1
|
Caballero Mateos AM, Cañadas de la Fuente GA, Gros B. Paradigm Shift in Inflammatory Bowel Disease Management: Precision Medicine, Artificial Intelligence, and Emerging Therapies. J Clin Med 2025; 14:1536. [PMID: 40095460 PMCID: PMC11899940 DOI: 10.3390/jcm14051536] [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: 12/23/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
Inflammatory bowel disease (IBD) management stands at the cusp of a transformative era, with recent breakthroughs heralding a paradigm shift in treatment strategies. Traditionally, IBD therapeutics revolved around immunosuppressants, but the landscape has evolved significantly. Recent approvals of etrasimod, upadacitinib, mirikizumab, and risankizumab have introduced novel mechanisms of action, offering renewed hope for IBD patients. These medications represent a departure from the status quo, breaking years of therapeutic stagnation. Precision medicine, involving Artificial Intelligence, is a pivotal aspect of this evolution, tailoring treatments based on genetic profiles, disease characteristics, and individual responses. This approach optimizes treatment efficacy, and paves the way for personalized care. Yet, the rising cost of IBD therapies, notably biologics, poses challenges, impacting healthcare budgets and patient access. Ongoing research strives to assess cost-effectiveness, guiding policy decisions to ensure equitable access to advanced treatments. Looking ahead, the future of IBD management holds great promise. Emerging therapies, precision medicine, and ongoing research into novel targets promise to reshape the IBD treatment landscape. As these advances continue to unfold, IBD patients can anticipate a brighter future, one marked by more effective, personalized, and accessible treatments.
Collapse
Affiliation(s)
- Antonio M. Caballero Mateos
- Department of Internal Medicine, Gastroenterology Unit, Hospital Santa Ana, 18600 Motril, Spain
- Institute of Biosanitary Research (IBS) Precision Medicine, 18012 Granada, Spain
| | - Guillermo A. Cañadas de la Fuente
- Department of Nursing, Faculty of Health Sciences, University of Granada, 18016 Granada, Spain;
- Brain, Mind and Behaviour Research Center (CIMCYC), University of Granada, Campus Universitario de Cartuja s/n, 18011 Granada, Spain
| | - Beatriz Gros
- Department of Gastroenterology and Hepatology, Reina Sofía University Hospital, IMIBIC, University of Cordoba, 14004 Cordoba, Spain;
- Biomedical Research Center in Hepatic and Digestive Disease, CIBEREHD, 28029 Madrid, Spain
| |
Collapse
|
2
|
Jurickova I, Dreskin BW, Angerman E, Bonkowski E, Nguyen J, Villarreal R, Tominaga K, Iwasawa K, Braun T, Takebe T, Helmrath MA, Haberman Y, Wells JM, Denson LA. Eicosatetraynoic Acid Regulates Profibrotic Pathways in an Induced Pluripotent Stem Cell-Derived Macrophage-Human Intestinal Organoid Model of Crohn's Disease. J Crohns Colitis 2025; 19:jjae139. [PMID: 39212594 PMCID: PMC11836882 DOI: 10.1093/ecco-jcc/jjae139] [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: 12/05/2023] [Revised: 07/19/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND AIMS We previously identified small molecules predicted to reverse an ileal gene signature for future Crohn's Disease (CD) strictures. Here we used a new human intestinal organoid (HIO) model system containing macrophages to test a lead candidate, eicosatetraynoic acid (ETYA). METHODS Induced pluripotent stem cell lines (iPSC) were derived from CD patients and differentiated into macrophages and HIOs. Macrophages and macrophage-HIO cocultures were exposed to lipopolysaccharide (LPS) with and without ETYA pretreatment. Cytospin and flow cytometry characterized macrophage morphology and activation markers, and RNA sequencing defined the global pattern of macrophage gene expression. TaqMan low-density array, Luminex multiplex assay, immunohistologic staining, and sirius red polarized light microscopy were performed to measure macrophage cytokine production and HIO profibrotic gene expression and collagen content. RESULTS Induced PSC-derived macrophages exhibited morphology similar to primary macrophages and expressed inflammatory macrophage cell surface markers including CD64 and CD68. LPS-stimulated macrophages expressed a global pattern of gene expression enriched in CD ileal inflammatory macrophages and matrisome-secreted products and produced cytokines and chemokines including CCL2, IL1B, and OSM implicated in refractory disease. ETYA suppressed CD64 abundance and profibrotic gene expression pathways in LPS-stimulated macrophages. Coculture of LPS-primed macrophages with HIO led to upregulation of fibroblast activation genes including ACTA2 and COL1A1, and an increase in HIO collagen content. ETYA pretreatment prevented profibrotic effects of LPS-primed macrophages. CONCLUSIONS ETYA inhibits profibrotic effects of LPS-primed macrophages upon cocultured HIO. This model may be used in future untargeted screens for small molecules to treat refractory CD.
Collapse
Affiliation(s)
- Ingrid Jurickova
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Benjamin W Dreskin
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Elizabeth Angerman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Erin Bonkowski
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jack Nguyen
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Richard Villarreal
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kentaro Tominaga
- Division of Gastroenterology and Hepatology, Niigata University, Niigata, Japan
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kentaro Iwasawa
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tzipi Braun
- Department of Pediatrics, Sheba Medical Center, Tel-HaShomer, Affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Takanori Takebe
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Institute of Research, Tokyo Medical and Dental University, Tokyo, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), and Division of Stem Cell and Organoid Medicine, Osaka University, Suita, Osaka, Japan
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael A Helmrath
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Pediatric General and Thoracic Surgery, Department of Surgery, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Yael Haberman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, Sheba Medical Center, Tel-HaShomer, Affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - James M Wells
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lee A Denson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
3
|
Petronio L, Dal Buono A, Gabbiadini R, Migliorisi G, Privitera G, Ferraris M, Loy L, Bezzio C, Armuzzi A. Drug Development in Inflammatory Bowel Diseases: What Is Next? Pharmaceuticals (Basel) 2025; 18:190. [PMID: 40006003 PMCID: PMC11858795 DOI: 10.3390/ph18020190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/18/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Inflammatory bowel diseases (IBDs), which include Crohn's disease (CD) and ulcerative colitis (UC), are chronic conditions requiring long-term therapy to maintain remission and improve quality of life. Despite the approval of numerous drugs, IBD continues to present treatment challenges. This review aims to summarize novel therapeutic target agents in phases II and III of development, including sphingosine-1-phosphate receptor modulators (S1P), anti-interleukin-23 (IL-23), and other small molecules and monoclonal antibodies currently under investigation (e.g., anti-TL1A, obefazimod, NX-13, RIPK-inhibitors). Methods: A comprehensive literature search was conducted up to December 2024 to identify relevant articles published in English over the past three-five years, focusing on phase II/III studies for UC and CD. The search included databases such as PubMed, Google Scholar, and the ClinicalTrials.gov portal. Results: Clinical trials underline the potential of novel immunomodulators, including anti-TL1A, obefazimod, NX-13, RIPK inhibitors, and anti-IL-23p19 agents, as promising therapeutic options for IBD. Anti-IL23p19 therapies, such as risankizumab and mirikizumab, alongside guselkumab, exemplify this class's growing clinical relevance. While some are already in clinical use, others are nearing approval. Conclusions: Ongoing research into long-term safety and the development of personalized treatment strategies remains pivotal to enhance outcomes. Patient stratification and the strategic positioning of these therapies within the expanding treatment landscape are critical for optimizing their clinical impact.
Collapse
Affiliation(s)
- Lorenzo Petronio
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.P.); (A.D.B.); (R.G.); (G.M.); (G.P.); (M.F.); (L.L.); (C.B.)
- Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
| | - Arianna Dal Buono
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.P.); (A.D.B.); (R.G.); (G.M.); (G.P.); (M.F.); (L.L.); (C.B.)
- Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
| | - Roberto Gabbiadini
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.P.); (A.D.B.); (R.G.); (G.M.); (G.P.); (M.F.); (L.L.); (C.B.)
| | - Giulia Migliorisi
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.P.); (A.D.B.); (R.G.); (G.M.); (G.P.); (M.F.); (L.L.); (C.B.)
- Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
| | - Giuseppe Privitera
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.P.); (A.D.B.); (R.G.); (G.M.); (G.P.); (M.F.); (L.L.); (C.B.)
- Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
| | - Matteo Ferraris
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.P.); (A.D.B.); (R.G.); (G.M.); (G.P.); (M.F.); (L.L.); (C.B.)
- Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
| | - Laura Loy
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.P.); (A.D.B.); (R.G.); (G.M.); (G.P.); (M.F.); (L.L.); (C.B.)
| | - Cristina Bezzio
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.P.); (A.D.B.); (R.G.); (G.M.); (G.P.); (M.F.); (L.L.); (C.B.)
- Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
| | - Alessandro Armuzzi
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.P.); (A.D.B.); (R.G.); (G.M.); (G.P.); (M.F.); (L.L.); (C.B.)
- Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
| |
Collapse
|
4
|
Lusetti F, Bezzio C, De Bernardi A, Dota M, Manes G, Saibeni S. The TL1A inhibitors in IBD: what's in the pot? Expert Rev Gastroenterol Hepatol 2025; 19:15-25. [PMID: 39772947 DOI: 10.1080/17474124.2025.2450795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION Inflammatory bowel diseases (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), present ongoing challenges despite advances in pathophysiological understanding and therapeutic options. Current therapies often fail to achieve sustained remission, necessitating exploration of novel treatment targets. AREAS COVERED This review explores the role of Tumor Necrosis Factor-like cytokine 1A (TL1A) and its receptor DR3 in IBD pathogenesis, detailing their involvement in mucosal homeostasis and immune modulation. Recent studies on TL1A inhibitors highlight their potential in mitigating inflammation and fibrosis in IBD. EXPERT OPINION TL1A inhibition emerges as a promising therapeutic strategy, supported by encouraging outcomes in clinical trials for moderate to severe IBD. Future research may elucidate TL1A's broader impact on immunity, epithelial integrity and fibrosis, offering new avenues for therapeutic intervention and biomarker discovery. Ongoing phase 3 trials are pivotal in assessing TL1A inhibitors as effective and safe treatments for IBD. Additionally, exploration of TL1A's role in fibrosis-associated complications and its potential as a biomarker for treatment response holds promise for personalized medicine approaches. Consideration of TL1A inhibition in concurrent immune-mediated inflammatory diseases suggests broader therapeutic implications beyond gastrointestinal manifestations of IBD.
Collapse
Affiliation(s)
- F Lusetti
- Gastroenterology Unit, Foundation Policlinico San Matteo IRCCS, University of Pavia, Pavia, Italy
- IBD Center, Gastroenterology Unit, Rho Hospital, ASST Rhodense, Rho, Italy
| | - C Bezzio
- IBD Center, Humanitas Clinical and Research Centre, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - A De Bernardi
- IBD Center, Gastroenterology Unit, Rho Hospital, ASST Rhodense, Rho, Italy
| | - M Dota
- Gastroenterology Unit, Foundation Policlinico San Matteo IRCCS, University of Pavia, Pavia, Italy
- IBD Center, Gastroenterology Unit, Rho Hospital, ASST Rhodense, Rho, Italy
| | - G Manes
- IBD Center, Gastroenterology Unit, Rho Hospital, ASST Rhodense, Rho, Italy
| | - S Saibeni
- IBD Center, Gastroenterology Unit, Rho Hospital, ASST Rhodense, Rho, Italy
| |
Collapse
|
5
|
Bertin L, Crepaldi M, Zanconato M, Lorenzon G, Maniero D, de Barba C, Bonazzi E, Facchin S, Scarpa M, Ruffolo C, Angriman I, Buda A, Zingone F, Barberio B, Savarino EV. Advancing therapeutic frontiers: a pipeline of novel drugs for luminal and perianal Crohn's disease management. Therap Adv Gastroenterol 2024; 17:17562848241303651. [PMID: 39711916 PMCID: PMC11660281 DOI: 10.1177/17562848241303651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/12/2024] [Indexed: 12/24/2024] Open
Abstract
Crohn's disease (CD) is a chronic, complex inflammatory disorder of the gastrointestinal tract that presents significant therapeutic challenges. Despite the availability of a wide range of treatments, many patients experience primary non-response, secondary loss of response, or adverse events, limiting the overall effectiveness of current therapies. Clinical trials often report response rates below 60%, partly due to stringent inclusion criteria. Emerging therapies that target novel pathways offer promise in overcoming these limitations. This review explores the latest investigational drugs in phases I, II, and III clinical trials for treating both luminal and perianal CD. We highlight promising therapies that target known mechanisms, including selective Janus kinase inhibitors, anti-adhesion molecules, tumor necrosis factor inhibitors, and IL-23 selective inhibitors. In addition, we delve into novel therapeutic strategies such as sphingosine-1-phosphate receptor modulators, miR-124 upregulators, anti-fractalkine (CX3CL1), anti-TL1A, peroxisome proliferator-activated receptor gamma agonists, TGFBRI/ALK5 inhibitors, anti-CCR9 agents, and other innovative small molecules, as well as combination therapies. These emerging approaches, by addressing new pathways and mechanisms of action, have the potential to surpass the limitations of existing treatments and significantly improve CD management. However, the path to developing new therapies for inflammatory bowel disease (IBD) is fraught with challenges, including complex trial designs, ethical concerns regarding placebo use, recruitment difficulties, and escalating costs. The landscape of IBD clinical trials is shifting toward greater inclusivity, improved patient diversity, and innovative trial designs, such as adaptive and Bayesian approaches, to address these challenges. By overcoming these obstacles, the drug development pipeline can advance more effective, accessible, and timely treatments for CD.
Collapse
Affiliation(s)
- Luisa Bertin
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Martina Crepaldi
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Miriana Zanconato
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Greta Lorenzon
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Daria Maniero
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Caterina de Barba
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Erica Bonazzi
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Sonia Facchin
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Marco Scarpa
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padua, Italy
| | - Cesare Ruffolo
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padua, Italy
| | - Imerio Angriman
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padua, Italy
| | - Andrea Buda
- Gastroenterology Unit, Department of Oncological Gastrointestinal Surgery, Santa Maria del Prato Hospital, Feltre, Italy
| | - Fabiana Zingone
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Brigida Barberio
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Edoardo Vincenzo Savarino
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani, 2, Padua 35128, Italy
| |
Collapse
|
6
|
Jarmakiewicz-Czaja S, Sokal-Dembowska A, Ferenc K, Filip R. Mechanisms of Insulin Signaling as a Potential Therapeutic Method in Intestinal Diseases. Cells 2024; 13:1879. [PMID: 39594627 PMCID: PMC11593555 DOI: 10.3390/cells13221879] [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: 07/16/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Gastrointestinal diseases are becoming a growing public health problem. One of them is inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD). The incidence of IBD is increasing in developing countries and declining in developed countries, affecting people of all ages. Researchers have been exploring new treatment options including insulin signaling pathways in the inflammation of the gastrointestinal tract. It seems that a better understanding of the mechanism of IGF-1, GLP-1 and TL1A on the gut microbiota and inflammation may provide new advances in future therapeutic strategies for patients with IBD, but also other intestinal diseases. This review aims to synthesize insights into the effects of GLP, IGF and anti-TL1A on inflammation and the gut microbiota, which may enable their future use in therapy for people with intestinal diseases.
Collapse
Affiliation(s)
- Sara Jarmakiewicz-Czaja
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Aneta Sokal-Dembowska
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Katarzyna Ferenc
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland;
| | - Rafał Filip
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland;
- Department of Gastroenterology with IBD Unit, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| |
Collapse
|
7
|
Noor NM, Bourke A, Subramanian S. Review article: Novel therapies in inflammatory bowel disease - An update for clinicians. Aliment Pharmacol Ther 2024; 60:1244-1260. [PMID: 39403052 DOI: 10.1111/apt.18294] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/17/2024] [Accepted: 09/04/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Several new treatments including small molecules and biologics have been approved for the treatment of inflammatory bowel diseases in recent years. Clinicians and patients now have a wide variety of therapeutic options to choose from and these novel therapies provide several advantages including oral administration, lower immunogenicity, better selectivity and arguably better safety profiles. An increase in treatment options has increased the complexity of decision-making. Both patients and clinicians have had to become rapidly familiar with efficacy of new medications balanced against a range of pre-initiation requirements, dosing schedules and adverse event profiles. AIMS To provide a simple guide to practising clinicians on recently approved and emerging therapies and address key challenges around treatment strategies such as optimal sequencing and timing of treatment. METHODS We comprehensively searched the published literature and major conference abstracts to identify phase III placebo-controlled and active comparator trials for Crohn's disease and ulcerative colitis. RESULTS Data for recently approved therapies including selective Janus kinase inhibitors, sphingosine-1 receptor modulators and p19 interleukin (IL)-23 inhibitors have demonstrated improved patient outcomes in both Crohn's disease and ulcerative colitis. Further comparative head-to-head studies have improved our understanding of when and how to optimally use newer therapies, specifically for IL-23 inhibitors. Data for emerging treatment options and novel treatment strategies such as early effective treatment, combinations of treatments and implications for sequencing are continuing to transform IBD care continually. CONCLUSIONS Recently approved novel therapies have expanded the range of medical options available to treat IBD. However, further data from long-term extension studies, real-world studies and head-to-head trials are warranted to better inform the long-term safety and optimal sequencing of treatments for patients living with IBD.
Collapse
Affiliation(s)
- Nurulamin M. Noor
- Department of Gastroenterology, Cambridge University Hospital NHS Foundation Trust, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Aoibh Bourke
- Department of Gastroenterology, Cambridge University Hospital NHS Foundation Trust, Cambridge, UK
| | - Sreedhar Subramanian
- Department of Gastroenterology, Cambridge University Hospital NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
8
|
Chauhan G, Massey WJ, Veisman I, Rieder F. Anti-fibrotics in inflammatory bowel diseases: Challenges and successes. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:85-106. [PMID: 39521606 DOI: 10.1016/bs.apha.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Stricture formation leading to obstruction in Crohn's disease (CD) remains one of the largest unmet needs in the field of inflammatory bowel diseases (IBD). Despite this need no selective anti-stricture drug has been approved for use in CD patients. This contrasts with other fibrotic diseases, such as in the lung, liver or kidney, where multiple drug development programs crossed the starting line and two anti-fibrotics are now being approved for pulmonary fibrosis. Strictures are composed of a mix of inflammation, excessive deposition of extracellular matrix (ECM) and smooth muscle hyperplasia, likely all ultimately being responsible for the luminal narrowing driving patient symptoms. Our understanding of the pathogenesis of stricturing CD has evolved and indicates a multifactorial process involving immune and non-immune cells and their soluble mediators. This understanding has rendered target pathways for anti-stricture drug development. Significant progress was made in creating consensus definitions and tools to enable clinical trials with two clinical development programs having been conceived to date. In this chapter, we discuss stricture pathogenesis with a focus on the pathways being tested in clinical trials, and clinical trial endpoints developed for this indication.
Collapse
Affiliation(s)
- Gaurav Chauhan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - William J Massey
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Ido Veisman
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States; Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic Foundation, Cleveland, OH, United States.
| |
Collapse
|
9
|
Ma C, Huang J, Zheng Y, Na Y, Wei J, Shan J, Meng K, Zhang X, Zhang S, Wen Y, Ding J. Anti-TL1A monoclonal antibody modulates the dysregulation of Th1/Th17 cells and attenuates granuloma formation in sarcoidosis by inhibiting the PI3K/AKT signaling pathway. Int Immunopharmacol 2024; 137:112360. [PMID: 38852524 DOI: 10.1016/j.intimp.2024.112360] [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: 05/26/2024] [Accepted: 05/26/2024] [Indexed: 06/11/2024]
Abstract
Sarcoidosis is a systemic granulomatous disease characterized by non-caseating epithelioid cell granulomas. One of its immunological hallmarks is the differentiation of CD4 + naïve T cells into Th1/Th17 cells, accompanied by the release of numerous pro-inflammatory cytokines. The TL1A/DR3 signaling pathway plays a crucial role in activating effector lymphocytes, thereby triggering pro-inflammatory responses. The primary aim of this investigation was to scrutinize the impact of anti-TL1A monoclonal antibody on the dysregulation of Th1/Th17 cells and granuloma formation in sarcoidosis. Initially, the abnormal activation of the TL1A/DR3 signaling pathway in pulmonary tissues of sarcoidosis patients was confirmed using qPCR and immunohistochemistry techniques. Subsequently, employing a murine model of sarcoidosis, the inhibitory effects of anti-TL1A monoclonal antibody on the TL1A/DR3 signaling pathway in sarcoidosis were investigated through qPCR, immunohistochemistry, and Western blot experiments. The influence of anti-TL1A monoclonal antibody on granulomas was assessed through HE staining, while their effects on sarcoidosis Th1/Th17 cells and associated cytokine mRNA levels were evaluated using flow cytometry and qPCR, respectively. Immunofluorescence and Western blot experiments corroborated the inhibitory effects of anti-TL1A monoclonal antibody on the aberrant activation of the PI3K/AKT signaling pathway in sarcoidosis. The findings of this study indicate that the TL1A/DR3 signaling pathway is excessively activated in sarcoidosis. Anti-TL1A monoclonal antibody effectively inhibit this abnormal activation in sarcoidosis, thereby alleviating the dysregulation of Th1/Th17 cells and reducing the formation of pulmonary granulomas. This effect may be associated with the inhibition of the downstream PI3K/AKT signaling pathway. Anti-TL1A monoclonal antibody hold promise as a potential novel therapeutic intervention for sarcoidosis.
Collapse
Affiliation(s)
- Chengxing Ma
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiayi Huang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yin Zheng
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yuqi Na
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Jia Wei
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiajia Shan
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Kaifang Meng
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Xian Zhang
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Shiyun Zhang
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Yanting Wen
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China.
| | - Jingjing Ding
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
10
|
Schweckendiek D, Rogler G. Antibodies Targeting the Tumor Necrosis Factor-Like Ligand 1A in Inflammatory Bowel Disease: A New Kid on the (Biologics) Block? Digestion 2024; 105:411-418. [PMID: 39068930 PMCID: PMC11633870 DOI: 10.1159/000540421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND The treatment options for inflammatory bowel disease (IBD) have grown over the last years. However, a significant fraction of patients either do not respond to their treatment or lose response over time. SUMMARY Future treatment options could include antibodies that target the tumor necrosis factor-like ligand 1A (TL1A). TL1A is a key cytokine involved in the pathogenesis of a variety of autoimmune diseases including IBD. Studies have shown that IBD disease severity correlates well with serum levels of TL1A. Phase 2 data from two agents currently in clinical testing have been released. In line with requirements for modern therapeutics, companion diagnostic was part of these trials. This aims to identify those patients that are more likely to respond to the agents tested. KEY MESSAGES With regard to the available data the risk/benefit profile of TL1A inhibitors seems to be promising. This article gives a short update and overview, where we are at this point in time with antibodies targeting the TL1A protein in IBD.
Collapse
Affiliation(s)
- Daniel Schweckendiek
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Wang J, Yang B, Chandra J, Ivanov A, Brown JM, Florian R. Preventing fibrosis in IBD: update on immune pathways and clinical strategies. Expert Rev Clin Immunol 2024; 20:727-734. [PMID: 38475672 PMCID: PMC11180587 DOI: 10.1080/1744666x.2024.2330604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 03/11/2024] [Indexed: 03/14/2024]
Abstract
INTRODUCTION Intestinal fibrosis is a common and serious complication of inflammatory bowel diseases (IBD) driving stricture formation in Crohn's disease patients and leading to submucosal damage in ulcerative colitis. Recent studies provided novel insights into the role of immune and nonimmune components in the pathogenesis of intestinal fibrosis. Those new findings may accelerate the development of anti-fibrotic treatment in IBD patients. AREAS COVERED This review is designed to cover the recent progress in mechanistic research and therapeutic developments on intestinal fibrosis in IBD patients, including new cell clusters, cytokines, proteins, microbiota, creeping fat, and anti-fibrotic therapies. EXPERT OPINION Due to the previously existing major obstacle of missing consensus on stricture definitions and the absence of clinical trial endpoints, testing of drugs with an anti-fibrotic mechanism is just starting in stricturing Crohn's disease (CD). A biomarker to stratify CD patients at diagnosis without any complications into at-risk populations for future strictures would be highly desirable. Further investigations are needed to identify novel mechanisms of fibrogenesis in the intestine that are targetable and ideally gut specific.
Collapse
Affiliation(s)
- Jie Wang
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Bo Yang
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Jyotsna Chandra
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Andrei Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - J. Mark Brown
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Rieder Florian
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
- Program for Global Translational Inflammatory Bowel Diseases, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
12
|
Zhan RR, Wang D, Zhang XL. Progress in research of TNF-like cytokine 1A as a therapeutic target for inflammatory bowel disease. WORLD CHINESE JOURNAL OF DIGESTOLOGY 2024; 32:397-404. [DOI: 10.11569/wcjd.v32.i6.397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
|
13
|
Matellan C, Kennedy C, Santiago-Vela MI, Hochegger J, Ní Chathail MB, Wu A, Shannon C, Roche HM, Aceves SS, Godson C, Manresa MC. The TNFSF12/TWEAK Modulates Colonic Inflammatory Fibroblast Differentiation and Promotes Fibroblast-Monocyte Interactions. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1958-1970. [PMID: 38700420 PMCID: PMC11149899 DOI: 10.4049/jimmunol.2300762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/29/2024] [Indexed: 05/05/2024]
Abstract
Fibroblasts acquire a proinflammatory phenotype in inflammatory bowel disease, but the factors driving this process and how fibroblasts contribute to mucosal immune responses are incompletely understood. TNF superfamily member 12 (TNFSF12, or TNF-like weak inducer of apoptosis [TWEAK]) has gained interest as a mediator of chronic inflammation. In this study, we explore its role as a driver of inflammatory responses in fibroblasts and its contribution to fibroblast-monocyte interaction using human primary colonic fibroblasts, THP-1 and primary monocytes. Recombinant human TWEAK induced the expression of cytokines, chemokines, and immune receptors in primary colonic fibroblasts. The TWEAK upregulated transcriptome shared 29% homology with a previously published transcriptional profile of inflammatory fibroblasts from ulcerative colitis. TWEAK elevated surface expression of activated fibroblast markers and adhesion molecules (podoplanin [PDPN], ICAM-1, and VCAM-1) and secretion of IL-6, CCL2, and CXCL10. In coculture, fibroblasts induced monocyte adhesion and secretion of CXCL1 and IL-8, and they promoted a CD14high/ICAM-1high phenotype in THP-1 cells, which was enhanced when fibroblasts were prestimulated with TWEAK. Primary monocytes in coculture with TWEAK-treated fibroblasts had altered surface expression of CD16 and triggering receptor expressed on myeloid cells-1 (TREM-1) as well as increased CXCL1 and CXCL10 secretion. Conversely, inhibition of the noncanonical NF-κB pathway on colonic fibroblasts with a NF-κB-inducing kinase small molecule inhibitor impaired their ability to induce a CD14high phenotype on monocytes. Our results indicate that TWEAK promotes an inflammatory fibroblast-monocyte crosstalk that may be amenable for therapeutic intervention.
Collapse
Affiliation(s)
- Carlos Matellan
- School of Biomolecular and Biomedical Sciences, University College Dublin, Belfield, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, Conway Institute of Biomolecular and biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Ciarán Kennedy
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, Conway Institute of Biomolecular and biomedical Research, University College Dublin, Belfield, Dublin, Ireland
- Diabetes Complications Research Centre, University College Dublin, Belfield, Dublin, Ireland
| | - Miren Itxaso Santiago-Vela
- School of Biomolecular and Biomedical Sciences, University College Dublin, Belfield, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Johanna Hochegger
- School of Biomolecular and Biomedical Sciences, University College Dublin, Belfield, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Méabh B. Ní Chathail
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
- School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Belfield, Dublin, Ireland
| | - Amanda Wu
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA
| | - Christopher Shannon
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, Conway Institute of Biomolecular and biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Helen M. Roche
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
- School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Belfield, Dublin, Ireland
- Institute for Global Food Security, Queen’s University Belfast, Belfast, U.K
| | - Seema S. Aceves
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA
- Rady Children’s Hospital, San Diego, CA
| | - Catherine Godson
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, Conway Institute of Biomolecular and biomedical Research, University College Dublin, Belfield, Dublin, Ireland
- Diabetes Complications Research Centre, University College Dublin, Belfield, Dublin, Ireland
| | - Mario C. Manresa
- School of Biomolecular and Biomedical Sciences, University College Dublin, Belfield, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
14
|
Zhijia S, Meijuan X, Yanting Z, Fang L, Minyu F, Ruirui C, Li C, Jingyan Z, Limei Y. Illness Perceptions as a Predictor of Symptom Cluster Trajectories in Patients With Inflammatory Bowel Disease: A Latent Growth Mixture Model. Gastroenterol Nurs 2024; 47:111-121. [PMID: 38567854 DOI: 10.1097/sga.0000000000000793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 09/20/2023] [Indexed: 04/05/2024] Open
Abstract
The aims of this study were to (a) identify the trajectory of symptom clusters in patients with inflammatory bowel disease up to 28 weeks after initiation of infliximab therapy and (b) examine the illness perceptions associated with symptom cluster trajectories. This was a prospective study where participants completed the symptom cluster scale at baseline, 14 weeks, and 28 weeks. A latent growth mixture modeling was used to identify trajectories of symptom clusters that were predicted, using baseline covariates (Brief Illness Perception Questionnaire). A total of 206 patients were included and identified as three latent classes: moderate symptom cluster-stable decline group (C1), high symptom cluster-rapid decline group (C2), and stable symptom cluster-stable trend group (C3). C1 was predicted by cognitive illness perceptions (odds ratio [95% confidence interval]: 1.134 [1.071, 1.200], p < .001). C2 was also predicted by cognitive and emotional illness perceptions (odds ratio [95% confidence interval]: 1.169 [1.095, 1.248], p < .001; odds ratio [95% confidence interval]: 1.174 [1.038, 1.328], p = .011). Patients with inflammatory bowel disease, initiating infliximab therapy, had different symptom cluster trajectories. Illness perceptions were associated with symptom cluster classes, which underline the complexity of symptoms. Paying attention to these factors and providing necessary knowledge and psychological supporting care after infliximab therapy would effectively improve patients' symptom burden.
Collapse
Affiliation(s)
- Shen Zhijia
- Shen Zhijia, MSN, RN, is a Registered Nurse, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Xi Meijuan, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhou Yanting, BSN, RN, is a Head Nurse in Gastroenterology, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Li Fang, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Fan Minyu, MM, is Gastroenterologist, Oncology department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Chen Ruirui, MSN, RN, is a Registered Nurse, School of Medicine, Jiangsu University, Zhenjiang, China, and Gastroenterology Department, General Hospital of Eastern Theater Command, Jiangsu, China
- Chen Li, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhong Jingyan, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Yin Limei, BSN, RN, is a Head of the Nursing Department, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
| | - Xi Meijuan
- Shen Zhijia, MSN, RN, is a Registered Nurse, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Xi Meijuan, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhou Yanting, BSN, RN, is a Head Nurse in Gastroenterology, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Li Fang, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Fan Minyu, MM, is Gastroenterologist, Oncology department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Chen Ruirui, MSN, RN, is a Registered Nurse, School of Medicine, Jiangsu University, Zhenjiang, China, and Gastroenterology Department, General Hospital of Eastern Theater Command, Jiangsu, China
- Chen Li, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhong Jingyan, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Yin Limei, BSN, RN, is a Head of the Nursing Department, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
| | - Zhou Yanting
- Shen Zhijia, MSN, RN, is a Registered Nurse, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Xi Meijuan, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhou Yanting, BSN, RN, is a Head Nurse in Gastroenterology, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Li Fang, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Fan Minyu, MM, is Gastroenterologist, Oncology department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Chen Ruirui, MSN, RN, is a Registered Nurse, School of Medicine, Jiangsu University, Zhenjiang, China, and Gastroenterology Department, General Hospital of Eastern Theater Command, Jiangsu, China
- Chen Li, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhong Jingyan, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Yin Limei, BSN, RN, is a Head of the Nursing Department, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
| | - Li Fang
- Shen Zhijia, MSN, RN, is a Registered Nurse, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Xi Meijuan, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhou Yanting, BSN, RN, is a Head Nurse in Gastroenterology, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Li Fang, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Fan Minyu, MM, is Gastroenterologist, Oncology department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Chen Ruirui, MSN, RN, is a Registered Nurse, School of Medicine, Jiangsu University, Zhenjiang, China, and Gastroenterology Department, General Hospital of Eastern Theater Command, Jiangsu, China
- Chen Li, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhong Jingyan, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Yin Limei, BSN, RN, is a Head of the Nursing Department, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
| | - Fan Minyu
- Shen Zhijia, MSN, RN, is a Registered Nurse, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Xi Meijuan, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhou Yanting, BSN, RN, is a Head Nurse in Gastroenterology, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Li Fang, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Fan Minyu, MM, is Gastroenterologist, Oncology department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Chen Ruirui, MSN, RN, is a Registered Nurse, School of Medicine, Jiangsu University, Zhenjiang, China, and Gastroenterology Department, General Hospital of Eastern Theater Command, Jiangsu, China
- Chen Li, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhong Jingyan, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Yin Limei, BSN, RN, is a Head of the Nursing Department, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
| | - Chen Ruirui
- Shen Zhijia, MSN, RN, is a Registered Nurse, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Xi Meijuan, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhou Yanting, BSN, RN, is a Head Nurse in Gastroenterology, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Li Fang, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Fan Minyu, MM, is Gastroenterologist, Oncology department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Chen Ruirui, MSN, RN, is a Registered Nurse, School of Medicine, Jiangsu University, Zhenjiang, China, and Gastroenterology Department, General Hospital of Eastern Theater Command, Jiangsu, China
- Chen Li, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhong Jingyan, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Yin Limei, BSN, RN, is a Head of the Nursing Department, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
| | - Chen Li
- Shen Zhijia, MSN, RN, is a Registered Nurse, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Xi Meijuan, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhou Yanting, BSN, RN, is a Head Nurse in Gastroenterology, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Li Fang, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Fan Minyu, MM, is Gastroenterologist, Oncology department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Chen Ruirui, MSN, RN, is a Registered Nurse, School of Medicine, Jiangsu University, Zhenjiang, China, and Gastroenterology Department, General Hospital of Eastern Theater Command, Jiangsu, China
- Chen Li, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhong Jingyan, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Yin Limei, BSN, RN, is a Head of the Nursing Department, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
| | - Zhong Jingyan
- Shen Zhijia, MSN, RN, is a Registered Nurse, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Xi Meijuan, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhou Yanting, BSN, RN, is a Head Nurse in Gastroenterology, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Li Fang, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Fan Minyu, MM, is Gastroenterologist, Oncology department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Chen Ruirui, MSN, RN, is a Registered Nurse, School of Medicine, Jiangsu University, Zhenjiang, China, and Gastroenterology Department, General Hospital of Eastern Theater Command, Jiangsu, China
- Chen Li, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhong Jingyan, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Yin Limei, BSN, RN, is a Head of the Nursing Department, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
| | - Yin Limei
- Shen Zhijia, MSN, RN, is a Registered Nurse, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Xi Meijuan, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhou Yanting, BSN, RN, is a Head Nurse in Gastroenterology, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Li Fang, MM, is Gastroenterologist, Gastroenterology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Fan Minyu, MM, is Gastroenterologist, Oncology department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Chen Ruirui, MSN, RN, is a Registered Nurse, School of Medicine, Jiangsu University, Zhenjiang, China, and Gastroenterology Department, General Hospital of Eastern Theater Command, Jiangsu, China
- Chen Li, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Zhong Jingyan, BSN, RN, is a Clinical Nurse Specialist, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
- Yin Limei, BSN, RN, is a Head of the Nursing Department, Nursing Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, SuZhou, China
| |
Collapse
|
15
|
Niu W, Liu Q, Huo X, Luo Y, Zhang X. TL1A promotes metastasis and EMT process of colorectal cancer. Heliyon 2024; 10:e24392. [PMID: 38312710 PMCID: PMC10835226 DOI: 10.1016/j.heliyon.2024.e24392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 12/15/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
BACKGROUND Metastasis is the major problem of colorectal cancer (CRC) and is correlated with the high mortality. Tumor necrosis factor-like cytokine 1A (TL1A) is a novel regulatory factor for inflammatory diseases. This work aimed to investigate the role of TL1A in CRC metastasis. METHOD AOM/DSS-induced mouse model, xenograft tumor model and metastasis murine model were established to mimic the colitis-associated CRC and investigate CRC growth and metastasis in vivo. Colon tissues were assessed by hematoxylin/eosin (HE) staining and immunohistochemistry (IHC). CRC cell metastasis in vivo was observed using in vivo imaging system (IVIS). Cell viability and proliferation were examined using cell counting kit 8 (CCK-8) and EdU experiments. The expression of tumor growth factor β (TGFβ) and metastatic biomarkers were detected using western blotting experiment. The in vitro cell metastasis was measured by Transwell. RESULTS Knockdown of TL1A notably suppressed the generation of colonic tumors in azoxymethane/dextran sodium sulfate (AOM/DSS) model, suppressed in vivo CRC cell growth, as well as lung and liver metastasis. The inflammation response and inflammatory cell infiltration in tumor sites were decreased by TL1A depletion. The in vitro CRC cell growth and metastasis was also suppressed by shTL1A, along with altered expression of epithelial mesenchymal transition (EMT) biomarkers. TL1A depletion suppressed the level of the TGF-β1 receptor (TβRI) and phosphorylation of Smad3 in CRC cells. Stimulation with TGF-β recovered the CRC cell migration and invasion that suppressed by shTL1A. CONCLUSION Our work implicated TL1A as a promoter of CRC generation and metastasis and defines TGF-β/Smad3 signaling as mediator of TL1A-regualated CRC cell metastasis.
Collapse
Affiliation(s)
- Weiwei Niu
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| | - Qian Liu
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| | - Xiaoxia Huo
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| | - Yuxin Luo
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| | - Xiaolan Zhang
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| |
Collapse
|
16
|
Jurickova I, Dreskin BW, Angerman E, Bonkowski E, Tominaga K, Iwasawa K, Braun T, Takebe T, Helmrath MA, Haberman Y, Wells JM, Denson LA. Eicosatetraynoic Acid Regulates Pro-Fibrotic Pathways in an Induced Pluripotent Stem Cell Derived Macrophage:Human Intestinal Organoid Model of Crohn's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.30.577959. [PMID: 38352573 PMCID: PMC10862816 DOI: 10.1101/2024.01.30.577959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Background and Aims We previously identified small molecules predicted to reverse an ileal gene signature for future Crohn's Disease (CD) strictures. Here we used a new human intestinal organoid (HIO) model system containing macrophages to test a lead candidate, eicosatetraynoic acid (ETYA). Methods Induced pluripotent stem cell lines (iPSC) were derived from CD patients and differentiated into macrophages and HIOs. Macrophages and macrophage:HIO co-cultures were exposed to lipopolysaccharide (LPS) with and without ETYA pre-treatment. Cytospin and flow cytometry characterized macrophage morphology and activation markers, and RNA sequencing defined the global pattern of macrophage gene expression. TaqMan Low Density Array, Luminex multiplex assay, immunohistologic staining, and sirius red polarized light microscopy were performed to measure macrophage cytokine production and HIO pro-fibrotic gene expression and collagen content. Results iPSC-derived macrophages exhibited morphology similar to primary macrophages and expressed inflammatory macrophage cell surface markers including CD64 and CD68. LPS-stimulated macrophages expressed a global pattern of gene expression enriched in CD ileal inflammatory macrophages and matrisome secreted products, and produced cytokines and chemokines including CCL2, IL1B, and OSM implicated in refractory disease. ETYA suppressed CD64 abundance and pro-fibrotic gene expression pathways in LPS stimulated macrophages. Co-culture of LPS-primed macrophages with HIO led to up-regulation of fibroblast activation genes including ACTA2 and COL1A1 , and an increase in HIO collagen content. ETYA pre-treatment prevented pro-fibrotic effects of LPS-primed macrophages. Conclusions ETYA inhibits pro-fibrotic effects of LPS-primed macrophages upon co-cultured HIO. This model may be used in future untargeted screens for small molecules to treat refractory CD.
Collapse
|
17
|
Ratajczak F, Joblin M, Hildebrandt M, Ringsquandl M, Falter-Braun P, Heinig M. Speos: an ensemble graph representation learning framework to predict core gene candidates for complex diseases. Nat Commun 2023; 14:7206. [PMID: 37938585 PMCID: PMC10632370 DOI: 10.1038/s41467-023-42975-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023] Open
Abstract
Understanding phenotype-to-genotype relationships is a grand challenge of 21st century biology with translational implications. The recently proposed "omnigenic" model postulates that effects of genetic variation on traits are mediated by core-genes and -proteins whose activities mechanistically influence the phenotype, whereas peripheral genes encode a regulatory network that indirectly affects phenotypes via core gene products. Here, we develop a positive-unlabeled graph representation-learning ensemble-approach based on a nested cross-validation to predict core-like genes for diverse diseases using Mendelian disorder genes for training. Employing mouse knockout phenotypes for external validations, we demonstrate that core-like genes display several key properties of core genes: Mouse knockouts of genes corresponding to our most confident predictions give rise to relevant mouse phenotypes at rates on par with the Mendelian disorder genes, and all candidates exhibit core gene properties like transcriptional deregulation in disease and loss-of-function intolerance. Moreover, as predicted for core genes, our candidates are enriched for drug targets and druggable proteins. In contrast to Mendelian disorder genes the new core-like genes are enriched for druggable yet untargeted gene products, which are therefore attractive targets for drug development. Interpretation of the underlying deep learning model suggests plausible explanations for our core gene predictions in form of molecular mechanisms and physical interactions. Our results demonstrate the potential of graph representation learning for the interpretation of biological complexity and pave the way for studying core gene properties and future drug development.
Collapse
Affiliation(s)
- Florin Ratajczak
- Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Munich, Neuherberg, Germany
| | | | | | | | - Pascal Falter-Braun
- Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Munich, Neuherberg, Germany.
- Microbe-Host Interactions, Faculty of Biology, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
| | - Matthias Heinig
- Institute of Computational Biology (ICB), Helmholtz Munich, Neuherberg, Germany.
- Department of Computer Science, TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany.
- German Centre for Cardiovascular Research (DZHK), Munich Heart Association, Partner Site Munich, Berlin, Germany.
| |
Collapse
|
18
|
Bretto E, Ribaldone DG, Caviglia GP, Saracco GM, Bugianesi E, Frara S. Inflammatory Bowel Disease: Emerging Therapies and Future Treatment Strategies. Biomedicines 2023; 11:2249. [PMID: 37626745 PMCID: PMC10452708 DOI: 10.3390/biomedicines11082249] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a term used to represent a group of chronic, relapsing inflammatory disorders of the gastrointestinal tract. Crohn's disease (CD) and ulcerative colitis (UC) are the two major clinical forms. The global incidence and prevalence of IBD have increased over the last 2-4 decades. Despite the specific etiopathogenesis of IBD still being unknown, it is widely recognized that immunological, genetic, and environmental factors are implicated. A greater understanding of the multiple signaling pathways involved has led to the development of biologic therapies in the last two decades. Although these treatments have dramatically transformed the course of IBD, there is not a definitive cure and available therapies may cause adverse events (AEs), limiting their use, or have an inadequate effect in some patients. In this context, emerging therapies addressing new specific pathogenetic mechanisms have shown promising efficacy and safety data in early clinical trials. The purpose of this review is to highlight the available clinical trial data for these new drugs, such as more preferential JAK inhibitors, anti-IL-23 antibodies, sphingosine-1-phosphate receptor modulators, anti-integrin therapies, and other small molecules that are currently under research. We will emphasize the potential significance of these agents in shaping future treatment options.
Collapse
Affiliation(s)
- Elisabetta Bretto
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.B.)
- Unit of Gastroenterology, Città della Salute e della Scienza di Torino-Molinette Hospital, 10126 Turin, Italy
| | - Davide Giuseppe Ribaldone
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.B.)
- Unit of Gastroenterology, Città della Salute e della Scienza di Torino-Molinette Hospital, 10126 Turin, Italy
| | - Gian Paolo Caviglia
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.B.)
| | - Giorgio Maria Saracco
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.B.)
- Unit of Gastroenterology, Città della Salute e della Scienza di Torino-Molinette Hospital, 10126 Turin, Italy
| | - Elisabetta Bugianesi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.B.)
- Unit of Gastroenterology, Città della Salute e della Scienza di Torino-Molinette Hospital, 10126 Turin, Italy
| | - Simone Frara
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.B.)
- Unit of Gastroenterology, Città della Salute e della Scienza di Torino-Molinette Hospital, 10126 Turin, Italy
| |
Collapse
|
19
|
Tian L, Tao S, He C, Dong S, Chen Y, Chen L, Jiang S. Astragaloside IV regulates TL1A and NF-κB signal pathway to affect inflammation in necrotizing enterocolitis. Tissue Cell 2023; 83:102128. [PMID: 37413858 DOI: 10.1016/j.tice.2023.102128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/24/2023] [Accepted: 06/06/2023] [Indexed: 07/08/2023]
Abstract
AIM This study aims to explore the possible effect of Astragaloside IV (AS-IV) on necrotizing enterocolitis (NEC) neonatal rat models and verify the possible implication of TNF-like ligand 1 A (TL1A) and NF-κB signal pathway. METHODS NEC neonatal rat models were established through formula feeding, cold/asphyxia stress and Lipopolysaccharide (LPS) gavage method. The appearance, activity and skin as well as the pathological status of rats subjected to NEC modeling were assessed. The intestinal tissues were observed after H&E staining. The expression of oxidative stress biomarkers (SOD, MDA and GSH-Px) and inflammatory cytokines (TNF-α, IL-1β and IL-6) were detected by ELISA and qRT-PCR. Western blotting and immunohistochemistry were applied to detect expressions of TL1A and NF-κB signal pathway-related proteins. Cell apoptosis was assessed by TUNEL. RESULTS NEC neonatal rat models were established successfully, in which TL1A was highly expressed and NF-κB signal pathway was activated, while TL1A and NF-κB signal pathway can be suppressed by AS-IV treatment in NEC rats. Meanwhile, inflammatory response in intestinal tissues was increased in NEC rat models and AS-IV can attenuate inflammatory response in NEC rats through inhibiting TL1A and NF-κb signal pathway. CONCLUSION AS-IV can inhibit TL1A expression and NF-κb signal pathway to attenuate the inflammatory response in NEC neonatal rat models.
Collapse
Affiliation(s)
- Lin Tian
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China
| | - Shuang Tao
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China.
| | - Chunzhi He
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China
| | - Shanwu Dong
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China
| | - Yongli Chen
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China
| | - Ling Chen
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China
| | - Shu Jiang
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China
| |
Collapse
|
20
|
Higashiyama M, Hokari R. New and Emerging Treatments for Inflammatory Bowel Disease. Digestion 2022; 104:74-81. [PMID: 36366823 DOI: 10.1159/000527422] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/05/2022] [Indexed: 11/12/2022]
Abstract
BACKGROUND The specific etiopathogenesis of inflammatory bowel disease (IBD) is still unknown. Although the conventional anti-inflammatory or immunomodulatory drugs relatively nonspecific to pathogenesis have been quite useful in many cases, elucidating the pathogenesis has gradually facilitated developments of disease-specific therapies for refractory cases in the last 2 decades. SUMMARY With a greater understanding of the multiple overactive signaling pathways of the gut mucosal immune response and enhanced leukocyte trafficking, several biological agents or small molecule drugs following the first novel biologic, anti-tumor necrosis factor α (anti-TNFα), have been developed against several modes of action including adhesion molecules, sphingosine-1-phospate receptors, cytokines (IL-12/23, TL1A, and IL-36), Janus kinase (JAK), and phosphodiesterase. Although preceding biological agents have dramatically changed the IBD treatment strategy, many patients still require alternative therapies due to failure or side effects. Newer treatments are now expected to be provided for better efficacy with an improved adverse event profile. In addition, translational studies have highlighted the new therapeutic concepts' potential, including modulation of host-microbiome interactions, stem therapy for perianal fistula, regulation of fibrosis, regulation of the gut-brain axis, and control of previously less targeted immune cells (B cells and innate lymphoid cells). This paper comprehensively reviewed not only the latest already or shortly available therapies but also emerging promising treatments that will be hopefully established in the future for IBD. KEY MESSAGES Many kinds of new treatments are available, and promising treatments with new perspectives are expected to emerge for refractory IBD in the future.
Collapse
Affiliation(s)
- Masaaki Higashiyama
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Ryota Hokari
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| |
Collapse
|
21
|
Tackling Inflammatory Bowel Diseases: Targeting Proinflammatory Cytokines and Lymphocyte Homing. Pharmaceuticals (Basel) 2022; 15:ph15091080. [PMID: 36145301 PMCID: PMC9502105 DOI: 10.3390/ph15091080] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) are characterized by chronic inflammatory disorders that are a result of an abnormal immune response mediated by a cytokine storm and immune cell infiltration. Proinflammatory cytokine therapeutic agents, represented by TNF inhibitors, have developed rapidly over recent years and are promising options for treating IBD. Antagonizing interleukins, interferons, and Janus kinases have demonstrated their respective advantages in clinical trials and are candidates for anti-TNF therapeutic failure. Furthermore, the blockade of lymphocyte homing contributes to the excessive immune response in colitis and ameliorates inflammation and tissue damage. Factors such as integrins, selectins, and chemokines jointly coordinate the accumulation of immune cells in inflammatory regions. This review assembles the major targets and agents currently targeting proinflammatory cytokines and lymphatic trafficking to facilitate subsequent drug development.
Collapse
|