are the two major entities of inflammatory bowel disease (IBD). These disorders are known for their relapsing disease course and severe gastrointestinal symptoms including pain, diarrhoea and bloody stool. Accumulating evidence suggests that IBD is not only restricted to the gastrointestinal tract and that disease processes are able to reach distant organs including the brain. In fact, up to 35 % of IBD patients also suffer from neuropsychiatric disorders such as generalized anxiety disorder and major depressive disorder. Emerging research in this area indicates that in many cases these neuropsychiatric disorders are a secondary condition as a consequence of the disturbed communication between the gut and the brain via the microbiota-gut-brain axis. In this review, we summarise the current knowledge on IBD-associated neuropsychiatric disorders. We examine the role of different pathways of the microbiota-gut-brain axis in the development of CNS disorders highlighting altered neural, immunological, humoral and microbial communication. Finally, we discuss emerging therapies targeting the microbiota-gut-brain axis to alleviate IBD and neuropsychiatric symptoms including faecal microbiota transplantation, psychobiotics, microbial metabolites and vagus nerve stimulation.
Collapse
Affiliation(s)
- Giulia Petracco
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Isabella Faimann
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Florian Reichmann
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria; BiotechMed-Graz, Austria.
| |
Collapse
|
3
|
Kantham S, Yu H, Cantelli CR, Chen G, Ma C, Chan JJ, Yang H, Tsai K, Lassueur K, Vallance BA, Jacobson K, Young RN. Development of novel GI-centric prostaglandin E 2 receptor type 4 (EP4) agonist prodrugs as treatment for ulcerative colitis and other intestinal inflammatory diseases. Bioorg Med Chem Lett 2025; 119:130093. [PMID: 39793629 DOI: 10.1016/j.bmcl.2025.130093] [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: 05/11/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Prostaglandin E2 receptor type 4 (EP4) agonists have been shown to be effective in treating experimental ulcerative colitis (UC) in animals and in human clinical trials, but their development has been impeded by unacceptable systemic side effects. In this study, a series of methylene phosphate prodrugs of a highly potent and selective prostaglandin EP4 receptor agonist were designed to target and remain localized in the gastrointestinal (GI) tract after either oral or rectal instillation. The prodrugs were designed to be converted to liberate active EP4 agonist by intestinal alkaline phosphate (IAP), a ubiquitous enzyme found at the luminal of the intestinal wall thus exposing the colon epithelial barrier while reducing systemic exposure to the active agonist. The prodrugs were shown to hydrolyze in plasma and after contact with GI tissue slices from ileum and colon. When optimized prodrugs were dosed orally, systemic peak exposure to the active agonist was not reduced, presumably due to IAP activity in the duodenum and small intestine. However, when dosed rectally, the prodrugs gave much reduced levels of EP4 agonist in the blood. An optimized prodrug was shown to be retained in the colon, when compared with free agonist after rectal administration in healthy mice and to be efficacious in a model of UC (the DSS mouse model). Plasma exposure to the active agonist was also much reduced in the mouse model of UC after 4 days of rectal dosing but after 7 days, one DSS mouse showed elevated systemic levels of the free agonist in the blood. The concept of efficacy and intestinal retention of an EP4 agonist-methylene phosphate prodrug was proven for rectal instillation but in DSS treated mice, severe disease appears to compromise the epithelia barrier sufficiently to allow some absorption of the prodrug to occur. Thus, further optimization of these prodrugs is required before a candidate can be selected for development for treating severe ulcerative colitis.
Collapse
Affiliation(s)
- Srinivas Kantham
- Department of Chemistry, Simon Fraser University Burnaby British Columbia Canada
| | - Hongbing Yu
- Department of Pediatrics, University of British Columbia Vancouver British Columbia Canada
| | | | - Gang Chen
- Department of Chemistry, Simon Fraser University Burnaby British Columbia Canada
| | - Caixia Ma
- Department of Pediatrics, University of British Columbia Vancouver British Columbia Canada
| | - Jocelyn J Chan
- Department of Pediatrics, University of British Columbia Vancouver British Columbia Canada
| | - Hyungjun Yang
- Department of Pediatrics, University of British Columbia Vancouver British Columbia Canada
| | - Kevin Tsai
- Department of Pediatrics, University of British Columbia Vancouver British Columbia Canada
| | - Kristiana Lassueur
- Department of Chemistry, Simon Fraser University Burnaby British Columbia Canada
| | - Bruce A Vallance
- Department of Pediatrics, University of British Columbia Vancouver British Columbia Canada
| | - Kevan Jacobson
- Department of Pediatrics, University of British Columbia Vancouver British Columbia Canada.
| | - Robert N Young
- Department of Chemistry, Simon Fraser University Burnaby British Columbia Canada.
| |
Collapse
|
4
|
Desai K, Mekontso J, Deshpande K, Trujillo S. Preclinical Assessment of Living Therapeutic Materials: State-of-Art and Challenges. ACS Biomater Sci Eng 2025. [PMID: 40230223 DOI: 10.1021/acsbiomaterials.5c00247] [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: 04/16/2025]
Abstract
Living Therapeutic Materials represent a promising technology to tackle therapeutic problems that classical materials cannot address. Despite the advancements on new functions of these devices, new applications, and new fabrication methods, the preclinical evaluation of Living Therapeutic Materials is still very limited and new challenges appear when incorporating the living devices in contact with the host. This is a critical bottleneck in the path to translation to the clinic. Therefore, we have compiled the literature on Living Therapeutic Materials, with a focus on microorganism-based living therapeutic materials, and summarized the investigations carried out to assess their biocompatibility, safety, and efficacy. We have split the investigations in three parts: in vitro, ex vivo, and in vivo assessments, where we describe common practices and remaining challenges.
Collapse
Affiliation(s)
- Krupansh Desai
- INM-Leibniz Institute for New Materials, 66123 Saarbrucken, Germany
- Chemistry Department, Saarland University, 66123 Saarbrucken, Germany
| | - Joëlle Mekontso
- INM-Leibniz Institute for New Materials, 66123 Saarbrucken, Germany
- Chemistry Department, Saarland University, 66123 Saarbrucken, Germany
| | - Ketaki Deshpande
- INM-Leibniz Institute for New Materials, 66123 Saarbrucken, Germany
- Chemistry Department, Saarland University, 66123 Saarbrucken, Germany
| | - Sara Trujillo
- INM-Leibniz Institute for New Materials, 66123 Saarbrucken, Germany
| |
Collapse
|
5
|
Ma Z, McAninch S, Liu Z, Zhang C, Chen H, He J, Yang W, Panganiban RP, Cong Y, Yochum G, Brasier AR, Pinchuk IV, Tian B, Zhou J. Orally Bioavailable BRD4 BD1 Inhibitor ZL0516 Effectively Suppresses Colonic Inflammation in Animal Models of Inflammatory Bowel Disease. ACS Pharmacol Transl Sci 2025; 8:1152-1167. [PMID: 40242579 PMCID: PMC11997885 DOI: 10.1021/acsptsci.5c00068] [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: 01/22/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025]
Abstract
Inflammatory bowel disease (IBD), a chronic, progressive, and recurrent gastrointestinal inflammatory disorder, poses a significant threat to global health and exerts an adverse effect on the quality of life. Currently, there is a lack of effective therapies for IBD. Developing novel targeted therapies for IBD, particularly orally effective therapeutics, is a vital need for IBD patients. Herein, we first demonstrate that BRD4/NF-κB signaling is aberrantly activated in the colons of human IBD biopsy samples compared to that of normal healthy controls. ZL0516, a potent, selective, and orally bioavailable BRD4 BD1 inhibitor, significantly inhibits the TNFα- and LPS-induced expression of inflammatory cytokines in human colonic epithelial cells (HCECs) and peripheral blood mononuclear cells (PBMCs) with low cytotoxicity. Intriguingly, when administered in a preventive mode, ZL0516 significantly blocks dextran sulfate sodium (DSS)-induced murine colitis. When used in a therapeutic mode, ZL0516 effectively suppresses colonic inflammation in several IBD-relevant animal models: DSS-, oxazolone (OXA)-, and flagellin (Cbir1) T cell-induced chronic murine colitis models of IBD. ZL0516 suppresses IBD inflammatory responses in vitro and in vivo by blocking the activation of the BRD4/NF-κB signaling pathway. Also, we found that RVX208, a selective BRD4 BD2 inhibitor in Phase III clinical development, only displayed marginal effects in these IBD animal models. Collectively, our results demonstrate that specific BRD4 BD1 inhibition is a novel therapeutic strategy for IBD-associated colonic inflammation, and orally effective inhibitor ZL0516 is a promising candidate for the development of a novel therapeutic regimen against IBD.
Collapse
Affiliation(s)
- Zonghui Ma
- Chemical
Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Steven McAninch
- Department
of Medicine, Penn State Health Milton S.
Hershey Medical Center, Hershey, Pennsylvania 17033, United States
| | - Zhiqing Liu
- Chemical
Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Cun Zhang
- Chemical
Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Haiying Chen
- Chemical
Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Jing He
- Department
of Pathology, University of Texas Medical
Branch (UTMB), Galveston, Texas 77555, United States
| | - Wenjing Yang
- Division
of Gastroenterology and Hepatology, Department of Medicine, Feinberg
School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Ronaldo P. Panganiban
- Department
of Medicine, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Yingzi Cong
- Division
of Gastroenterology and Hepatology, Department of Medicine, Feinberg
School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Gregory Yochum
- Division
of Colon and Rectal Surgery, Department of Surgery, and Department
of Biochemistry and Molecular Biology, Penn
State Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033, United States
| | - Allan R. Brasier
- Institute
for Clinical and Translational Research (ICTR) School of Medicine
and Public Health, 4248 Health Sciences Learning Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Irina V. Pinchuk
- Department
of Medicine, Penn State Health Milton S.
Hershey Medical Center, Hershey, Pennsylvania 17033, United States
| | - Bing Tian
- Department
of Internal Medicine, University of Texas
Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Jia Zhou
- Chemical
Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| |
Collapse
|
6
|
Chieppa M, De Santis S, Verna G. Winnie Mice: A Chronic and Progressive Model of Ulcerative Colitis. Inflamm Bowel Dis 2025; 31:1158-1167. [PMID: 39912845 PMCID: PMC11985403 DOI: 10.1093/ibd/izaf006] [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: 12/23/2024] [Indexed: 02/07/2025]
Abstract
Recent trends show a continuous worldwide rise in the incidence of ulcerative colitis (UC), leading to increased interest in its etiology and pathogenesis, which is currently unknown. To gain a better mechanistic understanding of this disease, many mouse models have been developed over the last several years, with variations of dextran sodium sulfate administration representing the most widely employed. The Winnie mouse strain was created through elicited random mutations in Muc2, resulting in a progressive, chronic intestinal inflammation localized to the colon that worsens over time. Moreover, Winnie mice display immunologic and microbiota features that are similar to those that can be found in UC patients. Phenotypically, the presence, albeit rare, of rectal prolapse and other complications impacting quality of life can be observed in Winnie mice, as well as extraintestinal manifestations that are often associated with UC. While Winnie mice are currently less studied compared to other more established models of colitis, much has been discovered in the initial years of its use as a UC-like model. In summary, the use of Winnie mice adds to the growing armamentarium that is required to develop precision-based medicine for its future application in treating complex multifactorial diseases, such as UC.
Collapse
Affiliation(s)
- Marcello Chieppa
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy
| | - Stefania De Santis
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Giulio Verna
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
7
|
Lee NK, Lee Y, Shin DS, Choi YM, Lee J, Park E, Paik HD. Effect of Lactiplantibacillus plantarum DSW3805 Isolated from Kimchi for Gut Health Attenuating Colonic Inflammation in a Dextran Sulfate Sodium-Induced Mouse Model. Nutrients 2025; 17:1259. [PMID: 40219015 PMCID: PMC11990075 DOI: 10.3390/nu17071259] [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: 02/18/2025] [Revised: 03/19/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives:Lactiplantibacillus plantarum DSW3805 was isolated from Korean kimchi samples to examine its effect in a dextran sulfate sodium (DSS)-induced mouse model. Methods: To induce colitis, mice were treated with DSS for one week before sacrifice (n = 8 per group, total n = 40). Lacticaseibacillus rhamnosus GG (109 CFU/day) or probiotics (L. plantarum DSW3805; 108 or 109 CFU/day) were administered for two weeks. To assess colitis damage, we evaluated the disease activity index, colon tissue, inflammatory factors, the microbiome, short-chain fatty acids, and intestine-related factors. Results: DSS induced colonic tissue damage (colon length, mucus thickness, and colonic crypts), and L. plantarum DSW3805 alleviated the tissue damage. Induced inflammation was reduced by inhibiting TNF-α, IFN-γ, IL-1β, IL-6, IgA, IgG, LTB4, PGE2, and NF-κB protein expression. The ratio of Firmicutes to Bacteroidetes in the PC group (DSS-treated control) was lower than that in the NC (DSS-nontreated control); L. plantarum DSW3805 increased the ratio. Higher concentrations of acetic, propionic, and butyric acids were detected in probiotic groups. In addition, harmful factors, such as calprotectin and β-glucuronidase, were reduced in the probiotic groups. Conclusions:L. plantarum DSW3805 alleviates gut damage by colitis; therefore, it can be used as a functional food to improve gut health.
Collapse
Affiliation(s)
- Na-Kyoung Lee
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea;
| | - Yunjung Lee
- Department of Food and Nutrition, Kyungnam University, Changwon 51767, Republic of Korea; (Y.L.); (D.-S.S.)
| | - Da-Soul Shin
- Department of Food and Nutrition, Kyungnam University, Changwon 51767, Republic of Korea; (Y.L.); (D.-S.S.)
| | - Yong-Min Choi
- Daesang Wellife, Seoul 03130, Republic of Korea; (Y.-M.C.); (J.L.)
| | - Jinhyeuk Lee
- Daesang Wellife, Seoul 03130, Republic of Korea; (Y.-M.C.); (J.L.)
| | - Eunju Park
- Department of Food and Nutrition, Kyungnam University, Changwon 51767, Republic of Korea; (Y.L.); (D.-S.S.)
| | - Hyun-Dong Paik
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea;
| |
Collapse
|
8
|
Atalar K, Alim E, Yigman Z, Belen HB, Erten F, Sahin K, Soylu A, Dizakar SOA, Bahcelioglu M. Transauricular vagal nerve stimulation suppresses inflammatory responses in the gut and brain in an inflammatory bowel disease model. J Anat 2025; 246:602-615. [PMID: 39707162 PMCID: PMC11911132 DOI: 10.1111/joa.14178] [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: 07/01/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 12/23/2024] Open
Abstract
Inflammatory bowel disease (IBD) encompasses Crohn's disease (CD) and ulcerative colitis (UC), is a major health problem on a global scale and its treatment is unsatisfactory. We aimed to investigate the effects of transauricular vagal nerve stimulation (tVNS) on inflammation in rats with IBD induced by trinitrobenzene sulfonic acid (TNBS). A total of 36 adult female Sprague-Dawley rats were given TNBS, or vehicle, and tVNS, or sham, every other day for 30 min for 10 days. Postmortem macroscopic and microscopic colon morphology were evaluated by histological staining. Additionally, IL-1β, IL-6, IL-10, and TNF-α cytokine levels in the colon and the brain were evaluated by immunohistochemistry and western blotting analysis. TNBS induced epithelial damage, inflammation, ulceration, and thickened mucosal layer in the colonic tissues. Administration of tVNS significantly ameliorated the severity of TNBS-induced tissue damage and inflammatory response. TNBS also alters pro-inflammatory and anti-inflammatory balance in the brain tissue. TVNS application significantly suppressed the protein levels of pro-inflammatory cytokines, namely IL-1β, IL-6, and TNF- α while augmenting the level of anti-inflammatory cytokine IL-10 in the colonic and the brain tissue. We have shown that TNBS-mediated colonic inflammation and tissue damage are associated with neuroinflammatory responses in the brain tissue. Also demonstrated for the first time that neuroinflammatory response in the gut-brain axis is suppressed by tVNS in the IBD model. Non-invasive tVNS stands out as a new potential treatment option for types of IBD.
Collapse
Affiliation(s)
- Kerem Atalar
- Department of Anatomy, Faculty of Medicine, Neuroscience and Neurotechnology Center of Excellence (NÖROM) and Neuropsychiatry Center, Gazi University, Ankara, Türkiye
| | - Ece Alim
- Department of Anatomy, Faculty of Medicine, Neuroscience and Neurotechnology Center of Excellence (NÖROM), Gazi University, Ankara, Türkiye
| | - Zeynep Yigman
- Department of Histology and Embryology, Faculty of Medicine, Neuroscience and Neurotechnology Center of Excellence (NÖROM), Gazi University, Türkiye
| | - Hayrunnisa Bolay Belen
- Department of Neurology and Algology, Faculty of Medicine, Neuroscience and Neurotechnology Center of Excellence (NÖROM), Neuropsychiatry Center, Gazi University, Ankara, Türkiye
| | - Fusun Erten
- Department of Veterinary Medicine, Pertek Sakine Genc Vocational School, Munzur University, Tunceli, Türkiye
| | - Kazım Sahin
- Department of Animal Nutrition and Nutritional Diseases, Faculty of Veterinary Medicine, Fırat University, Elazığ, Türkiye
| | - Ayse Soylu
- Department of Anatomy Faculty of Medicine, Gazi University, Ankara, Türkiye
| | | | - Meltem Bahcelioglu
- Department of Anatomy, Faculty of Medicine, Neuroscience and Neurotechnology Center of Excellence (NÖROM) and Neuropsychiatry Center, Gazi University, Ankara, Türkiye
| |
Collapse
|
9
|
Muro P, Jing C, Zhao Z, Jin T, Mao F. The emerging role of honeysuckle flower in inflammatory bowel disease. Front Nutr 2025; 12:1525675. [PMID: 40225345 PMCID: PMC11985448 DOI: 10.3389/fnut.2025.1525675] [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: 11/10/2024] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Crohn's disease (CD) and ulcerative colitis (UC), referred to as inflammatory bowel disease (IBD), pose considerable challenges in treatment because they are chronic conditions that easily relapse. The occurrence of IBD continues to rise in developing countries. Nonetheless, the existing therapies for IBD have limitations and fail to address the needs of the patients thoroughly. There is an increasing need for new, safe, and highly effective alternative medications for IBD patients. Traditional Chinese Medicine (TCM) is employed in drug development and disease management due to its wide-range of biological activities, minimal toxicity, and limited side effects. Extensive research has shown that certain TCM exhibits significant therapeutic benefits for IBD treatments. Honeysuckle (Lonicera japonica) was used in TCM research and clinical settings for the treatment of IBD. Bioactive metabolites in L. japonica, such as luteolin, quercetin, cyanidin, chlorogenic acid (CGA), caffeic acid (CA), and saponin, exhibit significant therapeutic benefits for managing IBD. The honeysuckle flower is a potential candidate in the treatment of IBD due to its anti-inflammatory, immune system-regulating, and antioxidant qualities. This paper reviews the metabolites of the honeysuckle flower as a candidate for the treatment of IBD. It discusses the fundamental mechanism of L. japonica and the potential of its bioactive metabolites in the prevention and treatment of IBD.
Collapse
Affiliation(s)
- Peter Muro
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Caihong Jing
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu, China
| | - Zhihan Zhao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Tao Jin
- Department of Gastrointestinal and Endoscopy, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Fei Mao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
10
|
Liu J, Dai Y, Yang W, Chen ZY. Role of Mushroom Polysaccharides in Modulation of GI Homeostasis and Protection of GI Barrier. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6416-6441. [PMID: 40063730 PMCID: PMC11926878 DOI: 10.1021/acs.jafc.5c00745] [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: 03/20/2025]
Abstract
Edible and medicinal mushroom polysaccharides (EMMPs) have been widely studied for their various biological activities. It has been shown that EMMPs could modulate microbiota in the large intestine and improve intestinal health. However, the role of EMMPs in protecting the gastric barrier, regulating gastric microbiota, and improving gastric health cannot be ignored. Hence, this review will elucidate the effect of EMMPs on gastric and intestinal barriers, with emphasis on the interaction of EMMPs with microbiota in maintaining overall gastrointestinal health. Additionally, this review highlights the gastroprotective effects and underlying mechanisms of EMMPs against gastric mucosa injury, gastritis, gastric ulcer, and gastric cancer. Furthermore, the effects of EMMPs on intestinal diseases, including inflammatory bowel disease, colorectal cancer, and intestinal infection, are also summarized. This review will also discuss the future perspective and challenges in the use of EMMPs as a dietary supplement or a nutraceutical in preventing and treating gastrointestinal diseases.
Collapse
Affiliation(s)
- Jianhui Liu
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong 999077, China
| | - Yi Dai
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Wenjian Yang
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong 999077, China
| |
Collapse
|
11
|
van der Geest R, Lee JS. Role of the basic leucine zipper transcription factor BATF2 in modulating immune responses and inflammation in health and disease. J Leukoc Biol 2025; 117:qiae245. [PMID: 39504573 PMCID: PMC11953073 DOI: 10.1093/jleuko/qiae245] [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: 07/20/2024] [Accepted: 11/06/2024] [Indexed: 11/08/2024] Open
Abstract
BATF2 is a transcription factor known to exhibit tumor-suppressive activity in cancer cells. Within recent years, however, BATF2 has also emerged as an important transcriptional regulator of the immune system. Through its immunomodulatory function, BATF2 has been implicated in a variety of (patho)physiological processes, including host defense against infection, antitumor immunity, and maintenance of tissue inflammatory homeostasis. Below, we discuss recent literature that has provided insight into the role of BATF2 as a transcriptional regulator of immune responses in health and disease, including the cell types that express BATF2, the different diseases in which the immunomodulatory effects of BATF2 have been shown to play a role, and the molecular mechanisms through which BATF2 is thought to exert those effects. In doing so, we highlight that the immunological effects of BATF2 are highly context dependent, and we point out the overlap between the mechanisms of action of BATF2 in infectious and noninfectious diseases. We also discuss areas of interest for future research, the clinical relevance of better understanding BATF2 function, and potential strategies for therapeutic modulation of BATF2.
Collapse
Affiliation(s)
- Rick van der Geest
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, 3459 Fifth Avenue, Pittsburgh, PA 15213, United States
| | - Janet S Lee
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, 3459 Fifth Avenue, Pittsburgh, PA 15213, United States
- Vascular Medicine Institute, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, United States
- Division of Pulmonary and Critical Care Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St. Louis, MO 63110, United States
| |
Collapse
|
12
|
Bhatnagar P, Elhariri S, Burud IAS, Eid N. Perianal fistulizing Crohn's disease: Mechanisms and treatment options focusing on cellular therapy. World J Gastroenterol 2025; 31:100221. [PMID: 40061590 PMCID: PMC11886048 DOI: 10.3748/wjg.v31.i9.100221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 01/06/2025] [Accepted: 01/21/2025] [Indexed: 02/18/2025] Open
Abstract
Perianal fistulizing Crohn's disease (PFCD) is a common presentation of CD, which affects the patients' quality of life, including social and sexual function. The management of PFCD remains a critical challenge in inflammatory bowel disease, primarily due to limited understanding of the mechanisms involved in its pathogenesis, complicating medical treatment. Increased production of inflammatory cytokines such as tumor necrosis factor and interleukin-13 by infiltrating macrophages and other inflammatory cells stimulate the epithelial-to-mesenchymal transition, resulting in activation of myofibroblasts and elevation of matrix metalloproteinases, leading to fistula formation. Given the potential for malignant transformation, PFCD screening is critical. Cytokine and inflammation-targeted therapies can help control this disease, but recurrence is a common complication. Surgical interventions such as fistulotomy represent viable therapeutic options, with magnetic resonance imaging serving as an important diagnostic tool for delineating fistula tract anatomy. Animal models and clinical trials demonstrate that injection of mesenchymal stem cells (MSCs) into the fistula results in suppression of the inflammatory cells and cytokines and complete resolution of PFCD. Recently, MSC-derived extracellular vesicles were found to stimulate fistula healing, with encouraging results. In this article, we comment on the review article by Pacheco et al, summarizing the various lines of PFCD treatment and highlighting the role of screening for this disease. Importantly, we focus on the various mechanisms involved in the pathogenesis of PFCD, the therapeutic roles of MSCs and related extracellular vesicles, and explore the potential role of autophagy in enhancing the therapeutic efficacy of these cells, which may help in the treatment of this disease.
Collapse
Affiliation(s)
- Payal Bhatnagar
- Department of Pharmaceutical Technology, School of Pharmacy, IMU University, Kuala Lumpur 57000, Malaysia
| | - Sherreen Elhariri
- Department of Surgery, IMU University, Clinical Campus, Seremban 70300, Negeri Sembilan, Malaysia
| | - Ismail A S Burud
- Department of Surgery, IMU University, Clinical Campus, Seremban 70300, Negeri Sembilan, Malaysia
| | - Nabil Eid
- Department of Anatomy, Division of Human Biology, School of Medicine, IMU University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
13
|
Kumar A, Husain N, Anbazhagan AN, Jayawardena D, Priyamvada S, Singhal M, Jain C, Kaur P, Guzman G, Saksena S, Dudeja PK. Dexamethasone Upregulates the Expression of the Human SLC26A3 (DRA, Down-Regulated in Adenoma) Transporter (an IBD Susceptibility Gene) in Intestinal Epithelial Cells and Attenuates Gut Inflammation. Inflamm Bowel Dis 2025; 31:625-635. [PMID: 39657154 DOI: 10.1093/ibd/izae271] [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: 07/19/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Down-Regulated in Adenoma (DRA) plays a critical role in intestinal chloride absorption and a decrease in its expression is a key event in diarrheal disorders. Recently, DRA has emerged as an Inflammatory Bowel Disease (IBD) susceptibility gene. Therefore, the strategies to upregulate DRA expression are potentially novel approaches to not only treat IBD-associated diarrhea but also gut inflammation. In this study, the effect of dexamethasone (DEX), an anti-inflammatory corticosteroid on DRA expression was investigated. METHODS GR (glucocorticoid receptor) overexpressed Caco-2 cells and C57BL/6/J mice and anti-αIL-10R mAb model of IBD were used. Protein expression was assessed by immunoblotting and immunofluorescence. Transcript levels were assessed by quantative-real-time polymerase chain reaction (qRT-PCR) and promoter activity was measured by luciferase assays. RESULTS Our results showed that DEX significantly increased DRA mRNA and protein expression in GR overexpressing Caco-2 cells. DEX-induced upregulation of DRA was GR dependent and appeared at least in part to occur via a transcriptional mechanism, as promoter activity of the DRA construct (-1183/+114 bp) was significantly increased in response to DEX. The increase in DRA mRNA was abrogated in the presence of MKP-1 inhibitor, triptolide. Administration of DEX (2 mg/kg body weight) to mice for 24 and 48 hours significantly increased the DRA expression in mouse colon. DEX treatment to mice for 7 days in the αIL-10R mAb model of colitis was able to significantly attenuate the gut inflammation and associated decrease in DRA expression. CONCLUSIONS We demonstrate that DEX stimulates DRA expression via transcriptional mechanisms and suggest that upregulation of DRA may contribute to both anti-inflammatory and pro-absorptive effects of DEX.
Collapse
Affiliation(s)
- Anoop Kumar
- Jesse Brown VA Medical Center, Research and Development, Chicago, IL, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Nazim Husain
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Arivarasu N Anbazhagan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Dulari Jayawardena
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Shubha Priyamvada
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Megha Singhal
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Charu Jain
- Jesse Brown VA Medical Center, Research and Development, Chicago, IL, USA
| | - Prabhdeep Kaur
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Grace Guzman
- Department of Pathology, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Seema Saksena
- Jesse Brown VA Medical Center, Research and Development, Chicago, IL, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Pradeep K Dudeja
- Jesse Brown VA Medical Center, Research and Development, Chicago, IL, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| |
Collapse
|
14
|
Chen D, Bi X, Feng Q, Sun Y. Supplementation with Lentil ( Lens culinaris) Hull Soluble Dietary Fiber Ameliorates Sodium Dextran Sulfate-Induced Colitis and Behavioral Deficits via the Gut-Brain Axis. Foods 2025; 14:870. [PMID: 40077572 PMCID: PMC11898428 DOI: 10.3390/foods14050870] [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: 02/08/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
In this study, the impact of lentil hull soluble dietary fibers (SDFs) on colitis and behavioral deficits in mice was assessed. Structural characterizations of SDFs confirmed that cellulase-modified soluble dietary fiber exhibited better physicochemical properties: more porous microstructure; similar polysaccharide structure; more stable particle size distribution; higher crystallinity; better adsorption capacity; and lower viscosity. Additionally, we explored its potential cognitive benefits via the gut-brain axis by behavioral tests, histopathology, 16S rRNA sequencing, gas chromatography and metabolomics analysis. The results showed that SDFs significantly improved inflammatory symptoms in colon and brain and cognitive behaviors. LSDF had better efficacy than HSDF. LSDF intervention decreased the harmful bacteria abundance (Bacteroides, Flexispira and Escherichia, etc.) and increased beneficial bacteria abundance (Aggregatibacter and Helicobacter, etc.). LSDF also affected brain metabolites through the sphingolipid metabolism. Spearman correlation analysis showed that there was a positive correlation between harmful bacteria with inflammatory factors (LPS, IL-1β, IL-6, and TNF-α, etc.) and sphingolipid metabolites, while beneficial bacteria were positively correlated with brain-derived neurotrophic factor (BDNF), IL-10, and cognitive behavior. This study highlights the value of SDFs in future diet-based therapeutic strategies targeting gut-brain interactions.
Collapse
Affiliation(s)
- Dongying Chen
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China;
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China;
| | - Xin Bi
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China;
| | - Qian Feng
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China;
| | - Yong Sun
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China;
| |
Collapse
|
15
|
Shi S, Jiang H, Ma W, Guan Z, Han M, Man S, Wu Z, He S. Preclinical studies of natural flavonoids in inflammatory bowel disease based on macrophages: a systematic review with meta-analysis and network pharmacology. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2293-2318. [PMID: 39422746 DOI: 10.1007/s00210-024-03501-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
Flavonoid is a category of bioactive polyphenolic compounds that are extensively distributed in plants with specific pharmacological properties, such as anti-inflammatory and anti-oxidant. Importantly, natural flavonoids have shown the protected function on the dextran sulfate sodium (DSS)-induced colitis in animals and lipopolysaccharides (LPS)-induced inflammatory response in macrophages. The purpose of this systematic review is to explore the efficacy of natural flavonoids in animal models of IBD (inflammatory bowel disease) and potential mechanisms in macrophages by meta-analysis and network pharmacology in preclinical studies. Relevant foundation studies were searched from January 2010 to November 2023 in databases like PubMed, Elsevier ScienceDirect, and Web of Science. Then, OriginPro software was used to extract values from images, and the analysis was performed using Review Manager 5.3. The retrieved data was analyzed according to the fixed-effects model and random-effects model. Subsequently, heterogeneity was evaluated using the I2 statistics. Lastly, network pharmacology was applied to confirm mechanisms of natural flavonoids on IBD. According to the results of meta-analysis, we found the natural flavonoids exhibited powerful therapeutic effects against IBD, which not only reversed colonic shortness (WMD = 1.33, 95% CI (1.07, 1.59), P < 0.00001), but also reduced histological score (SMD = - 2.66, 95% CI (- 3.77, - 1.95), P < 0.00001) between natural flavonoid treatment groups compared with the experimental IBD model. Furthermore, treatment with natural flavonoids decreased the levels of tumor necrosis factor-α (TNF-α) in macrophages. Mechanistically, our summarized data substantiate that natural flavonoids alleviate LPS-induced M1 macrophage polarization, anti-oxidant, anti-inflammatory, maintain intestinal barrier, and inhibit the activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome in macrophages. Moreover, the results of network pharmacology also support this. This systematic review demonstrated the efficiency of natural flavonoids in treating IBD in preclinical research by meta-analysis and network pharmacology, which offered supporting evidence for clinical trial implementation. However, some limitations remain present, such as technique quality shortage, missed reports on account of negative results, failure to count sample size, and the risk of bias.
Collapse
Affiliation(s)
- Shasha Shi
- Pharmacy School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Hao Jiang
- Pharmacy School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wenke Ma
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zitong Guan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Mengxue Han
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Shuai Man
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Zhuzhu Wu
- Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Shan He
- Pharmacy School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
16
|
Liu X, Song Y, Shen M, Liu X, Zhang W, Jiang H, Han M. Smooth Muscle Silent Information Regulator 1 Contributes to Colitis in Mice. Int J Mol Sci 2025; 26:1807. [PMID: 40076434 PMCID: PMC11898484 DOI: 10.3390/ijms26051807] [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: 01/08/2025] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Smooth muscle cells (SMCs) are an essential component of the intestine, play an important role to maintain intestine structure, and produce peristaltic and segmentation movements. The silent information regulator 1 (SIRT1) has a dual role along with possible mechanisms in the different experimental models of inflammatory bowel disease (IBD). However, very little is known about other putative roles that overexpression of SIRT1 in SMCs may have. Here, we explored the role of SMC SIRT1 in colonic mucosa regeneration and recovery after DSS-induced colitis. We showed that smooth-muscle-specific SIRT1 transgene (Sirt1-Tg) mice have abnormal baseline intestinal architecture. The overexpression of SIRT1 impaired the recovery after DSS-induced injury. Furthermore, we showed that smooth-muscle SIRT1 affected the intestinal epithelial regeneration after damage by releasing cZFP609, which inhibited the hypoxia-inducible factor (HIF)-1α nuclear translocation. Together, we identify an important signaling axis cZFP609-HIF-1α linking SMCs and intestinal epithelium, which is involved in colitis development.
Collapse
Affiliation(s)
| | | | | | | | | | - Haibin Jiang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
17
|
Ramadan Q, Hazaymeh R, Zourob M. A Versatile and Modular Microfluidic System for Dynamic Cell Culture and Cellular Interactions. MICROMACHINES 2025; 16:237. [PMID: 40047680 PMCID: PMC11857257 DOI: 10.3390/mi16020237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/07/2025] [Accepted: 02/15/2025] [Indexed: 03/09/2025]
Abstract
A versatile and modular microfluidic system for cell co-culture has been developed. Microfluidic chips, each featuring dual compartments separated by a porous membrane, have been fabricated and assembled within the system to facilitate fluidic interconnection and cell-cell communication through the chip assembly. A set of fluidic valves has been successfully integrated to regulate the flow through the chip assembly. The system allows for chip assembly in various arrangements, including in parallel, in series, and complex connections. Individual chips can be interconnected or disconnected within the system at any time. Moreover, the spatial order and orientation of the chips can be adjusted as needed, enabling the study of different cell-cell arrangements and the impact of the presence or absence of specific cell types. The utility of the system has been evaluated by culturing and interconnecting multi-monolayers of intestinal epithelial cells as a model of the complex cellular system. Epithelial monolayers were grown in multiple chips and interconnected in various configurations. The transepithelial electrical resistance and permeability profiles were investigated in detail for these configurations upon treatment of the cells with dextran sulfate sodium. Immune cells were stimulated through the epithelial layers and the expression of inflammatory cytokines was detected. This miniaturized platform offers controlled conditions for co-culturing key cellular components and assessing potential therapeutic agents in a physiologically relevant setting.
Collapse
Affiliation(s)
- Qasem Ramadan
- College of Science & General Studies, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Rana Hazaymeh
- College of Pharmacy, Almaarefa University, Riyadh 13713, Saudi Arabia;
| | - Mohammed Zourob
- College of Science & General Studies, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
18
|
Dong XQ, Zhang YH, Luo J, Li MJ, Ma LQ, Qi YT, Miao YL. Keratin 1 modulates intestinal barrier and immune response via kallikrein kinin system in ulcerative colitis. World J Gastroenterol 2025; 31:102070. [PMID: 39958441 PMCID: PMC11752705 DOI: 10.3748/wjg.v31.i6.102070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/06/2024] [Accepted: 12/19/2024] [Indexed: 01/10/2025] Open
Abstract
BACKGROUND External factors in ulcerative colitis (UC) exacerbate colonic epithelial permeability and inflammatory responses. Keratin 1 (KRT1) is crucial in regulating these alterations, but its specific role in the progression of UC remains to be fully elucidated. AIM To explore the role and mechanisms of KRT1 in the regulation of colonic epithelial permeability and inflammation in UC. METHODS A KRT1 antibody concentration gradient test, along with a dextran sulfate sodium (DSS)-induced animal model, was implemented to investigate the role of KRT1 in modulating the activation of the kallikrein kinin system (KKS) and the cleavage of bradykinin (BK)/high molecular weight kininogen (HK) in UC. RESULTS Treatment with KRT1 antibody in Caco-2 cells suppressed cell proliferation, induced apoptosis, reduced HK expression, and increased BK expression. It further downregulated intestinal barrier proteins, including occludin, zonula occludens-1, and claudin, and negatively impacted the coagulation factor XII. These changes led to enhanced activation of BK and HK cleavage, thereby intensifying KKS-mediated inflammation in UC. In the DSS-induced mouse model, administration of KRT1 antibody mitigated colonic injury, increased colon length, alleviated weight loss, and suppressed inflammatory cytokines such as interleukin (IL)-1, IL-6, tumor necrosis factor-α. It also facilitated repair of the intestinal barrier, reducing DSS-induced injury. CONCLUSION KRT1 inhibits BK expression, suppresses inflammatory cytokines, and enhances markers of intestinal barrier function, thus ameliorating colonic damage and maintaining barrier integrity. KRT1 is a viable therapeutic target for UC.
Collapse
Affiliation(s)
- Xiang-Qian Dong
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan Province, China
| | - Ying-Hui Zhang
- Department of Gastroenterology, Affiliated Hospital of Yunnan University, Kunming 650021, Yunnan Province, China
| | - Juan Luo
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan Province, China
| | - Mao-Juan Li
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan Province, China
| | - Lan-Qing Ma
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan Province, China
| | - Ya-Ting Qi
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan Province, China
| | - Ying-Lei Miao
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan Province, China
| |
Collapse
|
19
|
Scharf E, Schlattmann P, Stallhofer J, Stallmach A. Do Antibiotics Cause Inflammatory Bowel Disease? A Systematic Review and Meta-Analysis. Visc Med 2025; 41:32-47. [PMID: 39927188 PMCID: PMC11801854 DOI: 10.1159/000541601] [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: 05/18/2024] [Accepted: 09/23/2024] [Indexed: 02/11/2025] Open
Abstract
Introduction Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), exhibits a multifactorial pathogenesis influenced by genetic and environmental factors. Antibiotic usage has been implicated in modifying the gut microbiome, potentially leading to dysbiosis and contributing to IBD risk. Despite existing literature, the relationship remains inconclusive. This meta-analysis aimed to evaluate the association between prior antibiotic use and the onset of IBD. Methods A systematic literature search in PubMed was conducted to identify studies exploring the link between antibiotic use and subsequent IBD diagnosis. Studies reporting CD, UC, or both as primary outcomes were included. The meta-analysis, performed according to PRISMA guidelines, summarized risk estimates, represented as odds ratios (ORs), and corresponding confidence intervals (CIs). Subgroup analyses involved the categorization of antibiotics and the determination of the minimum number of antibiotic therapy courses administered. Results Out of 722 publications, 31 studies comprising 102,103 individuals met eligibility criteria. The pooled OR for IBD in those with prior antibiotic exposure was 1.40 (95% CI: 1.25-1.56). Antibiotic use was associated with an increased risk of IBD (OR: 1.52, 95% CI: 1.19-1.94). Notably, this association was confined to CD (OR: 1.50, 95% CI: 1.27-1.77), while no significant association was observed with UC (OR: 1.21, 95% CI: 1.00-1.47). Risk augmentation for IBD correlated positively with the number of antibiotic courses (OR: 1.08, 95% CI: 1.05-1.12). Conclusion Previous antibiotic use is associated with the later development of CD. A positive dose-response effect was also observed. Against this background, antibiotics should be used rationally.
Collapse
Affiliation(s)
- Ellen Scharf
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Peter Schlattmann
- Institute for Medical Statistics, Informatics, and Data Science, Jena University Hospital, Jena, Germany
| | - Johannes Stallhofer
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Andreas Stallmach
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| |
Collapse
|
20
|
Ham N, Park M, Bae YA, Yeo EJ, Jung Y. Differential pathological changes in colon microenvironments in acute and chronic mouse models of inflammatory bowel disease. Anim Cells Syst (Seoul) 2025; 29:100-112. [PMID: 39839656 PMCID: PMC11748878 DOI: 10.1080/19768354.2025.2451408] [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: 10/13/2024] [Revised: 12/09/2024] [Accepted: 01/03/2025] [Indexed: 01/23/2025] Open
Abstract
Inflammatory bowel disease is a chronic condition characterized by inflammation of the gastrointestinal tract, resulting from an abnormal immune response to normal stimuli, such as food and intestinal flora. Since the etiology of this disease remains largely unknown, murine models induced by the consumption of dextran-sodium sulfate serve as a pivotal tool for studying colon inflammation. In this study, we employed both acute and chronic colitis mouse models induced by varying durations of dextran-sodium sulfate consumption to investigate the pathological and immunologic characteristics throughout the disease course. During the acute phase, activated innate inflammation marked by M1 macrophage infiltration was prominent. In contrast, the chronic phase was characterized by tissue remodeling, with a significant increase in M2 macrophages and lymphocytes. RNA-sequencing revealed genetic changes in acute and chronic colitis, marked by the maintenance of genomic integrity in the acute phase and extracellular matrix dynamics in the chronic phase. These phase-specific alterations reflect the multifaceted physiological processes involved in the initiation and progression of inflammation in the large intestine, underscoring the necessity for distinct experimental approaches for each phase. The findings demonstrate that the factors shaping the large intestinal immune microenvironment change specifically during the acute and chronic phases of experimental inflammatory bowel disease, highlighting the importance of developing therapeutic strategies that align with the disease course.
Collapse
Affiliation(s)
- NaYeon Ham
- Department of Microbiology, Graduate School of Medicine, Gachon University, Incheon, South Korea
| | - Minji Park
- Department of Health Science and Technology, Gachon Advanced Institute for Health Science & Technology, Gachon University, Incheon, South Korea
| | - Young-An Bae
- Department of Microbiology, College of Medicine, Lee Gil Ya Cancer and Diabetes Institute Incheon, Gachon University, Incheon, South Korea
| | - Eui-Ju Yeo
- Department of Biochemistry, College of Medicine, Lee Gil Ya Cancer and Diabetes Institute Incheon, Gachon University, Incheon, South Korea
| | - YunJae Jung
- Department of Microbiology, Graduate School of Medicine, Gachon University, Incheon, South Korea
- Department of Health Science and Technology, Gachon Advanced Institute for Health Science & Technology, Gachon University, Incheon, South Korea
- Department of Microbiology, College of Medicine, Lee Gil Ya Cancer and Diabetes Institute Incheon, Gachon University, Incheon, South Korea
| |
Collapse
|
21
|
Borup A, Sharifpour MF, Rossen LS, Whitehead B, Boysen AT, Olesen R, Bohn AB, Ridolfi A, Brucale M, Valle F, Paolini L, Radeghieri A, Bergese P, Miles K, Veitch M, Thomas T, Ruscher R, Wangchuk P, Giacomin P, Loukas A, Nejsum P. Helminth extracellular vesicles co-opt host monocytes to drive T cell anergy. J Extracell Vesicles 2025; 14:e70027. [PMID: 39815783 PMCID: PMC11735955 DOI: 10.1002/jev2.70027] [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: 07/05/2024] [Accepted: 11/30/2024] [Indexed: 01/18/2025] Open
Abstract
Parasitic helminths secrete extracellular vesicles (EVs) into their host tissues to modulate immune responses, but the underlying mechanisms are poorly understood. We demonstrate that Ascaris EVs are efficiently internalised by monocytes in human peripheral blood mononuclear cells and increase the percentage of classical monocytes. Furthermore, EV treatment of monocytes induced a novel anti-inflammatory phenotype characterised by CD14+, CD16-, CC chemokine receptor 2 (CCR2-) and programmed death-ligand 1 (PD-L1)+ cells. In addition, Ascaris EVs induced T cell anergy in a monocyte-dependent mechanism. Targeting professional phagocytes to induce both direct and indirect pathways of immune modulation presents a highly novel and efficient mechanism of EV-mediated host-parasite communication. Intra-peritoneal administration of EVs induced protection against gut inflammation in the dextran sodium sulphate model of colitis in mice. Ascaris EVs were shown to affect circulating immune cells and protect against gut inflammation; this highlights their potential as a subject for further investigation in inflammatory conditions driven by dysregulated immune responses. However, their clinical translation would require further studies and careful consideration of ethical implications.
Collapse
Affiliation(s)
- Anne Borup
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Infectious DiseasesAarhus University HospitalAarhusDenmark
| | | | - Litten S. Rossen
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Infectious DiseasesAarhus University HospitalAarhusDenmark
| | - Bradley Whitehead
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Infectious DiseasesAarhus University HospitalAarhusDenmark
| | - Anders T. Boysen
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Infectious DiseasesAarhus University HospitalAarhusDenmark
| | - Rikke Olesen
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Infectious DiseasesAarhus University HospitalAarhusDenmark
| | - Anja B. Bohn
- Department of Biomedicine, FACS Core FacilityAarhus UniversityAarhusDenmark
| | - Andrea Ridolfi
- Department of Physics and Astronomy and LaserLaB AmsterdamVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Marco Brucale
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase (CSGI)University of FlorenceFlorenceItaly
- Consiglio Nazionale delle Ricerche (CNR)Istituto per lo Studio dei Materiali Nanostrutturati (ISMN)University of BolognaBolognaItaly
| | - Francesco Valle
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase (CSGI)University of FlorenceFlorenceItaly
- Consiglio Nazionale delle Ricerche (CNR)Istituto per lo Studio dei Materiali Nanostrutturati (ISMN)University of BolognaBolognaItaly
| | - Lucia Paolini
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase (CSGI)University of FlorenceFlorenceItaly
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
| | - Annalisa Radeghieri
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase (CSGI)University of FlorenceFlorenceItaly
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
| | - Paolo Bergese
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase (CSGI)University of FlorenceFlorenceItaly
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Consiglio Nazionale delle Ricerche (CNR), Institute for Research and Biomedical Innovation (IRIB)University of PalermoPalermoItaly
| | - Kim Miles
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsQueenslandAustralia
| | - Margaret Veitch
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsQueenslandAustralia
| | - Tamara Thomas
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsQueenslandAustralia
| | - Roland Ruscher
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsQueenslandAustralia
| | - Phurpa Wangchuk
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsQueenslandAustralia
| | - Paul Giacomin
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsQueenslandAustralia
| | - Alex Loukas
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsQueenslandAustralia
| | - Peter Nejsum
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Infectious DiseasesAarhus University HospitalAarhusDenmark
| |
Collapse
|
22
|
Kelly C, Sartor RB, Rawls JF. Early subclinical stages of the inflammatory bowel diseases: insights from human and animal studies. Am J Physiol Gastrointest Liver Physiol 2025; 328:G17-G31. [PMID: 39499254 PMCID: PMC11901386 DOI: 10.1152/ajpgi.00252.2024] [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: 08/16/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/07/2024]
Abstract
The inflammatory bowel diseases (IBD) occur in genetically susceptible individuals that mount inappropriate immune responses to their microbiota leading to chronic intestinal inflammation. The natural history of IBD progression begins with early subclinical stages of disease that occur before clinical diagnosis. Improved understanding of those early subclinical stages could lead to new or improved strategies for IBD diagnosis, prognostication, or prevention. Here, we review our current understanding of the early subclinical stages of IBD in humans including studies from first-degree relatives of patients with IBD and members of the general population who go on to develop IBD. We also discuss representative mouse models of IBD that can be used to investigate disease dynamics and host-microbiota relationships during these early stages. In particular, we underscore how mouse models of IBD that develop disease later in life with variable penetrance may present valuable opportunities to discern early subclinical mechanisms of disease before histological inflammation and other severe symptoms become apparent.
Collapse
Affiliation(s)
- Cecelia Kelly
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| | - R Balfour Sartor
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
23
|
Zeng C, Zhu Q, Peng W, Huang C, Chen H, Huang H, Zhou Y, Zhao C. The protective effect of amitriptyline on experimental colitis through inhibiting TLR-4/MD-2 signaling pathway. J Pharmacol Exp Ther 2025; 392:100024. [PMID: 39892990 DOI: 10.1124/jpet.124.002207] [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: 03/08/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 10/10/2024] Open
Abstract
Amitriptyline, a pleiotropic tricyclic antidepressant, possesses antioxidant and anti-inflammatory properties. Despite its diverse benefits, the specific effects of amitriptyline on inflammatory bowel disease (IBD) are not yet well defined. To explore this, we used a dextran sulfate sodium (DSS)-induced colitis model to examine the anti-inflammatory effects of amitriptyline and the underlying mechanisms by which it operates. Our research revealed that amitriptyline is effective in alleviating several pathological manifestations associated with colitis. This includes improving body weight retention, reducing disease activity index, lessening of colon length shortening, and repairing of colonic mucosal damage. Treatment with amitriptyline significantly protected mucosal injury by preserving the population of goblet cells and increasing the expression of tight junction proteins. Furthermore, we observed that amitriptyline effectively countered immune cell infiltration, specifically neutrophils and macrophages, while simultaneously lowering the levels of inflammatory cytokines such as tumor necrosis factor α, interleukin (IL)-1β, and IL-6. Additionally, RNA sequencing analysis pointed to the potential involvement of the Toll-like receptor (TLR) pathway in the anticolitic effects induced by amitriptyline. Subsequent Western blot analysis indicated that amitriptyline significantly inhibited the TLR-4-mediated nuclear factor (NF)-κB signaling pathway. To bolster our findings, in vitro studies demonstrated that amitriptyline downregulated the TLR-4/NF-κB/mitogen-activated protein kinase signaling cascades in mouse macrophages stimulated with lipopolysaccharide. Further molecular investigations revealed that amitriptyline was able to suppress the elevated expression of myeloid differentiation factor 2 that lipopolysaccharide stimulation typically induces. In summary, our findings suggest that amitriptyline effectively mitigates DSS-induced colitis in mice through the inhibition of TLR-4/myeloid differentiation 2 pathway signaling, indicating its potential repurposing for IBD treatment. SIGNIFICANCE STATEMENT: The potential of using amitriptyline in treating inflammatory bowel disease appears promising, leveraging its established safety and dosing profile as an antidepressant. The study results show that amitriptyline can alleviate pathological symptoms, inflammation, and intestinal mucosal damage in mice with colitis induced by DSS. The protective effect observed appears to be linked to the inhibition of TLR-4/myeloid differentiation 2 signaling pathway. By exploring novel applications for existing medications, we can optimize amitriptyline's efficacy and broaden its impact in both medical and commercial contexts.
Collapse
Affiliation(s)
- Chengcheng Zeng
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Qingqing Zhu
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Wu Peng
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Chen Huang
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Huiting Chen
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Hongli Huang
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Yongjian Zhou
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Chong Zhao
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China.
| |
Collapse
|
24
|
Araruna MEC, Alves Júnior EB, de Lima Serafim CA, Pessoa MMB, de Souza Pessôa ML, Alves VP, Sobral MV, da Silva MS, Alves AF, de Paiva Sousa MC, Araújo AA, Batista LM. (-)-Fenchone Ameliorates TNBS-Induced Colitis in Rats via Antioxidant, Immunomodulatory, and Cytoprotective Mechanisms. Pharmaceuticals (Basel) 2024; 18:18. [PMID: 39861081 PMCID: PMC11769309 DOI: 10.3390/ph18010018] [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: 06/20/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND (-)-Fenchone is a bicyclic monoterpene present in the plant species Foeniculum vulgare Mill, Thuja occidentalis L. (tuja), and Lavandula stoechas (lavender). These plants have therapeutic value in the treatment of intestinal disorders. AIM To evaluate intestinal anti-inflammatory activity in an acute and chronic trinitrobenzene sulphonic acid (TNBS)-induced colitis model in rats. METHODS Intestinal anti-inflammatory effects were assessed using the acute and chronic TNBS-induced colitis model in rats. The mechanisms were evaluated from colonic tissue fragments of the acute and chronic models. RESULTS Oral administration of the (-)-fenchone (37.5-300 mg/kg) acute phase or (150 mg/kg) (p < 0.001) chronic phase reduced the macroscopic lesion score, ulcerative area, intestinal weight/length ratio, and diarrheal index in TNBS-treated animals. At a dose of 150 mg/kg, the acute and chronic phase decreased malondialdehyde (MDA) and myeloperoxidase (MPO) (p < 0.001), restored glutathione (GSH) levels and superoxide dismutase (SOD) (p < 0.001), decreased immunomarking for factor nuclear kappa B (NF-κB) and levels of interleukin (IL)-1 and tumor necrosis factor α (TNF-α), and maintained IL-10 and TGF-β basal levels. Furthermore, increased immunostaining for zonula occludens 1 (ZO-1) was observed. CONCLUSIONS (-)-fenchone has intestinal anti-inflammatory activity related to cytoprotection of the intestinal barrier, as well as antioxidant and immunomodulatory effects.
Collapse
Affiliation(s)
- Maria Elaine Cristina Araruna
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa CEP 58051-970, PB, Brazil; (M.E.C.A.); (E.B.A.J.); (C.A.d.L.S.); (M.M.B.P.); (M.L.d.S.P.); (V.P.A.); (M.V.S.); (M.S.d.S.); (A.F.A.)
| | - Edvaldo Balbino Alves Júnior
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa CEP 58051-970, PB, Brazil; (M.E.C.A.); (E.B.A.J.); (C.A.d.L.S.); (M.M.B.P.); (M.L.d.S.P.); (V.P.A.); (M.V.S.); (M.S.d.S.); (A.F.A.)
| | - Catarina Alves de Lima Serafim
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa CEP 58051-970, PB, Brazil; (M.E.C.A.); (E.B.A.J.); (C.A.d.L.S.); (M.M.B.P.); (M.L.d.S.P.); (V.P.A.); (M.V.S.); (M.S.d.S.); (A.F.A.)
| | - Matheus Marley Bezerra Pessoa
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa CEP 58051-970, PB, Brazil; (M.E.C.A.); (E.B.A.J.); (C.A.d.L.S.); (M.M.B.P.); (M.L.d.S.P.); (V.P.A.); (M.V.S.); (M.S.d.S.); (A.F.A.)
| | - Michelle Liz de Souza Pessôa
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa CEP 58051-970, PB, Brazil; (M.E.C.A.); (E.B.A.J.); (C.A.d.L.S.); (M.M.B.P.); (M.L.d.S.P.); (V.P.A.); (M.V.S.); (M.S.d.S.); (A.F.A.)
| | - Vitória Pereira Alves
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa CEP 58051-970, PB, Brazil; (M.E.C.A.); (E.B.A.J.); (C.A.d.L.S.); (M.M.B.P.); (M.L.d.S.P.); (V.P.A.); (M.V.S.); (M.S.d.S.); (A.F.A.)
| | - Marianna Vieira Sobral
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa CEP 58051-970, PB, Brazil; (M.E.C.A.); (E.B.A.J.); (C.A.d.L.S.); (M.M.B.P.); (M.L.d.S.P.); (V.P.A.); (M.V.S.); (M.S.d.S.); (A.F.A.)
| | - Marcelo Sobral da Silva
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa CEP 58051-970, PB, Brazil; (M.E.C.A.); (E.B.A.J.); (C.A.d.L.S.); (M.M.B.P.); (M.L.d.S.P.); (V.P.A.); (M.V.S.); (M.S.d.S.); (A.F.A.)
| | - Adriano Francisco Alves
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa CEP 58051-970, PB, Brazil; (M.E.C.A.); (E.B.A.J.); (C.A.d.L.S.); (M.M.B.P.); (M.L.d.S.P.); (V.P.A.); (M.V.S.); (M.S.d.S.); (A.F.A.)
- Department of Physiology and Pathology, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa CEP 58051-970, PB, Brazil;
| | - Maria Carolina de Paiva Sousa
- Department of Physiology and Pathology, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa CEP 58051-970, PB, Brazil;
| | - Aurigena Antunes Araújo
- Department of Morphology, Histology and Basic Pathology, Biosciences Center, Federal University of Rio Grande do Norte (UFRN), Natal CEP 59078-970, RN, Brazil;
| | - Leônia Maria Batista
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa CEP 58051-970, PB, Brazil; (M.E.C.A.); (E.B.A.J.); (C.A.d.L.S.); (M.M.B.P.); (M.L.d.S.P.); (V.P.A.); (M.V.S.); (M.S.d.S.); (A.F.A.)
| |
Collapse
|
25
|
Apte A, Dutta Dey P, Julakanti SR, Midura-Kiela M, Skopp SM, Canchis J, Fauser T, Bardill J, Seal S, Jackson DM, Ghishan FK, Kiela PR, Zgheib C, Liechty KW. Oral Delivery of miR146a Conjugated to Cerium Oxide Nanoparticles Improves an Established T Cell-Mediated Experimental Colitis in Mice. Pharmaceutics 2024; 16:1573. [PMID: 39771552 PMCID: PMC11679827 DOI: 10.3390/pharmaceutics16121573] [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: 11/05/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Dysregulated inflammation and oxidative stress are strongly implicated in the pathogenesis of inflammatory bowel disease. We have developed a novel therapeutic that targets inflammation and oxidative stress. It is comprised of microRNA-146a (miR146a)-loaded cerium oxide nanoparticles (CNPs) (CNP-miR146a). We hypothesized that oral delivery of CNP-miR146a would reduce colonic inflammation in a mouse model of established, chronic, T cell-mediated colitis. Methods: The stability of CNP-miR146a and mucosal delivery was assessed in vitro with simulated gastrointestinal fluid and in vivo after oral gavage by quantitative real-time RT-PCR. The efficacy of orally administered CNP-miR146a was tested in mice with established colitis using the model of adoptive naïve T-cell transfer in recombinant activating gene 2 knockout (Rag2-/-) mice. Measured outcomes included histopathology; CD45+ immune cell infiltration; oxidative DNA damage (tissue 8-hydroxy-2'-deoxyguanosine; 8-OHdG); expression of IL-6 and TNF mRNA and protein, and flow cytometry analysis of lamina propria Th1 and Th17 cell populations. Results: miR146a expression remained stable in simulated gastric and intestinal conditions. miR146a expression increased in the intestines of mice six hours following oral gavage of CNP-miR146a. Oral delivery of CNP-miR146a in mice with colitis was associated with reduced inflammation and oxidative stress in the proximal and distal colons as evidenced by histopathology scoring, reduced immune cell infiltration, reduced IL-6 and TNF expression, and decreased populations of CD4+Tbet+IFNg+ Th1, CD4+RorgT+IL17+ Th17, as well as pathogenic double positive IFNg+IL17+ T cells. Conclusions: CNP-miR146a represents a novel orally available therapeutic with high potential to advance into clinical trials.
Collapse
Affiliation(s)
- Anisha Apte
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
| | - Pujarini Dutta Dey
- Department of Pediatrics, Daniel Cracchiolo Institute for Pediatric Autoimmune Disease Research, Steele Children’s Research Center, University of Arizona Health Sciences Center, Tucson, AZ 85621, USA
| | - Srisaianirudh Reddy Julakanti
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
| | - Monica Midura-Kiela
- Department of Pediatrics, Daniel Cracchiolo Institute for Pediatric Autoimmune Disease Research, Steele Children’s Research Center, University of Arizona Health Sciences Center, Tucson, AZ 85621, USA
| | - Stacy M. Skopp
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
| | - Jimena Canchis
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
| | - Tobias Fauser
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
| | - James Bardill
- Laboratory for Fetal and Regenerative Biology, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO 80045, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Nanoscience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | | | - Fayez K. Ghishan
- Department of Pediatrics, Daniel Cracchiolo Institute for Pediatric Autoimmune Disease Research, Steele Children’s Research Center, University of Arizona Health Sciences Center, Tucson, AZ 85621, USA
| | - Pawel R. Kiela
- Department of Pediatrics, Daniel Cracchiolo Institute for Pediatric Autoimmune Disease Research, Steele Children’s Research Center, University of Arizona Health Sciences Center, Tucson, AZ 85621, USA
- Department of Immunobiology, University of Arizona Health Sciences Center, Tucson, AZ 85621, USA
| | - Carlos Zgheib
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
- Ceria Therapeutics, Inc., Tucson, AZ 85721, USA
| | - Kenneth W. Liechty
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
- Ceria Therapeutics, Inc., Tucson, AZ 85721, USA
| |
Collapse
|
26
|
Shin HY, Jeong WB, Joung MY, Shin KS, Yu KW. Effects of Centella asiatica-isolated pectic polysaccharide on dextran sulfate sodium-induced colitis. Int J Biol Macromol 2024; 285:138237. [PMID: 39622374 DOI: 10.1016/j.ijbiomac.2024.138237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/15/2024] [Accepted: 11/29/2024] [Indexed: 12/08/2024]
Abstract
Dietary supplementation of polysaccharides demonstrates strong therapeutic actions on inflammatory bowel disease (IBD). Centella asiatica (CA) has traditionally been used in Ayurveda and Chinese medicine. However, the effects of CA-isolated pectic polysaccharides on IBD remain unknown. This study determined the effect of a CA-isolated pectic polysaccharide on IBD. The crude polysaccharide (CA-CP), isolated from a hot-water extract of CA, is a typical pectic polysaccharide composed mainly of galacturonic acid (40.6 %), galactose (27.6 %), arabinose (13.5 %), and rhamnose (8.5 %). CA-CP improved clinical symptoms in a DSS-induced colitis murine model, including weight change (9.9-12.0 %), disease activity index (31.6-51.9 %), colon length (13.2-21.5 %), and spleen weight (21.8-26.3 %). CA-CP effectively regulated the levels of inflammatory and junctional factors by mediating the MAPK and NF-κB pathways. CA-CP partially alleviated DSS-induced crypt destruction, submucosal edema, inflammatory infiltration, and mucin secretion. The content of total short-chain fatty acids increased substantially (cecum 29.8-53.7 %, feces 75.4-109.3 %) with oral CA-CP administration compared to the DSS group. Cecal microbial community analysis revealed that CA-CP administration regulated DSS-induced colitis by reducing the abundance of Escherichia (5.0-10.9 %) and Clostridium (0.3-0.4 %), while increasing the abundance of Bacteroidetes (6.8-8.5 %), Ligilactobacillus (2.7-6.0 %), and Bilophilia (0.3-0.6 %). The findings provide fundamental data for developing novel functional therapeutic agents for the prevention and treatment of colitis, using CA-isolated pectic polysaccharides. Furthermore, to the best of our knowledge, our study is the first to demonstrate the effects of pectic polysaccharides isolated from CA on IBD.
Collapse
Affiliation(s)
- Hyun Young Shin
- Transdisciplinary Major in Learning Health Systems, Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea.
| | - Won Bi Jeong
- Major in Food & Nutrition, Korea National University of Transportation, Chungbuk 27909, Republic of Korea.
| | - Mi Yeun Joung
- Corporation ChamSunJin Green Juice, Jincheon 27865, Republic of Korea.
| | - Kwang-Soon Shin
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea.
| | - Kwang-Won Yu
- Major in Food & Nutrition, Korea National University of Transportation, Chungbuk 27909, Republic of Korea.
| |
Collapse
|
27
|
Wang X, Wang P, Li Y, Guo H, Wang R, Liu S, Qiu J, Wang X, Hao Y, Zhao Y, Liao H, Zou Z, Thinwa J, Liu R. Procyanidin C1 Modulates the Microbiome to Increase FOXO1 Signaling and Valeric Acid Levels to Protect the Mucosal Barrier in Inflammatory Bowel Disease. ENGINEERING 2024; 42:108-120. [DOI: 10.1016/j.eng.2023.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
28
|
Stolz V, de Freitas e Silva R, Rica R, Zhu C, Preglej T, Hamminger P, Hainberger D, Alteneder M, Müller L, Waldherr M, Waltenberger D, Hladik A, Agerer B, Schuster M, Frey T, Krausgruber T, Knapp S, Campbell C, Schmetterer K, Trauner M, Bergthaler A, Bock C, Boucheron N, Ellmeier W. Nuclear receptor corepressor 1 controls regulatory T cell subset differentiation and effector function. eLife 2024; 13:e78738. [PMID: 39466314 PMCID: PMC11517256 DOI: 10.7554/elife.78738] [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: 03/17/2022] [Accepted: 09/25/2024] [Indexed: 10/29/2024] Open
Abstract
FOXP3+ regulatory T cells (Treg cells) are key for immune homeostasis. Here, we reveal that nuclear receptor corepressor 1 (NCOR1) controls naïve and effector Treg cell states. Upon NCOR1 deletion in T cells, effector Treg cell frequencies were elevated in mice and in in vitro-generated human Treg cells. NCOR1-deficient Treg cells failed to protect mice from severe weight loss and intestinal inflammation associated with CD4+ T cell transfer colitis, indicating impaired suppressive function. NCOR1 controls the transcriptional integrity of Treg cells, since effector gene signatures were already upregulated in naïve NCOR1-deficient Treg cells while effector NCOR1-deficient Treg cells failed to repress genes associated with naïve Treg cells. Moreover, genes related to cholesterol homeostasis including targets of liver X receptor (LXR) were dysregulated in NCOR1-deficient Treg cells. However, genetic ablation of LXRβ in T cells did not revert the effects of NCOR1 deficiency, indicating that NCOR1 controls naïve and effector Treg cell subset composition independent from its ability to repress LXRβ-induced gene expression. Thus, our study reveals that NCOR1 maintains naïve and effector Treg cell states via regulating their transcriptional integrity. We also reveal a critical role for this epigenetic regulator in supporting the suppressive functions of Treg cells in vivo.
Collapse
Affiliation(s)
- Valentina Stolz
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Rafael de Freitas e Silva
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Ramona Rica
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Ci Zhu
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Teresa Preglej
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Patricia Hamminger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Daniela Hainberger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Marlis Alteneder
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Lena Müller
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Monika Waldherr
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Darina Waltenberger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Anastasiya Hladik
- Medical University of Vienna, Vienna, Department of Medicine I, Laboratory of Infection BiologyViennaAustria
| | - Benedikt Agerer
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Michael Schuster
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Tobias Frey
- Medical University of Vienna, Department of Laboratory MedicineViennaAustria
| | - Thomas Krausgruber
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Medical University of Vienna, Center for Medical Statistics, Informatics, and Intelligent Systems, Institute of Artificial IntelligenceViennaAustria
| | - Sylvia Knapp
- Medical University of Vienna, Vienna, Department of Medicine I, Laboratory of Infection BiologyViennaAustria
| | - Clarissa Campbell
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Klaus Schmetterer
- Medical University of Vienna, Department of Laboratory MedicineViennaAustria
| | - Michael Trauner
- Medical University of Vienna, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Hans Popper Laboratory of Molecular HepatologyViennaAustria
| | - Andreas Bergthaler
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Medical University of Vienna, Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied ImmunologyViennaAustria
| | - Christoph Bock
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Medical University of Vienna, Center for Medical Statistics, Informatics, and Intelligent Systems, Institute of Artificial IntelligenceViennaAustria
| | - Nicole Boucheron
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Wilfried Ellmeier
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| |
Collapse
|
29
|
Caruso R, Lo BC, Chen GY, Núñez G. Host-pathobiont interactions in Crohn's disease. Nat Rev Gastroenterol Hepatol 2024:10.1038/s41575-024-00997-y. [PMID: 39448837 DOI: 10.1038/s41575-024-00997-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
The mammalian intestine is colonized by trillions of microorganisms that are collectively referred to as the gut microbiota. The majority of symbionts have co-evolved with their host in a mutualistic relationship that benefits both. Under certain conditions, such as in Crohn's disease, a subtype of inflammatory bowel disease, some symbionts bloom to cause disease in genetically susceptible hosts. Although the identity and function of disease-causing microorganisms or pathobionts in Crohn's disease remain largely unknown, mounting evidence from animal models suggests that pathobionts triggering Crohn's disease-like colitis inhabit certain niches and penetrate the intestinal tissue to trigger inflammation. In this Review, we discuss the distinct niches occupied by intestinal symbionts and the evidence that pathobionts triggering Crohn's disease live in the mucus layer or near the intestinal epithelium. We also discuss how Crohn's disease-associated mutations in the host disrupt intestinal homeostasis by promoting the penetration and accumulation of pathobionts in the intestinal tissue. Finally, we discuss the potential role of microbiome-based interventions in precision therapeutic strategies for the treatment of Crohn's disease.
Collapse
Affiliation(s)
- Roberta Caruso
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Bernard C Lo
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Grace Y Chen
- Department of Internal Medicine and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gabriel Núñez
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
30
|
Boaru DL, Fraile-Martinez O, De Leon-Oliva D, Garcia-Montero C, De Castro-Martinez P, Miranda-Gonzalez A, Saez MA, Muñon-Zamarron L, Castillo-Ruiz E, Barrena-Blázquez S, Cañonez-Zafra R, Alvarez-Mon MÁ, Toledo-Lobo MV, Minaya-Bravo AM, Lopez-Gonzalez L, Diaz-Pedrero R, Saz JV, Albillos A, Alvarez-Mon M, Guijarro LG, Ortega MA. Harnessing the Anti-Inflammatory Properties of Polyphenols in the Treatment of Inflammatory Bowel Disease. Int J Biol Sci 2024; 20:5608-5672. [PMID: 39494333 PMCID: PMC11528451 DOI: 10.7150/ijbs.98107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 08/25/2024] [Indexed: 11/05/2024] Open
Abstract
Inflammatory bowel disease (IBD) encompasses a spectrum of chronic inflammatory conditions affecting the gastrointestinal tract, notably ulcerative colitis (UC) and Crohn's disease (CD). Both UC and CD result from the interplay between genetic and environmental factors that trigger an exacerbated immune response against gut microorganisms, leading to non-resolving inflammatory damage in the mucosa of specific zones in the intestine. Despite extensive research, current treatments often entail invasive interventions with considerable adverse effects on patient well-being. Consequently, there is a pressing need to find alternative and complementary therapeutic strategies aimed at ameliorating chronic inflammation and restoring intestinal barrier integrity. Polyphenols are plant-based compounds formed naturally or as semi-synthetic/synthetic derivatives with proven health-promoting effects and translational applications in a broad spectrum of chronic diseases. Preclinical models of IBD largely support the efficacy of a broad variety of polyphenols due to their well-documented antioxidant and modulatory properties on the immune system and gut microbiota. Likewise, a growing number of studies using distinct types of polyphenols are being conducted in humans, although more efforts are still warranted. In the present review, the main polyphenols investigated in vitro and in vivo models of IBD will be summarized, as well as the available trials or observational data accessible in humans. Finally, the role of polyphenols in the clinical context of IBDs, along with the main problematics regarding their translational issues and concerns will be discussed, including bioavailability, their inclusion in healthy dietary patterns and foods, interaction with other drugs, and other important points to be addressed by future research.
Collapse
Affiliation(s)
- Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Alejandro Miranda-Gonzalez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Miguel A Saez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Leticia Muñon-Zamarron
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Elisa Castillo-Ruiz
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Rafael Cañonez-Zafra
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
| | - Miguel Ángel Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Maria V Toledo-Lobo
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Biomedicine and Biotechnology, University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Ana M Minaya-Bravo
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias University Hospital, Alcalá de Henares, Spain
| | - Jose V Saz
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Biomedicine and Biotechnology, University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Agustin Albillos
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Gastroenterology and Hepatology Departament. Ramón y Cajal University Hospital, Madrid, Spain; Ramón y Cajal Institute for Health Research (IRYCIS), Madrid, Spain; Network Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain; University of Alcalá, Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Departament and Internal Medicine (CIBEREHD), Príncipe de Asturias University Hospital, Alcalá de Henares, Spain
| | - Luis G Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
| | - Miguel A Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| |
Collapse
|
31
|
Dong Y, Gai Z, Han M, Zhao Y. Lacticaseibacillus rhamnosus LRa05 mediates dynamic regulation of intestinal microbiota in mice with low-dose DSS-induced chronic mild inflammation. Front Microbiol 2024; 15:1483104. [PMID: 39444683 PMCID: PMC11496787 DOI: 10.3389/fmicb.2024.1483104] [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: 08/19/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024] Open
Abstract
Aim This study aimed to investigate the effects of low-dose dextran sulfate sodium (DSS) on the induction of chronic mild inflammation in mice and to evaluate the therapeutic potential of Lacticaseibacillus rhamnosus LRa05 (LRa05) to ameliorate the associated effects. The focus was on investigating changes in inflammatory, gut microbiota, serum lipopolysaccharide (LPS) and inflammatory cytokines. Methods Mice were exposed to a low-dose of DSS to induce chronic mild inflammation and LRa05 was administered as a probiotic intervention. The experiment included determination of body weight, colon length, histological examinations, and analysis of LPS and inflammatory cytokines in serum over 12 weeks. In addition, liver function, oxidative stress and intestinal microbiota were examined to understand the comprehensive effects of DSS and LRa05. Results Low-dose DSS did not lead to significant changes in body weight, colon length or histologic signs of inflammation. However, it led to a significant increase in serum levels of LPS, tumor necrosis factor-alpha (TNFα) and interleukin-6 (IL6). Intervention with LRa05 effectively attenuated these changes, particularly by lowering LPS levels and normalizing inflammatory cytokines. In addition, LRa05 protected against DSS-induced liver function damage and attenuated oxidative stress in the liver. Analysis of the gut microbiota demonstrated dynamic regulatory effects, where LRa05 intervention led to significant shifts in microbial populations, promoting a balanced microbiota profile. These changes are indicative of dynamic regulation by LRa05 in response to chronic mild inflammation, highlighting the probiotic's role in modulating the gut environment. Conclusion The LRa05 intervention showed multi-layered regulation in the chronic mild inflammation model by reducing inflammatory cytokines, maintaining liver function and restoring the balance of the gut microbiota. This provides experimental support for the potential use of LRa05 in chronic inflammation-related diseases and emphasizes the importance of probiotics for overall health. The study suggests that LRa05 is a potential therapeutic agent for the treatment of chronic inflammation associated with gut dysbiosis.
Collapse
Affiliation(s)
- Yao Dong
- Department of Research and Development, Wecare Probiotics Co., Ltd., Suzhou, China
| | - Zhonghui Gai
- Department of Research and Development, Wecare Probiotics Co., Ltd., Suzhou, China
| | - Mei Han
- Department of Food Quality and Safety, Shanghai Business School, Shanghai, China
| | - Yunjiao Zhao
- Department of Research and Development, Wecare Probiotics Co., Ltd., Suzhou, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
32
|
Tong G, Shen Y, Li H, Qian H, Tan Z. NLRC4, inflammation and colorectal cancer (Review). Int J Oncol 2024; 65:99. [PMID: 39239759 PMCID: PMC11387119 DOI: 10.3892/ijo.2024.5687] [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: 07/09/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024] Open
Abstract
Chronic inflammation is recognized as a major risk factor for cancer and is involved in every phase of the disease. Inflammasomes are central to the inflammatory response and play a crucial role in cancer development. The present review summarizes the role of Nod‑like receptor C4 (NLRC4) in inflammation and colorectal cancer (CRC). Reviews of the literature were conducted using Web of Science, PubMed and CNKI, with search terms including 'NLRC4', 'colorectal cancer', 'auto‑inflammatory diseases' and 'prognosis'. Variants of NLRC4 can cause recessive immune dysregulation and autoinflammation or lead to ulcerative colitis as a heterozygous risk factor. Additionally, genetic mutations in inflammasome components may increase susceptibility to cancer. NLRC4 is considered a tumor suppressor in CRC. The role of NLRC4 in CRC signaling pathways is currently understood to involve five key aspects (caspase 1, NLRP3/IL‑8, IL‑1β/IL‑1, NAIP and p53). The mechanisms by which NLRC4 is involved in CRC are considered to be threefold (through pyroptosis, apoptosis, necroptosis and PANoptosis; regulating the immune response; and protecting intestinal epithelial cells to prevent CRC). However, the impact of NLRC4 mutations on CRC remains unclear. In conclusion, NLRC4 is a significant inflammasome that protects against CRC through various signaling pathways and mechanisms. The association between NLRC4 mutations and CRC warrants further investigation.
Collapse
Affiliation(s)
- Guojun Tong
- Department of Colorectal Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China
- Central Laboratory, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China
| | - Yan Shen
- Department of General Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China
| | - Hui Li
- Department of General Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China
| | - Hai Qian
- Department of General Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China
| | - Zhenhua Tan
- Department of General Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China
| |
Collapse
|
33
|
Chun KS, Kim EH, Kim DH, Song NY, Kim W, Na HK, Surh YJ. Targeting cyclooxygenase-2 for chemoprevention of inflammation-associated intestinal carcinogenesis: An update. Biochem Pharmacol 2024; 228:116259. [PMID: 38705538 DOI: 10.1016/j.bcp.2024.116259] [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: 02/13/2024] [Revised: 04/18/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Mounting evidence from preclinical and clinical studies suggests that persistent inflammation functions as a driving force in the journey to cancer. Cyclooxygenase-2 (COX-2) is a key enzyme involved in inflammatory signaling. While being transiently upregulated upon inflammatory stimuli, COX-2 has been found to be consistently overexpressed in human colorectal cancer and several other malignancies. The association between chronic inflammation and cancer has been revisited: cancer can arise when inflammation fails to resolve. Besides its proinflammatory functions, COX-2 also catalyzes the production of pro-resolving as well as anti-inflammatory metabolites from polyunsaturated fatty acids. This may account for the side effects caused by long term use of some COX-2 inhibitory drugs during the cancer chemopreventive trials. This review summarizes the latest findings highlighting the dual functions of COX-2 in the context of its implications in the development, maintenance, and progression of cancer.
Collapse
Affiliation(s)
- Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Korea
| | - Eun-Hee Kim
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam 13488, South Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, Gyeonggi-do 16227, South Korea
| | - Na-Young Song
- Department of Oral Biology, BK21 Four Project, Yonsei University College of Dentistry, Seoul 03722, South Korea
| | - Wonki Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Hye-Kyung Na
- Department of Food Science and Biotechnology, College of Knowledge-Based Services Engineering, Sungshin Women's University, Seoul 01133, South Korea
| | - Young-Joon Surh
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
34
|
Zhang Y, Thomas JP, Korcsmaros T, Gul L. Integrating multi-omics to unravel host-microbiome interactions in inflammatory bowel disease. Cell Rep Med 2024; 5:101738. [PMID: 39293401 PMCID: PMC11525031 DOI: 10.1016/j.xcrm.2024.101738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/11/2024] [Accepted: 08/21/2024] [Indexed: 09/20/2024]
Abstract
The gut microbiome is crucial for nutrient metabolism, immune regulation, and intestinal homeostasis with changes in its composition linked to complex diseases like inflammatory bowel disease (IBD). Although the precise host-microbial mechanisms in disease pathogenesis remain unclear, high-throughput sequencing have opened new ways to unravel the role of interspecies interactions in IBD. Systems biology-a holistic computational framework for modeling complex biological systems-is critical for leveraging multi-omics datasets to identify disease mechanisms. This review highlights the significance of multi-omics data in IBD research and provides an overview of state-of-the-art systems biology resources and computational tools for data integration. We explore gaps, challenges, and future directions in the research field aiming to uncover novel biomarkers and therapeutic targets, ultimately advancing personalized treatment strategies. While focusing on IBD, the proposed approaches are applicable for other complex diseases, like cancer, and neurodegenerative diseases, where the microbiome has also been implicated.
Collapse
Affiliation(s)
- Yiran Zhang
- Department of Surgery & Cancer, Imperial College London, London W12 0NN, UK; Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| | - John P Thomas
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK; UKRI MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, London W12 0HS, UK
| | - Tamas Korcsmaros
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK; NIHR Imperial BRC Organoid Facility, Imperial College London, London W12 0NN, UK; Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK.
| | - Lejla Gul
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK; Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK
| |
Collapse
|
35
|
Sawaed J, Zelik L, Levin Y, Feeney R, Naama M, Gordon A, Zigdon M, Rubin E, Telpaz S, Modilevsky S, Ben-Simon S, Awad A, Harshuk-Shabso S, Nuriel-Ohayon M, Werbner M, Schroeder BO, Erez A, Bel S. Antibiotics damage the colonic mucus barrier in a microbiota-independent manner. SCIENCE ADVANCES 2024; 10:eadp4119. [PMID: 39259805 PMCID: PMC11389797 DOI: 10.1126/sciadv.adp4119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 08/05/2024] [Indexed: 09/13/2024]
Abstract
Antibiotic use is a risk factor for development of inflammatory bowel diseases (IBDs). IBDs are characterized by a damaged mucus layer, which does not separate the intestinal epithelium from the microbiota. Here, we hypothesized that antibiotics affect the integrity of the mucus barrier, which allows bacterial penetrance and predisposes to intestinal inflammation. We found that antibiotic treatment led to breakdown of the colonic mucus barrier and penetration of bacteria into the mucus layer. Using fecal microbiota transplant, RNA sequencing followed by machine learning, ex vivo mucus secretion measurements, and antibiotic treatment of germ-free mice, we determined that antibiotics induce endoplasmic reticulum stress in the colon that inhibits colonic mucus secretion in a microbiota-independent manner. This antibiotic-induced mucus secretion flaw led to penetration of bacteria into the colonic mucus layer, translocation of microbial antigens into circulation, and exacerbation of ulcerations in a mouse model of IBD. Thus, antibiotic use might predispose to intestinal inflammation by impeding mucus production.
Collapse
Affiliation(s)
- Jasmin Sawaed
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Lilach Zelik
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Yehonatan Levin
- Racah Institute of Physics, The Hebrew University, Jerusalem, Israel
| | - Rachel Feeney
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå, Sweden
- Umeå Center for Microbial Research (UCMR), Umeå, Sweden
| | - Maria Naama
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Ateret Gordon
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Mor Zigdon
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Elad Rubin
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Shahar Telpaz
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | | | - Shira Ben-Simon
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Aya Awad
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | | | | | - Michal Werbner
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Bjoern O Schroeder
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå, Sweden
- Umeå Center for Microbial Research (UCMR), Umeå, Sweden
| | - Amir Erez
- Racah Institute of Physics, The Hebrew University, Jerusalem, Israel
| | - Shai Bel
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
36
|
Mohd Tamsir N, Mohd Esa N, Shafie NH, Hamzah H. Manilkara zapota (L.) P. Royen Leaf Mitigates Colitis-Associated Colon Cancer through Anti-inflammatory Modulation in BALB/C Mice. Adv Pharmacol Pharm Sci 2024; 2024:1137696. [PMID: 39290583 PMCID: PMC11407886 DOI: 10.1155/2024/1137696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 08/02/2024] [Accepted: 08/24/2024] [Indexed: 09/19/2024] Open
Abstract
Colitis-associated colon cancer (CAC) arises from prolonged inflammation of the inner colon lining. An alternative approach to treating or preventing CAC involves the use of natural products such as Manilkara zapota (L.) P. Royen or M. zapota, which has been studied for its medicinal and pharmacological properties. Previous research has demonstrated the anticancer effects of M. zapota leaf aqueous extract (MZLAE) on colon cancer cells. However, no animal study has investigated the effects of MZLAE on CAC. Therefore, this study aimed to assess the potential anti-inflammatory effects of MZLAE on CAC in mice. In the present study, CAC was induced using azoxymethane (AOM) and dextran sodium sulphate (DSS). The mice were randomly assigned into five groups: (a) normal, (b) AOM/DSS, (c) AOM/DSS + 50 mg/kg MZLAE, (d) AOM/DSS + 100 mg/kg MZLAE, and (e) AOM/DSS + 200 mg/kg MZLAE. Various parameters including disease activity index (DAI), colon length and weight, reactive oxygen species (ROS), superoxide, superoxide dismutase (SOD), histopathological assessment, and proinflammatory cytokines expression were analysed. The results indicated that MZLAE improved DAI scores, colon length, colon histological dysplasia and inflammation scores, and SOD level, while also reducing ROS production and expression of proinflammatory cytokines (tumour necrosis factor-alpha (TNF- α) and interleukin 6 (IL-6)). In conclusion, this study suggests that MZLAE may serve as a promising source of antioxidants and anti-inflammatory agents for alleviating CAC.
Collapse
Affiliation(s)
- Norain Mohd Tamsir
- Department of Nutrition Faculty of Medicine and Health Sciences Universiti Putra Malaysia 43400, Serdang, Selangor, Malaysia
| | - Norhaizan Mohd Esa
- Department of Nutrition Faculty of Medicine and Health Sciences Universiti Putra Malaysia 43400, Serdang, Selangor, Malaysia
- Natural Medicine and Product Research Laboratory (NaturMeds) Institute of Bioscience Universiti Putra Malaysia 43400, Serdang, Selangor, Malaysia
| | - Nurul Husna Shafie
- Department of Nutrition Faculty of Medicine and Health Sciences Universiti Putra Malaysia 43400, Serdang, Selangor, Malaysia
- Laboratory of UPM-MAKNA Cancer Research Institute of Bioscience Universiti Putra Malaysia 43400, Serdang, Selangor, Malaysia
| | - Hazilawati Hamzah
- Department of Veterinary Pathology and Microbiology Faculty of Veterinary Medicine Universiti Putra Malaysia 43400, Serdang, Selangor, Malaysia
| |
Collapse
|
37
|
Uttarwar RG, Mekonnen SA, Van Beeck W, Wang A, Finnegan P, Roberts RF, Merenstein D, Slupsky CM, Marco ML. Effects of Bifidobacterium animalis subsp. lactis BB-12 and yogurt on mice during oral antibiotic administration. Microbiol Res 2024; 286:127794. [PMID: 38852301 DOI: 10.1016/j.micres.2024.127794] [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: 02/16/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/11/2024]
Abstract
Probiotics have the potential to prevent disruptions to normal gastrointestinal function caused by oral antibiotic use. In this study, we examined the capacity of Bifidobacterium animalis subspecies lactis BB-12 (BB-12) and yogurt, separately and combined, to mitigate the effects of the antibiotic amoxicillin-clavulanate (AMC) on the gut microbiota and metabolomes of C57BL/6 J mice. Male and female mice were administered either BB-12, yogurt, BB-12 in yogurt, or saline for 10 days concurrent with the inclusion of AMC in the drinking water. Male mice exposed to AMC exhibited significant reductions (p<0.05) in body weight over the course of the study compared to sham (no AMC) controls whereas no such effects were observed for female mice. AMC administration resulted in rapid alterations to the intestinal microbiota in both sexes irrespective of BB-12 or yogurt treatment, including significant (p<0.05) losses in bacterial cell numbers and changes in microbial alpha-diversity and beta-diversity in the feces and cecal contents. The effects of AMC on the gut microbiota were observed within one day of administration and the bacterial contents continued to change over time, showing a succession marked by rapid reductions in Muribaculaceae and Lachnospiraceae and temporal increases in proportions of Acholeplasmataceae (day 1) and Streptococcaceae and Leuconostocaceae (day 5). By day 10 of AMC intake, high proportions of Gammaproteobacteria assigned as Erwiniaceae or Enterobacteriaceae (average of 63 %), were contained in the stools and were similarly enriched in the cecum. The cecal contents of mice given AMC harbored significantly reduced concentrations of (branched) short-chain fatty acids (SCFA), aspartate, and other compounds, whereas numerous metabolites, including formate, lactate, and several amino acids and amino acid derivatives were significantly enriched. Despite the extensive impact of AMC, starting at day 7 of the study, the body weights of male mice given yogurt or BB-12 (in saline) with AMC were similar to the healthy controls. BB-12 (in saline) and yogurt intake was associated with increased Streptococcaceae and both yogurt and BB-12 resulted in lower proportions of Erwiniaceae in the fecal and cecal contents. The cecal contents of mice fed BB-12 in yogurt contained levels of formate, glycine, and glutamine that were equivalent to the sham controls. These findings highlight the potential of BB-12 and yogurt to mitigate antibiotic-induced gut dysbiosis.
Collapse
Affiliation(s)
- Ruchita G Uttarwar
- Department of Food Science & Technology, University of California, Davis, USA
| | - Solomon A Mekonnen
- Department of Food Science & Technology, University of California, Davis, USA
| | - Wannes Van Beeck
- Department of Food Science & Technology, University of California, Davis, USA
| | - Aidong Wang
- Department of Food Science & Technology, University of California, Davis, USA; Department of Nutrition, University of California, Davis, CA, USA
| | - Peter Finnegan
- Department of Food Science & Technology, University of California, Davis, USA
| | | | - Daniel Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Carolyn M Slupsky
- Department of Food Science & Technology, University of California, Davis, USA; Department of Nutrition, University of California, Davis, CA, USA
| | - Maria L Marco
- Department of Food Science & Technology, University of California, Davis, USA.
| |
Collapse
|
38
|
Özcan Ö, Akyol Ö, Akyol A. Amine Oxidase, Copper Containing 3 ( Aoc3) Knockout Mice Are More Prone to DSS-induced Colitis and Colonic Tumorigenesis. In Vivo 2024; 38:2300-2309. [PMID: 39187313 PMCID: PMC11363779 DOI: 10.21873/invivo.13695] [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: 04/21/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND/AIM Inflammatory bowel diseases and colorectal cancer are a major cause of morbidity and mortality. Amine oxidase, copper-containing 3 (AOC3) is a critical enzyme in the physiological trafficking of leukocytes and the regulation of inflammation. This study aimed to examine the effects of Aoc3 deficiency in mice models of colitis and colorectal tumorigenesis. MATERIALS AND METHODS C57BL/6 and Aoc3 knockout mice were used for Dextran Sodium Sulfate (DSS) induced acute colitis and the Azoxymethane (AOM)/DSS model of inflammation-related colon cancer. We also evaluated the effect of Aoc3 in an Apc mutant mice model of intestinal and colonic tumorigenesis. RESULTS We observed that Aoc3 deficient mice were more prone to colitis induced by DSS in early phases and their survival was shorter. We also showed that Aoc3 deficient mice developed more tumors both in AOM/DSS and Apc mutant mice models. Furthermore, colonic tumors in the AOM/DSS groups in Aoc3 mutant mice were generally invasive type adenocarcinomas. CONCLUSION Aoc3 deficiency promotes colitis and colonic tumorigenesis in mouse models.
Collapse
Affiliation(s)
- Özge Özcan
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Özge Akyol
- Department of Pathology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Aytekin Akyol
- Department of Pathology, Hacettepe University Faculty of Medicine, Ankara, Turkey;
- Hacettepe University Transgenic Animal Technologies Research and Application Center, Ankara, Turkey
- Molecular Pathology Application and Research Center, Hacettepe University, Ankara, Turkey
| |
Collapse
|
39
|
Park N, Kim KS, Park CG, Jung HD, Park W, Na K. Adipose-derived stem cell-based anti-inflammatory paracrine factor regulation for the treatment of inflammatory bowel disease. J Control Release 2024; 374:384-399. [PMID: 39173953 DOI: 10.1016/j.jconrel.2024.08.027] [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: 05/24/2024] [Revised: 07/17/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Stem cell-based therapies offer promising avenues for treating inflammatory diseases owing to their immunomodulatory properties. However, challenges persist regarding their survival and efficacy in inflamed tissues. Our study introduces a novel approach by engineering adipose-derived stem cells (ADSCs) to enhance their viability in inflammatory environments and boost the secretion of paracrine factors for treating inflammatory bowel disease (IBD). An arginine-glycine-aspartate peptide-poly (ethylene glycol)-chlorin e6 conjugate (RPC) was synthesized and coupled with ADSCs, resulting in RPC-labeled ADSCs (ARPC). This conjugation strategy employed RGD-integrin interaction to shield stem cells and allowed visualization and tracking using chlorin e6. The engineered ARPC demonstrated enhanced viability and secretion of paracrine factors upon light irradiation, regulating the inflammatory microenvironment. RNA-sequencing analysis unveiled pathways favoring angiogenesis, DNA repair, and exosome secretion in ARPC(+) while downregulating inflammatory pathways. In in vivo models of acute and chronic IBD, ARPC(+) treatment led to reduced inflammation, preserved colon structure, and increased populations of regulatory T cells, highlighting its therapeutic potential. ARPC(+) selectively homed to inflammatory sites, demonstrating its targeted effect. Overall, ARPC(+) exhibits promise as an effective and safe therapeutic strategy for managing inflammatory diseases like IBD by modulating immune responses and creating an anti-inflammatory microenvironment.
Collapse
Affiliation(s)
- Naeun Park
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Kyoung Sub Kim
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Seobu-ro 2066, Suwon, Gyeonggi 16419, Republic of Korea; Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Seobu-ro 2066, Suwon, Gyeonggi 16419, Republic of Korea
| | - Hyun-Do Jung
- Division of Materials Science and Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi 16419, Republic of Korea
| | - Kun Na
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea.
| |
Collapse
|
40
|
Tanwar H, Gnanasekaran JM, Allison D, Chuang LS, He X, Aimetti M, Baima G, Costalonga M, Cross RK, Sears C, Mehandru S, Cho J, Colombel JF, Raufman JP, Thumbigere-Math V. Unravelling the Oral-Gut Axis: Interconnection Between Periodontitis and Inflammatory Bowel Disease, Current Challenges, and Future Perspective. J Crohns Colitis 2024; 18:1319-1341. [PMID: 38417137 PMCID: PMC11324343 DOI: 10.1093/ecco-jcc/jjae028] [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: 08/21/2023] [Revised: 12/04/2023] [Accepted: 02/27/2024] [Indexed: 03/01/2024]
Abstract
As the opposite ends of the orodigestive tract, the oral cavity and the intestine share anatomical, microbial, and immunological ties that have bidirectional health implications. A growing body of evidence suggests an interconnection between oral pathologies and inflammatory bowel disease [IBD], implying a shift from the traditional concept of independent diseases to a complex, reciprocal cycle. This review outlines the evidence supporting an 'oral-gut' axis, marked by a higher prevalence of periodontitis and other oral conditions in IBD patients and vice versa. We present an in-depth examination of the interconnection between oral pathologies and IBD, highlighting the shared microbiological and immunological pathways, and proposing a 'multi-hit' hypothesis in the pathogenesis of periodontitis-mediated intestinal inflammation. Furthermore, the review underscores the critical need for a collaborative approach between dentists and gastroenterologists to provide holistic oral-systemic healthcare.
Collapse
Affiliation(s)
- Himanshi Tanwar
- Division of Periodontology, University of Maryland School of Dentistry, Baltimore, MD, USA
| | | | - Devon Allison
- Division of Periodontology, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Ling-shiang Chuang
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xuesong He
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Mario Aimetti
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - Giacomo Baima
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - Massimo Costalonga
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Raymond K Cross
- Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cynthia Sears
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Saurabh Mehandru
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Judy Cho
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean-Pierre Raufman
- Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vivek Thumbigere-Math
- Division of Periodontology, University of Maryland School of Dentistry, Baltimore, MD, USA
- National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD, USA
| |
Collapse
|
41
|
Jyotsna, Sarkar B, Yadav M, Deka A, Markandey M, Sanyal P, Nagarajan P, Gaikward N, Ahuja V, Mohanty D, Basak S, Gokhale RS. A hepatocyte-specific transcriptional program driven by Rela and Stat3 exacerbates experimental colitis in mice by modulating bile synthesis. eLife 2024; 12:RP93273. [PMID: 39137024 PMCID: PMC11321761 DOI: 10.7554/elife.93273] [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] [Indexed: 08/15/2024] Open
Abstract
Hepatic factors secreted by the liver promote homeostasis and are pivotal for maintaining the liver-gut axis. Bile acid metabolism is one such example wherein, bile acid synthesis occurs in the liver and its biotransformation happens in the intestine. Dysfunctional interactions between the liver and the intestine stimulate varied pathological outcomes through its bidirectional portal communication. Indeed, aberrant bile acid metabolism has been reported in inflammatory bowel disease (IBD). However, the molecular mechanisms underlying these crosstalks that perpetuate intestinal permeability and inflammation remain obscure. Here, we identify a novel hepatic gene program regulated by Rela and Stat3 that accentuates the inflammation in an acute experimental colitis model. Hepatocyte-specific ablation of Rela and Stat3 reduces the levels of primary bile acids in both the liver and the gut and shows a restricted colitogenic phenotype. On supplementation of chenodeoxycholic acid (CDCA), knock-out mice exhibit enhanced colitis-induced alterations. This study provides persuasive evidence for the development of multi-organ strategies for treating IBD and identifies a hepatocyte-specific Rela-Stat3 network as a promising therapeutic target.
Collapse
Affiliation(s)
- Jyotsna
- Immunometabolism Laboratory, National Institute of ImmunologyNew DelhiIndia
| | - Binayak Sarkar
- Immunometabolism Laboratory, National Institute of ImmunologyNew DelhiIndia
| | - Mohit Yadav
- Immunometabolism Laboratory, National Institute of ImmunologyNew DelhiIndia
| | - Alvina Deka
- System Immunology Laboratory, National Institute of ImmunologyNew DelhiIndia
| | - Manasvini Markandey
- Department of GastroEnterology, All India Institute of Medical SciencesNew DelhiIndia
| | | | - Perumal Nagarajan
- Immunometabolism Laboratory, National Institute of ImmunologyNew DelhiIndia
| | | | - Vineet Ahuja
- Department of GastroEnterology, All India Institute of Medical SciencesNew DelhiIndia
| | - Debasisa Mohanty
- Immunometabolism Laboratory, National Institute of ImmunologyNew DelhiIndia
| | - Soumen Basak
- System Immunology Laboratory, National Institute of ImmunologyNew DelhiIndia
| | - Rajesh S Gokhale
- Immunometabolism Laboratory, National Institute of ImmunologyNew DelhiIndia
- Department of Biology, Indian Institute of Science Education and ResearchPashanIndia
| |
Collapse
|
42
|
Miller PF. Targeting microbial pathogenic mechanisms as a novel therapeutic strategy in IBD. Mol Med 2024; 30:122. [PMID: 39135000 PMCID: PMC11321147 DOI: 10.1186/s10020-024-00840-9] [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: 12/14/2023] [Accepted: 05/19/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Current therapy for patients suffering from inflammatory bowel diseases (IBD) is focused on inflammatory mechanisms exclusively and not the dysbiotic microbiota, despite growing evidence implicating a role for intestinal microbes in disease. MAIN BODY Ongoing research into the intestinal microbiota of IBD patients, using new technologies and/or deeper application of existing ones, has identified a number of microorganisms whose properties and behaviors warrant consideration as causative factors in disease. Such studies have implicated both bacteria and fungi in the pathogenesis of disease. Some of these organisms manifest mechanisms that should be amenable to therapeutic intervention via either conventional or novel drug discovery platforms. Of particular note is a deeper characterization of microbial derived proteases and their destructive potential. CONCLUSION Given the steady progress on the mechanistic role of the microbiota in inflammatory diseases, it is reasonable to anticipate a future in which therapeutics targeting microbial derived pathogenic factors play an important role in improving the lives of IBD patients.
Collapse
Affiliation(s)
- Paul F Miller
- Lighthouse Biopharma Consulting, LLC, 39 Emerald Glen Lane, Salem, CT, 06420, USA.
| |
Collapse
|
43
|
Young KA, Wojdyla K, Lai T, Mulholland KE, Aldaz Casanova S, Antrobus R, Andrews SR, Biggins L, Mahler-Araujo B, Barton PR, Anderson KR, Fearnley GW, Sharpe HJ. The receptor protein tyrosine phosphatase PTPRK promotes intestinal repair and catalysis-independent tumour suppression. J Cell Sci 2024; 137:jcs261914. [PMID: 38904097 PMCID: PMC11298714 DOI: 10.1242/jcs.261914] [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: 12/18/2023] [Accepted: 05/28/2024] [Indexed: 06/22/2024] Open
Abstract
PTPRK is a receptor tyrosine phosphatase that is linked to the regulation of growth factor signalling and tumour suppression. It is stabilized at the plasma membrane by trans homophilic interactions upon cell-cell contact. PTPRK regulates cell-cell adhesion but is also reported to regulate numerous cancer-associated signalling pathways. However, the signalling mechanism of PTPRK remains to be determined. Here, we find that PTPRK regulates cell adhesion signalling, suppresses invasion and promotes collective, directed migration in colorectal cancer cells. In vivo, PTPRK supports recovery from inflammation-induced colitis. In addition, we confirm that PTPRK functions as a tumour suppressor in the mouse colon and in colorectal cancer xenografts. PTPRK regulates growth factor and adhesion signalling, and suppresses epithelial to mesenchymal transition (EMT). Contrary to the prevailing notion that PTPRK directly dephosphorylates EGFR, we find that PTPRK regulation of both EGFR and EMT is independent of its catalytic function. This suggests that additional adaptor and scaffold functions are important features of PTPRK signalling.
Collapse
Affiliation(s)
| | | | - Tiffany Lai
- Signalling programme, Babraham Institute, Cambridge CB22 3AT, UK
| | | | | | - Robin Antrobus
- Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, UK
| | | | - Laura Biggins
- Bioinformatics, Babraham Institute, Cambridge CB22 3AT, UK
| | | | - Philippa R. Barton
- Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, UK
| | - Keith R. Anderson
- Molecular biology department, Genentech, South San Francisco, CA 94080, USA
| | | | - Hayley J. Sharpe
- Signalling programme, Babraham Institute, Cambridge CB22 3AT, UK
| |
Collapse
|
44
|
McKelvey M, Uddin MB, Palani S, Shao S, Sun K. IL-10 Counteracts IFN-γ to Alleviate Acute Lung Injury in a Viral-Bacterial Superinfection Model. Am J Respir Cell Mol Biol 2024; 71:110-120. [PMID: 38574279 PMCID: PMC11225870 DOI: 10.1165/rcmb.2023-0437oc] [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: 12/10/2023] [Accepted: 04/03/2024] [Indexed: 04/06/2024] Open
Abstract
Immune activation is essential for lung control of viral and bacterial infection, but an overwhelming inflammatory response often leads to the onset of acute respiratory distress syndrome. IL-10 plays a crucial role in regulating the balance between antimicrobial immunity and immunopathology. In the present study, we investigated the role of IL-10 in acute lung injury induced by influenza A virus and methicillin-resistant Staphylococcus aureus coinfection. This unique coinfection model resembles patients with acute pneumonia undergoing appropriate antibiotic therapies. Using global IL-10 and IL-10 receptor gene-deficient mice, as well as in vivo neutralizing antibodies, we show that IL-10 deficiency promotes IFN-γ-dominant cytokine responses and triggers acute animal death. Interestingly, this extreme susceptibility is fully preventable by IFN-γ neutralization during coinfection. Further studies using mice with Il10ra deletion in selective myeloid subsets reveal that IL-10 primarily acts on mononuclear phagocytes to prevent IFN-γ/TNF-α hyperproduction and acute mortality. Importantly, this antiinflammatory IL-10 signaling is independent of its inhibitory effect on antiviral and antibacterial defense. Collectively, our results demonstrate a key mechanism of IL-10 in preventing hypercytokinemia and acute respiratory distress syndrome pathogenesis by counteracting the IFN-γ response.
Collapse
Affiliation(s)
| | - Md Bashir Uddin
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Sunil Palani
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Shengjun Shao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Keer Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
45
|
Sun Q, Li H, Lv J, Shi W, Bai Y, Pan K, Chen A. Dopamine β-hydroxylase shapes intestinal inflammation through modulating T cell activation. Cell Immunol 2024; 401-402:104839. [PMID: 38850753 DOI: 10.1016/j.cellimm.2024.104839] [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: 03/19/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic and relapsing disease characterized by immune-mediated dysfunction of intestinal homeostasis. Alteration of the enteric nervous system and the subsequent neuro-immune interaction are thought to contribute to the initiation and progression of IBD. However, the role of dopamine beta-hydroxylase (DBH), an enzyme converting dopamine into norepinephrine, in modulating intestinal inflammation is not well defined. METHODS CD4+CD45RBhighT cell adoptive transfer, and 2,4-dinitrobenzene sulfonic acid (DNBS) or dextran sodium sulfate (DSS)-induced colitis were collectively conducted to uncover the effects of DBH inhibition by nepicastat, a DBH inhibitor, in mucosal ulceration, disease severity, and T cell function. RESULTS Inhibition of DBH by nepicastat triggered therapeutic effects on T cell adoptive transfer induced chronic mouse colitis model, which was consistent with the gene expression of DBH in multiple cell populations including T cells. Furthermore, DBH inhibition dramatically ameliorated the disease activity and colon shortening in chemically induced acute and chronic IBD models, as evidenced by morphological and histological examinations. The reshaped systemic inflammatory status was largely associated with decreased pro-inflammatory mediators, such as TNF-α, IL-6 and IFN-γ in plasma and re-balanced Th1, Th17 and Tregs in mesenteric lymph nodes (MLNs) upon colitis progression. Additionally, the conversion from dopamine (DA) to norepinephrine (NE) was inhibited resulting in increase in DA level and decrease in NE level and DA/NE showed immune-modulatory effects on the activation of immune cells. CONCLUSION Modulation of neurotransmitter levels via inhibition of DBH exerted protective effects on progression of murine colitis by modulating the neuro-immune axis. These findings suggested a promising new therapeutic strategy for attenuating intestinal inflammation.
Collapse
Affiliation(s)
- Qiaoling Sun
- Asieris Pharmaceuticals Co., Ltd, Shanghai, China.
| | - Heng Li
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Jing Lv
- Asieris Pharmaceuticals Co., Ltd, Shanghai, China
| | - Weilin Shi
- Asieris Pharmaceuticals Co., Ltd, Shanghai, China
| | - Yanfeng Bai
- Asieris Pharmaceuticals Co., Ltd, Shanghai, China
| | - Ke Pan
- Asieris Pharmaceuticals Co., Ltd, Shanghai, China
| | - Alice Chen
- Asieris Pharmaceuticals Co., Ltd, Palo Alto, CA, USA.
| |
Collapse
|
46
|
Reardon C. Comment on "No consistent evidence for the anti-inflammatory effect of vagus nerve stimulation in humans: A systematic review and meta-analysis". Brain Behav Immun 2024; 119:1018. [PMID: 38447885 DOI: 10.1016/j.bbi.2024.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/18/2024] [Accepted: 03/03/2024] [Indexed: 03/08/2024] Open
Affiliation(s)
- Colin Reardon
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, UC Davis, Davis, CA, United States
| |
Collapse
|
47
|
Madan U, Verma B, Awasthi A. Cenicriviroc, a CCR2/CCR5 antagonist, promotes the generation of type 1 regulatory T cells. Eur J Immunol 2024; 54:e2350847. [PMID: 38643381 DOI: 10.1002/eji.202350847] [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: 10/20/2023] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/22/2024]
Abstract
Cenicriviroc, a dual CCR2/CCR5 antagonist, initially developed as an anti-HIV drug, has shown promising results in nonalcoholic steatohepatitis phase 2 clinical trials. It inhibits the infiltration and activation of CCR2+/CCR5+ monocytes and macrophages to the site of liver injury, preventing liver fibrosis. However, the role of Cenicriviroc in the modulation of helper T cell differentiation and functions remains to be explored. In inflamed colons of Crohn's disease patients, CCR2+ and CCR5+ CD4+ T cells are enriched. Considering the role of CCR2+ and CCR5+ T cells in IBD pathogenesis, we investigated the potential role of Cenicriviroc in colitis. Our in vitro studies revealed that Cenicriviroc inhibits Th1-, Th2-, and Th17-cell differentiation while promoting the generation of type 1 regulatory T cells (Tr1), known for preventing inflammation through induction of IL-10. This study is the first to report that Cenicriviroc promotes Tr1 cell generation by up-regulating the signature of Tr1 cell transcription factors such as c-Maf, Prdm1, Irf-1, Batf, and EGR-2. Cenicriviroc displayed a protective effect in experimental colitis models by preventing body weight loss and intestinal inflammation and preserving epithelial barrier integrity. We show that Cenicriviroc induced IL-10 and inhibited the generation of pro-inflammatory cytokines IFN-γ, IL-17, IL-6, and IL-1β during colitis. Based on our data, we propose Cenicriviroc as a potential therapeutic in controlling tissue inflammation by inhibiting the generation and functions of effector T cells and promoting the induction of anti-inflammatory Tr1 cells.
Collapse
Affiliation(s)
- Upasna Madan
- Centre for Immuno-biology and Immunotherapy, NCR-Biotech Science Cluster, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Bhawna Verma
- Centre for Immuno-biology and Immunotherapy, NCR-Biotech Science Cluster, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Amit Awasthi
- Centre for Immuno-biology and Immunotherapy, NCR-Biotech Science Cluster, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, NCR Biotech Science Cluster, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
48
|
Qiu X, Luo W, Li H, Li T, Huang Y, Huang Q, Zhou R. A Traditional Chinese Medicine, Zhenqi Granule, Potentially Alleviates Dextran Sulfate Sodium-Induced Mouse Colitis Symptoms. BIOLOGY 2024; 13:427. [PMID: 38927307 PMCID: PMC11200386 DOI: 10.3390/biology13060427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024]
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease that causes chronic inflammation in the large intestine. The etiology of UC is complex and incompletely understood, with potential contributing factors including genetic susceptibility, environmental influences, immune dysregulation, and gut barrier dysfunction. Despite available therapeutic drugs, the suboptimal cure rate for UC emphasizes the necessity of developing novel therapeutics. Traditional Chinese Medicine (TCM) has attracted great interest in the treatment of such chronic inflammatory diseases due to its advantages, such as multi-targets and low side effects. In this study, a mouse model of Dextran Sulfate Sodium (DSS)-induced acute colitis was established and the efficacy of Zhenqi Granule, a TCM preparation composed of the extractives from Astragali Radix and Fructus Ligustri Lucidi, was evaluated. The results showed that treatment with Zhenqi Granule prior to or post-DSS induction could alleviate the symptoms of colitis, including weight loss, diarrhea, hematochezia, colon length shortening, and pathological damage of colon tissues of the DSS-treated mice. Further, network pharmacology analysis showed that there were 98 common targets between the active components of Zhenqi Granule and the targets of UC, and the common targets were involved in the regulation of inflammatory signaling pathways. Our results showed that Zhenqi Granule had preventive and therapeutic effects on acute colitis in mice, and the mechanism may be that the active components of Zhenqi Granule participated in the regulation of inflammatory response. This study provided data reference for further exploring the mechanism of Zhenqi Granule and also provided potential treatment strategies for UC.
Collapse
Affiliation(s)
- Xiuxiu Qiu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (X.Q.); (W.L.); (H.L.); (T.L.); (Y.H.)
| | - Wentao Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (X.Q.); (W.L.); (H.L.); (T.L.); (Y.H.)
| | - Haotian Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (X.Q.); (W.L.); (H.L.); (T.L.); (Y.H.)
| | - Tingting Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (X.Q.); (W.L.); (H.L.); (T.L.); (Y.H.)
| | - Yaxue Huang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (X.Q.); (W.L.); (H.L.); (T.L.); (Y.H.)
| | - Qi Huang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (X.Q.); (W.L.); (H.L.); (T.L.); (Y.H.)
- International Research Center for Animal Disease, Ministry of Science & Technology of China, Wuhan 430070, China
- The Cooperative Innovation Center of Sustainable Pig Production, Wuhan 430070, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (X.Q.); (W.L.); (H.L.); (T.L.); (Y.H.)
- International Research Center for Animal Disease, Ministry of Science & Technology of China, Wuhan 430070, China
- The Cooperative Innovation Center of Sustainable Pig Production, Wuhan 430070, China
| |
Collapse
|
49
|
Miljkovic R, Marinkovic E, Lukic I, Kovacevic A, Lopandic Z, Popovic M, Gavrovic-Jankulovic M, Schabussova I, Inic-Kanada A, Stojanovic M. Banana Lectin: A Novel Immunomodulatory Strategy for Mitigating Inflammatory Bowel Disease. Nutrients 2024; 16:1705. [PMID: 38892639 PMCID: PMC11175119 DOI: 10.3390/nu16111705] [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: 04/28/2024] [Revised: 05/25/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Compared to the general population, patients with inflammatory bowel disease (IBD) are less likely to be vaccinated, putting them at an increased risk of vaccine-preventable illnesses. This risk is further compounded by the immunosuppressive therapies commonly used in IBD management. Therefore, developing new treatments for IBD that maintain immune function is crucial, as successful management can lead to better vaccination outcomes and overall health for these patients. Here, we investigate the potential of recombinant banana lectin (rBanLec) as a supporting therapeutic measure to improve IBD control and possibly increase vaccination rates among IBD patients. By examining the therapeutic efficacy of rBanLec in a murine model of experimental colitis, we aim to lay the foundation for its application in improving vaccination outcomes. After inducing experimental colitis in C57BL/6 and BALB/c mice with 2,4,6-trinitrobenzene sulfonic acid, we treated animals orally with varying doses of rBanLec 0.1-10 µg/mL (0.01-1 µg/dose) during the course of the disease. We assessed the severity of colitis and rBanLec's modulation of the immune response compared to control groups. rBanLec administration resulted in an inverse dose-response reduction in colitis severity (less pronounced weight loss, less shortening of the colon) and an improved recovery profile, highlighting its therapeutic potential. Notably, rBanLec-treated mice exhibited significant modulation of the immune response, favoring anti-inflammatory pathways (primarily reduction in a local [TNFα]/[IL-10]) crucial for effective vaccination. Our findings suggest that rBanLec could mitigate the adverse effects of immunosuppressive therapy on vaccine responsiveness in IBD patients. By improving the underlying immune response, rBanLec may increase the efficacy of vaccinations, offering a dual benefit of disease management and prevention of vaccine-preventable illnesses. Further studies are required to translate these findings into clinical practice.
Collapse
Affiliation(s)
- Radmila Miljkovic
- Department of Research and Development, Institute of Immunology, Virology, Vaccines and Sera—Torlak, 11152 Belgrade, Serbia; (R.M.); (E.M.); (I.L.); (A.K.)
| | - Emilija Marinkovic
- Department of Research and Development, Institute of Immunology, Virology, Vaccines and Sera—Torlak, 11152 Belgrade, Serbia; (R.M.); (E.M.); (I.L.); (A.K.)
| | - Ivana Lukic
- Department of Research and Development, Institute of Immunology, Virology, Vaccines and Sera—Torlak, 11152 Belgrade, Serbia; (R.M.); (E.M.); (I.L.); (A.K.)
| | - Ana Kovacevic
- Department of Research and Development, Institute of Immunology, Virology, Vaccines and Sera—Torlak, 11152 Belgrade, Serbia; (R.M.); (E.M.); (I.L.); (A.K.)
| | - Zorana Lopandic
- Institute for Chemistry in Medicine, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Mina Popovic
- Faculty of Ecology and Environmental Protection, University Union—Nikola Tesla, 11158 Belgrade, Serbia;
| | | | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Aleksandra Inic-Kanada
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Marijana Stojanovic
- Department of Molecular Biology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, 11108 Belgrade, Serbia
| |
Collapse
|
50
|
Apte A, Bardill JR, Canchis J, Skopp SM, Fauser T, Lyttle B, Vaughn AE, Seal S, Jackson DM, Liechty KW, Zgheib C. Targeting Inflammation and Oxidative Stress to Improve Outcomes in a TNBS Murine Crohn's Colitis Model. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:894. [PMID: 38786849 PMCID: PMC11124096 DOI: 10.3390/nano14100894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
Inflammation and oxidative stress are implicated in the pathogenesis of Crohn's disease. Cerium oxide nanoparticle (CNP) conjugated to microRNA 146a (miR146a) (CNP-miR146a) is a novel compound with anti-inflammatory and antioxidative properties. We hypothesized that local administration of CNP-miR146a would improve colitis in a 2,4,6-Trinitrobenzenesulfonic acid (TNBS) mouse model for Crohn's disease by decreasing colonic inflammation. Balb/c mice were instilled with TNBS enemas to induce colitis. Two days later, the mice received cellulose gel enema, cellulose gel with CNP-miR146a enema, or no treatment. Control mice received initial enemas of 50% ethanol and PBS enemas on day two. The mice were monitored daily for weight loss and clinical disease activity. The mice were euthanized on days two or five to evaluate their miR146a expression, inflammation on histology, and colonic IL-6 and TNF gene expressions and protein concentrations. CNP-miR146a enema successfully increased colonic miR146a expression at 12 h following delivery. At the end of five days from TNBS instillation, the mice treated with CNP-miR146a demonstrated reduced weight loss, improved inflammation scores on histology, and reduced gene expressions and protein concentrations of IL-6 and TNF. The local delivery of CNP-miR146a in a TNBS mouse model of acute Crohn's colitis dramatically decreased inflammatory signaling, resulting in improved clinical disease.
Collapse
Affiliation(s)
- Anisha Apte
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, Tucson, AZ 85721, USA (K.W.L.)
| | - James R. Bardill
- Department of Surgery, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA; (J.R.B.)
| | - Jimena Canchis
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, Tucson, AZ 85721, USA (K.W.L.)
| | - Stacy M. Skopp
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, Tucson, AZ 85721, USA (K.W.L.)
| | - Tobias Fauser
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, Tucson, AZ 85721, USA (K.W.L.)
| | - Bailey Lyttle
- Department of Surgery, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA; (J.R.B.)
| | - Alyssa E. Vaughn
- Department of Surgery, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA; (J.R.B.)
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center, University of Central Florida, Orlando, FL 32827, USA
| | | | - Kenneth W. Liechty
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, Tucson, AZ 85721, USA (K.W.L.)
- Ceria Therapeutics, Inc., Tucson, AZ 85721, USA
| | - Carlos Zgheib
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, Tucson, AZ 85721, USA (K.W.L.)
- Ceria Therapeutics, Inc., Tucson, AZ 85721, USA
| |
Collapse
|