1
|
Górecka A, Komosinska-Vassev K. Neutrophil Elastase and Elafin in Inflammatory Bowel Diseases: Urinary Biomarkers Reflecting Intestinal Barrier Dysfunction and Proteolytic Activity. J Clin Med 2025; 14:2466. [PMID: 40217915 PMCID: PMC11989340 DOI: 10.3390/jcm14072466] [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: 03/03/2025] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), is a chronic inflammatory disorder driven by a complex interplay of immune and proteolytic mechanisms. Neutrophil elastase (NE), released at sites of inflammation, plays a central role by promoting inflammation, degrading the extracellular matrix (ECM), and disturbing intestinal barrier integrity via NF-κB activation and E-cadherin degradation. Elafin, an endogenous NE inhibitor, mitigates proteolytic damage, reinforces the intestinal barrier, and exerts anti-inflammatory effects by suppressing NF-κB and reducing pro-inflammatory cytokines. Since the NE/elafin balance is critical in IBD, assessing their ratio may provide a more precise measure of proteolytic dysregulation. This study aimed to evaluate the diagnostic and prognostic utility of urinary NE, elafin, and their ratio in IBD patients. Methods: Urinary concentrations of NE and elafin were measured by immunoassay in 88 subjects including ulcerative colitis and Crohn's disease patients and healthy individuals. The diagnostic accuracy of these biomarkers was assessed using receiver operating characteristic (ROC) curve analysis. Results: Urinary NE levels were significantly elevated in both UC and CD patients compared to controls, with a 17-fold increase in the UC patients and a 28-fold increase in the CD patients (p < 0.0001). Elafin levels were also increased in IBD patients. The NE/elafin ratio was significantly increased in both disease groups, with a 4.5-fold increase in the UC and 5.6-fold increase in the CD patients compared to healthy controls. The ROC curve analysis demonstrated that the NE/elafin ratio is the most effective biomarker for distinguishing CD patients from healthy individuals (AUC = 0.896), with a high sensitivity (92.9%) and specificity (69.7%), making it a strong diagnostic tool. NE also showed an excellent diagnostic performance both in CD (AUC = 0.842) and UC (AUC = 0.880). The elafin urinary profile had a high diagnostic value, with a better accuracy in the UC patients (AUC = 0.772) than the CD patients (AUC = 0.674), though it was inferior to NE and NE/elafin. Conclusions: Our findings indicate that urinary NE, elafin, the and NE/elafin ratio have significant diagnostic value in differentiating IBD patients from healthy controls. The NE/elafin ratio and NE proved to be the most reliable urinary biomarkers in both CD and UC diagnosis, with a high predictive value and strong discriminatory power.
Collapse
Affiliation(s)
- Aleksandra Górecka
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland;
| | | |
Collapse
|
2
|
Baker ZR, Zhang Y, Zhang H, Franklin HC, Serpa PBS, Southard T, Li L, Hsu BB. Sustained in situ protein production and release in the mammalian gut by an engineered bacteriophage. Nat Biotechnol 2025:10.1038/s41587-025-02570-7. [PMID: 39966654 DOI: 10.1038/s41587-025-02570-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/20/2025] [Indexed: 02/20/2025]
Abstract
Oral administration of biologic drugs is challenging because of the degradative activity of the upper gastrointestinal tract. Strategies that use engineered microbes to produce biologics in the lower gastrointestinal tract are limited by competition with resident commensal bacteria. Here we demonstrate the engineering of bacteriophage (phage) that infect resident commensals to express heterologous proteins released during cell lysis. Working with the virulent T4 phage, which targets resident, nonpathogenic Escherichia coli, we first identify T4-specific promoters with maximal protein expression and minimal impact on T4 phage titers. We engineer T4 phage to express a serine protease inhibitor of a pro-inflammatory enzyme with increased activity in ulcerative colitis and observe reduced enzyme activity in a mouse model of colitis. We also apply the approach to reduce weight gain and inflammation in mouse models of diet-induced obesity. This work highlights an application of virulent phages in the mammalian gut as engineerable vectors to release therapeutics from resident gut bacteria.
Collapse
Affiliation(s)
- Zachary R Baker
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA
- Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Yao Zhang
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Haiyan Zhang
- Metabolism Core, Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA
| | - Hollyn C Franklin
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA
- Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Priscila B S Serpa
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Teresa Southard
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA.
| | - Bryan B Hsu
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA.
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA.
- Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Virginia Tech, Blacksburg, VA, USA.
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
3
|
Jun YK, Oh HJ, Lee JA, Choi Y, Shin CM, Park YS, Kim N, Lee DH, Yoon H. The Potential of Molecular Remission: Tissue Neutrophil Elastase Is Better Than Histological Activity for Predicting Long-Term Relapse in Patients With Ulcerative Colitis in Endoscopic Remission. Inflamm Bowel Dis 2025; 31:514-523. [PMID: 39191527 DOI: 10.1093/ibd/izae194] [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: 05/17/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Growing interest exists in deep remission, beyond clinical and endoscopic remission, to enhance long-term prognosis in patients with ulcerative colitis (UC). Our study aimed to evaluate the risk of relapse according to tissue expression levels of calprotectin and neutrophil elastase (NE) in patients with quiescent UC. METHODS Rectal biopsies were performed on 218 patients with UC in clinical and endoscopic remission. Histological activity was prospectively scored using the Robarts Histological Index. Tissue calprotectin and NE levels were evaluated using immunohistochemistry. Optimal tissue calprotectin and NE cutoffs for relapse were determined using log-rank analysis. Cox proportional hazard analyses evaluated relapse risk factors. RESULTS Tissue calprotectin and NE levels were significantly higher in patients with histological activity than in those in histological remission (P < .001). The optimal cutoffs of tissue calprotectin and NE for relapse were 10.61 and 22.08 per mm2, respectively. The 3-year clinical relapse risk was significantly lower in the low-tissue NE group than in the high-tissue NE group (P = .009); however, it did not differ between the low- and high-tissue calprotectin group (P = .094). In multivariate analyses, a low level of tissue NE expression was independently associated with a lower risk of 3-year clinical relapse (adjusted hazard ratio = 0.453, 95% confidence interval = 0.225-0.911, P = .026), unlike histological index and tissue calprotectin. CONCLUSIONS In patients with UC who have achieved clinical and endoscopic remission, tissue expression of NE is a better predictor of long-term relapse than histological activity.
Collapse
Affiliation(s)
- Yu Kyung Jun
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyeon Jeong Oh
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Ji Ae Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Yonghoon Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
4
|
Alphonse N, Sécher T, Heuzé-Vourc'h N. A breath of fresh air: inhaled antibodies to combat respiratory infectious diseases - a clinical trial overview. Expert Opin Drug Deliv 2025; 22:197-218. [PMID: 39711323 DOI: 10.1080/17425247.2024.2446608] [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: 09/18/2024] [Revised: 12/03/2024] [Accepted: 12/20/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION With the worldwide growing burden of respiratory tract infections (RTIs), innovative therapeutic approaches are in high demand. Inhaled antibodies (Abs) represent a promising avenue, offering targeted treatment options with potentially better therapeutic index compared to traditional delivery methods. AREAS COVERED This comprehensive review summarizes the challenges faced in delivering Abs by (intranasal and pulmonary) inhalation. It outlines the physiological and biological barriers encountered by inhaled drugs, as well as the influence of delivery devices and formulation on the deposition and efficacy of inhaled molecules. Moreover, it provides a detailed overview of the current clinical trial landscape of inhaled anti-RTI Abs, highlighting the progress in the development of inhaled Abs targeting a range of pathogens, such as severe acute respiratory syndrome coronavirus 2 and respiratory syncytial virus. The mechanism of action, therapeutic targets, and clinical outcomes of these novel therapies are detailed. EXPERT OPINION Delivery of Abs by inhalation faces several challenges. Addressing these challenges and developing specific approaches to deliver inhaled Abs represent a promising avenue for the development of the next generation of inhaled Abs. By offering targeted, localized therapy with the potential for a better therapeutic index, inhaled Abs could significantly improve outcomes for patients with RTIs.
Collapse
Affiliation(s)
- Noémie Alphonse
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires, Tours, France
| | - Thomas Sécher
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires, Tours, France
| | - Nathalie Heuzé-Vourc'h
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires, Tours, France
| |
Collapse
|
5
|
Wilton ZER, Jamus AN, Core SB, Frietze KM. Pathogenic and Protective Roles of Neutrophils in Chlamydia trachomatis Infection. Pathogens 2025; 14:112. [PMID: 40005489 PMCID: PMC11858174 DOI: 10.3390/pathogens14020112] [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: 12/13/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Chlamydia trachomatis (Ct) is an obligate intracellular pathogen that causes the most commonly diagnosed bacterial sexually transmitted infection (STI) and is a leading cause of preventable blindness globally. Ct infections can generate a strong pro-inflammatory immune response, leading to immune-mediated pathology in infected tissues. Neutrophils play an important role in mediating both pathology and protection during infection. Excessive neutrophil activation, migration, and survival are associated with host tissue damage during Chlamydia infections. In contrast, neutrophils also perform phagocytic killing of Chlamydia in the presence of IFN-γ and anti-Chlamydia antibodies. Neutrophil extracellular traps (NETs) and many neutrophil degranulation products have also demonstrated strong anti-Chlamydia functions. To counteract this neutrophil-mediated protection, Chlamydia has developed several evasion strategies. Various Chlamydia proteins can limit potentially protective neutrophil responses by directly targeting receptors present on the surface of neutrophils or neutrophil degranulation products. In this review, we provide a survey of current knowledge regarding the role of neutrophils in pathogenesis and protection, including the ways that Chlamydia circumvents neutrophil functions, and we propose critical areas for future research.
Collapse
Affiliation(s)
| | | | | | - Kathryn M. Frietze
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences, Albuquerque, NM 87131, USA
| |
Collapse
|
6
|
Obata‐Ninomiya K, Jayaraman T, Ziegler SF. From the bench to the clinic: basophils and type 2 epithelial cytokines of thymic stromal lymphopoietin and IL-33. Clin Transl Immunology 2024; 13:e70020. [PMID: 39654685 PMCID: PMC11626414 DOI: 10.1002/cti2.70020] [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: 03/26/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024] Open
Abstract
Type 2 epithelial cytokines, including thymic stromal lymphopoietin and IL-33, play central roles in modulation of type 2 immune cells, such as basophils. Basophils are a small subset of granulocytes within the leukocyte population that predominantly exist in the blood. They have non-redundant roles in allergic inflammation in peripheral tissues such as the lung, skin and gut, where they increase and accumulate at inflammatory lesions and exclusively produce large amounts of IL-4, a type 2 cytokine. These inflammatory reactions are known to be, to some extent, phenocopies of infectious diseases of ticks and helminths. Recently, biologics related to both type 2 epithelial cytokines and basophils have been approved by the US Food and Drug Administration for treatment of allergic diseases. We summarised the roles of Type 2 epithelial cytokines and basophils in basic science to translational medicine, including recent findings.
Collapse
Affiliation(s)
| | | | - Steven F Ziegler
- Center of Fundamental ImmunologyBenaroya Research InstituteSeattleWAUSA
- Department of ImmunologyUniversity of Washington School of MedicineSeattleWAUSA
| |
Collapse
|
7
|
Redeghieri P, Moray J, Kerff F, Gohy S, Leal T, Muyldermans S, Vanbever R, Morales‐Yánez FJ, Dumoulin M. Enzymatic, structural, and biophysical characterization of a single-domain antibody (VHH) selectively and tightly inhibiting neutrophil elastase and exhibiting favorable developability properties. Protein Sci 2024; 33:e5227. [PMID: 39604162 PMCID: PMC11602439 DOI: 10.1002/pro.5227] [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: 05/10/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
Human neutrophil elastase (hNE), a serine protease released by neutrophils during inflammation, plays a major role in the pathophysiology of several conditions especially in inflammatory lung diseases. Its inhibition constitutes, therefore, a promising therapeutic strategy to combat these diseases. In this work, we characterized the in vitro properties of a VHH (i.e., the antigen binding domain of camelid heavy chain-only antibodies), referred to as NbE201. This VHH is able to inhibit tightly, selectively and competitively both human and murine elastases with the inhibition constants (Ki) of 4.1 ± 0.9 nM and 36.8 ± 3.9 nM, respectively. The IC50 for the inhibition of the hydrolysis of elastin is in the same range to that of alpha-1 antitrypsin (i.e., the main endogenous inhibitor of hNE also used in the clinic) and 14 times better than that of Sivelestat (i.e., the 2nd clinically approved hNE inhibitor). The X-ray crystal structure of the NbE201-hNE complex reveals that the Complementarity Determining Regions CDR1 and CDR3 of the VHH bind into the substrate binding pocket of hNE and prevent the access to small or macromolecular substrates. They do not, however, bind deep enough into the pocket to be hydrolyzed. NbE201 is highly stable towards oxidation, deamidation, and chemical or thermal denaturation. NbE201 is therefore likely to tolerate manufacturing processes during drug development. These results highlight the high potential of NbE201 as a (pre)clinical tool to diagnose and treat diseases associated with excessive hNE activity, and for fundamental research to better understand the role of hNE in these conditions.
Collapse
Affiliation(s)
- Paola Redeghieri
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| | - Joël Moray
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| | - Frédéric Kerff
- Biological Macromolecule Crystallography, Centre for Protein Engineering, InBios, Department of Life SciencesUniversity of LiègeLiègeBelgium
| | - Sophie Gohy
- Department of Pneumology, ENT and Dermatology, Institute of Experimental and Clinical Research (IREC)Université Catholique de LouvainBrusselsBelgium
- Cystic Fibrosis Reference CentreCliniques Universitaires Saint‐LucBrusselsBelgium
| | - Teresinha Leal
- Louvain Center for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC)Université Catholique de LouvainBrusselsBelgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular ImmunologyVrije Universiteit BrusselBrusselsBelgium
| | - Rita Vanbever
- Louvain Drug Research Institute (LDRI)Université Catholique de LouvainBrusselsBelgium
| | - Francisco Javier Morales‐Yánez
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| | - Mireille Dumoulin
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| |
Collapse
|
8
|
Matsuo K, Ikemoto M, Okada K. Intraperitoneal Administration of S100A8 Ameliorates Experimental Acute Colitis in Rats. BIOLOGY 2024; 13:916. [PMID: 39596871 PMCID: PMC11592024 DOI: 10.3390/biology13110916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/01/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024]
Abstract
S100A8 is a protein that is abundant in neutrophils and macrophages (MΦ), but its role in inflammation remains unclear. This study aimed to assess the immunological role(s) of S100A8 in acute intestinal inflammation in rats and its role in MΦ. Rat recombinant S100A8 (rr-S100A8, 1.0 mg/kg) was intraperitoneally administered daily to rats with 3% dextran sulfate sodium (DSS) (DSS + A8 group)-induced experimental acute colitis. The histological severity score (6.50 ± 0.51, p = 0.038) in the DSS + A8 group rats remained lower than that (9.75 ± 1.48) of the rats without S100A8 (DSS group) administration. The tumor necrosis factor-alpha (TNF-α) production in the colon tissues of the rats in the DSS + A8 group (4.76 ± 0.90 pg/mL/g, p = 0.042) was significantly suppressed, compared with that of the DSS group (10.45 ± 2.04 pg/mL/g). To stimulate rat peritoneal MΦ, rr-S100A8, the anti-rat S100A8 antibody, and a lipopolysaccharide (LPS) were used in the in vitro experiments. In the MΦ stimulated with rr-S100A8 for 2 h, the mRNA level of intracellular S100A8 (47.41 ± 24.44, p = 0.002) increased in an autocrine manner, whereas that of S100A9 (0.24 ± 0.43, p = 0.782) was not significant. The TNF-α mRNA level in the MΦ treated with LPS and the anti-rat S100A8 antibody significantly increased (102.26 ± 18.60, p = 0.001) compared to that with LPS alone (16.9 ± 8.56). These results indicate that S100A8 can serve as an anti-inflammatory protein in acute inflammation by negatively regulating S100A9 and TNF-α production through inflammatory signaling pathways in MΦ.
Collapse
Affiliation(s)
- Kano Matsuo
- Graduate School of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
| | - Masaki Ikemoto
- Division of Clinical Immunology and Rheumatology, Department of Internal Medicine (IV), Faculty of Osaka Medical College, Osaka 569-8686, Japan
| | - Kohki Okada
- Department of Medical Technology and Sciences, Faculty of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
| |
Collapse
|
9
|
Vilardi A, Przyborski S, Mobbs C, Rufini A, Tufarelli C. Current understanding of the interplay between extracellular matrix remodelling and gut permeability in health and disease. Cell Death Discov 2024; 10:258. [PMID: 38802341 PMCID: PMC11130177 DOI: 10.1038/s41420-024-02015-1] [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: 01/18/2024] [Revised: 03/25/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
The intestinal wall represents an interactive network regulated by the intestinal epithelium, extracellular matrix (ECM) and mesenchymal compartment. Under healthy physiological conditions, the epithelium undergoes constant renewal and forms an integral and selective barrier. Following damage, the healthy epithelium is restored via a series of signalling pathways that result in remodelling of the scaffolding tissue through finely-regulated proteolysis of the ECM by proteases such as matrix metalloproteinases (MMPs). However, chronic inflammation of the gastrointestinal tract, as occurs in Inflammatory Bowel Disease (IBD), is associated with prolonged disruption of the epithelial barrier and persistent damage to the intestinal mucosa. Increased barrier permeability exhibits distinctive signatures of inflammatory, immunological and ECM components, accompanied by increased ECM proteolytic activity. This narrative review aims to bring together the current knowledge of the interplay between gut barrier, immune and ECM features in health and disease, discussing the role of barrier permeability as a discriminant between homoeostasis and IBD.
Collapse
Affiliation(s)
- Aurora Vilardi
- Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, United Kingdom
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham, DH1 3LE, United Kingdom
| | - Claire Mobbs
- Department of Biosciences, Durham University, Durham, DH1 3LE, United Kingdom
| | - Alessandro Rufini
- Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, United Kingdom.
- Department of Biosciences, University of Milan, Milan, 20133, Italy.
| | - Cristina Tufarelli
- Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, United Kingdom.
| |
Collapse
|
10
|
Awasthi D, Sarode A. Neutrophils at the Crossroads: Unraveling the Multifaceted Role in the Tumor Microenvironment. Int J Mol Sci 2024; 25:2929. [PMID: 38474175 DOI: 10.3390/ijms25052929] [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: 01/31/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Over the past decade, research has prominently established neutrophils as key contributors to the intricate landscape of tumor immune biology. As polymorphonuclear granulocytes within the innate immune system, neutrophils play a pivotal and abundant role, constituting approximately ∼70% of all peripheral leukocytes in humans and ∼10-20% in mice. This substantial presence positions them as the frontline defense against potential threats. Equipped with a diverse array of mechanisms, including reactive oxygen species (ROS) generation, degranulation, phagocytosis, and the formation of neutrophil extracellular traps (NETs), neutrophils undeniably serve as indispensable components of the innate immune system. While these innate functions enable neutrophils to interact with adaptive immune cells such as T, B, and NK cells, influencing their functions, they also engage in dynamic interactions with rapidly dividing tumor cells. Consequently, neutrophils are emerging as crucial regulators in both pro- and anti-tumor immunity. This comprehensive review delves into recent research to illuminate the multifaceted roles of neutrophils. It explores their diverse functions within the tumor microenvironment, shedding light on their heterogeneity and their impact on tumor recruitment, progression, and modulation. Additionally, the review underscores their potential anti-tumoral capabilities. Finally, it provides valuable insights into clinical therapies targeting neutrophils, presenting a promising approach to leveraging innate immunity for enhanced cancer treatment.
Collapse
Affiliation(s)
- Deepika Awasthi
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Aditya Sarode
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
11
|
Hou JJ, Ding L, Yang T, Yang YF, Jin YP, Zhang XP, Ma AH, Qin YH. The proteolytic activity in inflammatory bowel disease: insight from gut microbiota. Microb Pathog 2024; 188:106560. [PMID: 38272327 DOI: 10.1016/j.micpath.2024.106560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic, recurrent inflammatory disease caused by the destruction of the intestinal mucosal epithelium that affects a growing number of people worldwide. Although the etiology of IBD is complex and still elucidated, the role of dysbiosis and dysregulated proteolysis is well recognized. Various studies observed altered composition and diversity of gut microbiota, as well as increased proteolytic activity (PA) in serum, plasma, colonic mucosa, and fecal supernatant of IBD compared to healthy individuals. The imbalance of intestinal microecology and intestinal protein hydrolysis were gradually considered to be closely related to IBD. Notably, the pivotal role of intestinal microbiota in maintaining proteolytic balance received increasing attention. In summary, we have speculated a mesmerizing story, regarding the hidden role of PA and microbiota-derived PA hidden in IBD. Most importantly, we provided the diagnosis and therapeutic targets for IBD as well as the formulation of new treatment strategies for other digestive diseases and protease-related diseases.
Collapse
Affiliation(s)
- Jun-Jie Hou
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Liang Ding
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Tao Yang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yan-Fei Yang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yue-Ping Jin
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Xiao-Ping Zhang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - A-Huo Ma
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yue-Hua Qin
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China.
| |
Collapse
|
12
|
Blin T, Parent C, Pichon G, Guillon A, Jouan Y, Allouchi H, Aubrey N, Boursin F, Domain R, Korkmaz B, Sécher T, Heuzé-Vourc'h N. The proteolytic airway environment associated with pneumonia acts as a barrier for treatment with anti-infective antibodies. Eur J Pharm Biopharm 2024; 195:114163. [PMID: 38086491 DOI: 10.1016/j.ejpb.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 01/29/2024]
Abstract
Like pneumonia, coronavirus disease 2019 (COVID-19) is characterized by a massive infiltration of innate immune cells (such as polymorphonuclear leukocytes) into the airways and alveolar spaces. These cells release proteases that may degrade therapeutic antibodies and thus limit their effectiveness. Here, we investigated the in vitro and ex vivo impact on anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) IgG1s and other IgG subclasses (IgG2 and IgG4) of the neutrophil elastase, proteinase 3 and cathepsin G (the three main neutrophil serine proteases) found in endotracheal aspirates from patients with severe COVID-19. Although the IgGs were sensitive to neutrophil serine proteases, IgG2 was most resistant to proteolytic degradation. The two anti-SARS CoV2 antibodies (casirivimab and imdevimab) were sensitive to the lung's proteolytic environment, although neutrophil serine protease inhibitors only partly limited the degradation. Overall, our results show that the pneumonia-associated imbalance between proteases and their inhibitors in the airways contributes to degradation of antiviral antibodies.
Collapse
Affiliation(s)
- Timothée Blin
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France; Tours University Hospital (CHRU), Department of Pulmonary Medicine, Cystic Fibrosis Resource Center, F-37032 Tours, France
| | - Christelle Parent
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France
| | - Gabrielle Pichon
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France
| | - Antoine Guillon
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France; Tours University Hospital (CHRU), Critical Care Department, F-37032 Tours, France
| | - Youenn Jouan
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France; Tours University Hospital (CHRU), Cardiac Surgery Department, F-37032 Tours, France
| | - Hassan Allouchi
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France; Tours University Hospital (CHRU), Pharmacy Department, F-37032 Tours, France
| | - Nicolas Aubrey
- University of Tours, F-37032 Tours, France; UMR INRA ISP 1282, BioMap Team, F-37032 Tours, France
| | - Fanny Boursin
- University of Tours, F-37032 Tours, France; UMR INRA ISP 1282, BioMap Team, F-37032 Tours, France
| | - Roxane Domain
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France
| | - Baris Korkmaz
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France
| | - Thomas Sécher
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France
| | - Nathalie Heuzé-Vourc'h
- INSERM, Respiratory Disease Research Centre, U1100, F-37032 Tours, France; University of Tours, F-37032 Tours, France.
| |
Collapse
|
13
|
Pehrsson M, Domislovic V, Alexdottir MS, Brinar M, Karsdal MA, Barisic A, Krznaric Z, Mortensen JH. Blood-Based Biomarkers Reflecting Protease 3 and MMP-12 Catalyzed Elastin Degradation as Potential Noninvasive Surrogate Markers of Endoscopic and Clinical Disease in Inflammatory Bowel Disease. J Clin Med 2023; 13:21. [PMID: 38202027 PMCID: PMC10779348 DOI: 10.3390/jcm13010021] [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: 11/14/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Chronic inflammation in inflammatory bowel disease (IBD) triggers significant extracellular matrix remodeling, including elastin remodeling, leading to severe clinical complications. Novel methods to assess intestinal tissue destruction may act as surrogate markers of endoscopic disease activity, relieving patients of invasive endoscopy. We explored the noninvasive blood-based biomarkers ELP-3 and ELM-12, measuring elastin degradation in IBD. In a study involving 104 Crohn's disease (CD), 39 ulcerative colitis (UC), and 29 healthy donors, we assessed these biomarkers' association with endoscopic and clinical disease activity using ELISA. Patients were evaluated based on the SES-CD and CDAI for CD patients and modified MES and partial Mayo for UC patients. ELP-3 and ELM-12 were elevated in patients with IBD. Discerning CD patients in endoscopic remission and mild from moderate to severe, ELP-3 provided an AUC of 0.69 and ELM-12 an AUC of 0.73. The ELP-3 biomarker was associated with UC patients and provided the highest diagnostic power of 0.87 for remission vs. active clinical disease. The data suggest an association of ELP-3 with active CD and ELM-12 with endoscopic remission in CD patients. Additionally, ELP-3 could identify UC patients with active clinical disease from patients in remission. The noninvasive biomarkers ELP-3 and ELM-12 could be potential surrogate biomarkers of elastin degradation and endoscopic and clinical disease markers.
Collapse
Affiliation(s)
- Martin Pehrsson
- Biomarkers and Research, Nordic Bioscience A/S, 2730 Herlev, Denmark; (M.S.A.); (M.A.K.); (J.H.M.)
| | - Viktor Domislovic
- Department of Gastroenterology and Hepatology, University Hospital Center Zagreb, 10000 Zagreb, Croatia; (V.D.); (M.B.); (Z.K.)
| | | | - Marko Brinar
- Department of Gastroenterology and Hepatology, University Hospital Center Zagreb, 10000 Zagreb, Croatia; (V.D.); (M.B.); (Z.K.)
| | - Morten Asser Karsdal
- Biomarkers and Research, Nordic Bioscience A/S, 2730 Herlev, Denmark; (M.S.A.); (M.A.K.); (J.H.M.)
| | - Ana Barisic
- Center for Clinical Nutrition, University Hospital Centre Zagreb, 10000 Zagreb, Croatia;
| | - Zeljko Krznaric
- Department of Gastroenterology and Hepatology, University Hospital Center Zagreb, 10000 Zagreb, Croatia; (V.D.); (M.B.); (Z.K.)
| | - Joachim Høg Mortensen
- Biomarkers and Research, Nordic Bioscience A/S, 2730 Herlev, Denmark; (M.S.A.); (M.A.K.); (J.H.M.)
| |
Collapse
|
14
|
Riaz B, Sohn S. Neutrophils in Inflammatory Diseases: Unraveling the Impact of Their Derived Molecules and Heterogeneity. Cells 2023; 12:2621. [PMID: 37998356 PMCID: PMC10670008 DOI: 10.3390/cells12222621] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
Inflammatory diseases involve numerous disorders and medical conditions defined by an insufficient level of self-tolerance. These diseases evolve over the course of a multi-step process through which environmental variables play a crucial role in the emergence of aberrant innate and adaptive immunological responses. According to experimental data accumulated over the past decade, neutrophils play a significant role as effector cells in innate immunity. However, neutrophils are also involved in the progression of numerous diseases through participation in the onset and maintenance of immune-mediated dysregulation by releasing neutrophil-derived molecules and forming neutrophil extracellular traps, ultimately causing destruction of tissues. Additionally, neutrophils have a wide variety of functional heterogeneity with adverse effects on inflammatory diseases. However, the complicated role of neutrophil biology and its heterogeneity in inflammatory diseases remains unclear. Moreover, neutrophils are considered an intriguing target of interventional therapies due to their multifaceted role in a number of diseases. Several approaches have been developed to therapeutically target neutrophils, involving strategies to improve neutrophil function, with various compounds and inhibitors currently undergoing clinical trials, although challenges and contradictions in the field persist. This review outlines the current literature on roles of neutrophils, neutrophil-derived molecules, and neutrophil heterogeneity in the pathogenesis of autoimmune and inflammatory diseases with potential future therapeutic strategies.
Collapse
Affiliation(s)
- Bushra Riaz
- Department of Biomedical Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
| | - Seonghyang Sohn
- Department of Biomedical Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
15
|
Sécher T, Heuzé-Vourc'h N. Barriers for orally inhaled therapeutic antibodies. Expert Opin Drug Deliv 2023; 20:1071-1084. [PMID: 37609943 DOI: 10.1080/17425247.2023.2249821] [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: 05/12/2023] [Revised: 07/17/2023] [Accepted: 08/16/2023] [Indexed: 08/24/2023]
Abstract
INTRODUCTION Respiratory diseases represent a worldwide health issue. The recent Sars-CoV-2 pandemic, the burden of lung cancer, and inflammatory respiratory diseases urged the development of innovative therapeutic solutions. In this context, therapeutic antibodies (Abs) offer a tremendous opportunity to benefit patients with respiratory diseases. Delivering Ab through the airways has been demonstrated to be relevant to improve their therapeutic index. However, few inhaled Abs are on the market. AREAS COVERED This review describes the different barriers that may alter the fate of inhaled therapeutic Abs in the lungs at steady state. It addresses both physical and biological barriers and discusses the importance of taking into consideration the pathological changes occurring during respiratory disease, which may reinforce these barriers. EXPERT OPINION The pulmonary route remains rare for delivering therapeutic Abs, with few approved inhaled molecules, despite promising evidence. Efforts must focus on the intertwined barriers associated with lung diseases to develop appropriate Ab-formulation-device combo, ensuring optimal Ab deposition in the respiratory tract. Finally, randomized controlled clinical trials should be carried out to establish inhaled Ab therapy as prominent against respiratory diseases.
Collapse
Affiliation(s)
- Thomas Sécher
- INSERM, Centre d'Etude des Pathologies Respiratoires, Tours, France
- Université de Tours, Tours, France
| | - Nathalie Heuzé-Vourc'h
- INSERM, Centre d'Etude des Pathologies Respiratoires, Tours, France
- Université de Tours, Tours, France
| |
Collapse
|
16
|
Whaley KG, Xiong Y, Karns R, Hyams JS, Kugathasan S, Boyle BM, Walters TD, Kelsen J, LeLeiko N, Shapiro J, Waddell A, Fox S, Bezold R, Bruns S, Widing R, Haberman Y, Collins MH, Mizuno T, Minar P, D'Haens GR, Denson LA, Vinks AA, Rosen MJ. Multicenter Cohort Study of Infliximab Pharmacokinetics and Therapy Response in Pediatric Acute Severe Ulcerative Colitis. Clin Gastroenterol Hepatol 2023; 21:1338-1347. [PMID: 36031093 PMCID: PMC9968822 DOI: 10.1016/j.cgh.2022.08.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/02/2022] [Accepted: 08/15/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS We aimed to model infliximab (IFX) pharmacokinetics (PK) in pediatric acute severe ulcerative colitis (ASUC) and assess the association between PK parameters, including drug exposure, and clinical response. METHODS We studied a multicenter prospective cohort of hospitalized children initiating IFX for ASUC or IBD-unclassified. Serial IFX serum concentrations over 26 weeks were used to develop a PK model. We tested the association of PK parameter estimates with day 7 clinical response, week 8 clinical remission, week 26 corticosteroid-free clinical remission (CSF-CR) (using the Pediatric Ulcerative Colitis Activity Index), and colectomy-free survival. RESULTS Thirty-eight participants received IFX (median initial dose, 9.9 mg/kg). Day 7 clinical response, week 8 clinical remission, and week 26 CSF-CR occurred in 71%, 55%, and 43%, respectively. Albumin, C-reactive protein, white blood cell count, platelets, weight, and antibodies to IFX were significant covariates incorporated into a PK model. Week 26 non-remitters exhibited faster IFX clearance than remitters (P = .013). However, cumulative IFX exposure did not differ between clinical response groups. One (2.7%) and 4 (10.8%) participants underwent colectomy by week 26 and 2 years, respectively. Day 3 IFX clearance >0.02 L/h was associated with colectomy (hazard ratio, 58.2; 95% confidence interval, 6.0-568.6; P < .001). CONCLUSIONS At median higher-than-label IFX dosing for pediatric ASUC, baseline faster IFX CL was associated with colectomy and at week 26 with lack of CSF-CR. IFX exposure was not predictive of clinical outcomes. Higher IFX dosing may sufficiently optimize early outcomes in pediatric ASUC. Larger studies are warranted to determine whether sustained intensification can overcome rapid clearance and improve later outcomes. CLINICALTRIALS gov identifier: NCT02799615.
Collapse
Affiliation(s)
- Kaitlin G Whaley
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ye Xiong
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Rebekah Karns
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jeffrey S Hyams
- Division of Digestive Diseases, Hepatology, Nutrition, Connecticut Children's Medical Center, Hartford, Connecticut
| | - Subra Kugathasan
- Division of Pediatric Gastroenterology, Emory University School of Medicine, Atlanta, Georgia
| | - Brendan M Boyle
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Nationwide Children's Hospital, Columbus, Ohio
| | - Thomas D Walters
- Division of Pediatric Gastroenterology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Judith Kelsen
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Neal LeLeiko
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Morgan Stanley Children's Hospital, New York, New York
| | - Jason Shapiro
- IBD Center, Department of Pediatrics, Hasbro Children's Hospital, Providence, Rhode Island
| | - Amanda Waddell
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sejal Fox
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ramona Bezold
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stephanie Bruns
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Robin Widing
- Office for Clinical and Translational Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Yael Haberman
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Sheba Medical Center, Tel Hashomer, affiliated with the Tel Aviv University, Israel
| | - Margaret H Collins
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Tomoyuki Mizuno
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Phillip Minar
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Geert R D'Haens
- Department of Gastroenterology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Lee A Denson
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Alexander A Vinks
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michael J Rosen
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
17
|
Azcutia V, Kelm M, Fink D, Cummings RD, Nusrat A, Parkos CA, Brazil JC. Sialylation regulates neutrophil transepithelial migration, CD11b/CD18 activation, and intestinal mucosal inflammatory function. JCI Insight 2023; 8:e167151. [PMID: 36719745 PMCID: PMC10077474 DOI: 10.1172/jci.insight.167151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
Polymorphonuclear neutrophils (PMNs) play a critical role in clearing invading microbes and promoting tissue repair following infection/injury. However, dysregulated PMN trafficking and associated tissue damage is pathognomonic of numerous inflammatory mucosal diseases. The final step in PMN influx into mucosal lined organs (including the lungs, kidneys, skin, and gut) involves transepithelial migration (TEpM). The β2-integrin CD11b/CD18 plays an important role in mediating PMN intestinal trafficking, with recent studies highlighting that terminal fucose and GlcNAc glycans on CD11b/CD18 can be targeted to reduce TEpM. However, the role of the most abundant terminal glycan, sialic acid (Sia), in regulating PMN epithelial influx and mucosal inflammatory function is not well understood. Here we demonstrate that inhibiting sialidase-mediated removal of α2-3-linked Sia from CD11b/CD18 inhibits PMN migration across intestinal epithelium in vitro and in vivo. Sialylation was also found to regulate critical PMN inflammatory effector functions, including degranulation and superoxide release. Finally, we demonstrate that sialidase inhibition reduces bacterial peptide-mediated CD11b/CD18 activation in PMN and blocks downstream intracellular signaling mediated by spleen tyrosine kinase (Syk) and p38 MAPK. These findings suggest that sialylated glycans on CD11b/CD18 represent potentially novel targets for ameliorating PMN-mediated tissue destruction in inflammatory mucosal diseases.
Collapse
Affiliation(s)
- Veronica Azcutia
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthias Kelm
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Dylan Fink
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard D. Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Charles A. Parkos
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer C. Brazil
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
18
|
Deraison C, Bonnart C, Langella P, Roget K, Vergnolle N. Elafin and its precursor trappin-2: What is their therapeutic potential for intestinal diseases? Br J Pharmacol 2023; 180:144-160. [PMID: 36355635 PMCID: PMC10098471 DOI: 10.1111/bph.15985] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 04/22/2022] [Accepted: 05/07/2022] [Indexed: 11/12/2022] Open
Abstract
Elafin and its precursor trappin-2 are known for their contribution to the physiological mucosal shield against luminal microbes. Such a contribution seems to be particularly relevant in the gut, where the exposure of host tissues to heavy loads of microbes is constant and contributes to mucosa-associated pathologies. The expression of trappin-2/elafin has been shown to be differentially regulated in diseases associated with gut inflammation. Accumulating evidence has demonstrated the protective effects of trappin-2/elafin in gut intestinal disorders associated with acute or chronic inflammation, or with gluten sensitization disorders. The protective effects of trappin-2/elafin in the gut are discussed in terms of their pleiotropic modes of action: acting as protease inhibitors, transglutaminase substrates, antimicrobial peptides or as a regulator of pro-inflammatory transcription factors. Further, the question of the therapeutic potential of trappin-2/elafin delivery at the intestinal mucosa surface is raised. Whether trappin-2/elafin mucosal delivery should be considered to ensure intestinal tissue repair is also discussed.
Collapse
Affiliation(s)
- Céline Deraison
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Chrystelle Bonnart
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Philippe Langella
- Université Paris-Saclay, AgroParisTech, Micalis Institute, INRAE, Jouy-en-Josas, France
| | | | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France.,Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
19
|
Cao D, Qian K, Zhao Y, Hong J, Chen H, Wang X, Yang N, Zhang C, Cao J, Jia K, Wu G, Zhu M, Shen J, Zhang Y, Cui Z, Wang Z. Association of neutrophil extracellular traps with fistula healing in patients with complex perianal fistulising Crohn's disease. J Crohns Colitis 2022; 17:580-592. [PMID: 36322703 DOI: 10.1093/ecco-jcc/jjac171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Perianal fistulising Crohn's disease (pfCD) is a disabling phenotype of Crohn's disease (CD) with suboptimal outcomes. We assessed neutrophil extracellular traps (NETs) in perianal fistulas and implicated their roles in pfCD healing. METHODS Patients with complex pfCD who developed preplaced seton drainage were recruited during the verified maintenance of remission in CD. Fistula tracts were sampled during definitive surgery plus seton removal. Patient demographics, CD classification, medication strategy, and healing of pfCD were recorded. RNA sequencing was applied for transcriptomic profile analysis. NETs components, including myeloperoxidase (MPO), neutrophil elastase (NE), and citrullinated histone H3 (CitH3), were identified using immunofluorescence. Serum infliximab (IFX), anti-IFX antibodies, and tissue levels of IFX, adalimumab (ADA), MPO and CitH3 were determined using enzyme-linked immunosorbent assays. Peptidyl arginine deiminase IV (PAD4), tumour necrosis factor (TNF)-α, and NE were detected using immunohistochemistry. Gene expression levels of PAD family members were assessed with qPCR. RESULTS Twenty-one patients were included, 15 of whom adopted IFX as maintenance treatment. RNA-seq revealed difference in neutrophil associated pathways between unhealed and healed fistulas. NETs components (MPO/NE/CitH3) were detectable in the fistulas and were parallel with the PAD4 levels. Eleven of 21 (52%) patients experienced complete healing of the pfCD 108 weeks post-operatively. Fistula NETs were significantly increased in patients with unhealed pfCD. Increased NETs were associated with abundant TNF-α production and the absence of IFX in fistulas. CONCLUSIONS NETs exist in pfCD fistulas, which are associated with unhealed post-operative fistulas in pfCD, suggesting their prognostic roles in pfCD.
Collapse
Affiliation(s)
- Dongxing Cao
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Keyu Qian
- Laboratory of Medicine, Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200444, China
| | - Ying Zhao
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai 200001, China
| | - Jie Hong
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai 200001, China
| | - Haoyan Chen
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai 200001, China
| | - Xiaohui Wang
- Department of General Surgery, Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200444, China
| | - Nailin Yang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Cheng Zhang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jingkai Cao
- Department of General Surgery, Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200444, China
| | - Keyu Jia
- Laboratory of Medicine, Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200444, China
| | - Guangyu Wu
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200444, China
| | - Mingming Zhu
- Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Ren Ji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Ye Zhang
- Laboratory of Medicine, Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200444, China
| | - Zhe Cui
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; Department of General Surgery, Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.1058, Huan Zhen Bei Rd, Baoshan District, Shanghai 200444, China
| | - Zheng Wang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
20
|
Takahashi T, Kato S, Ito J, Shimizu N, Parida IS, Itaya-Takahashi M, Sakaino M, Imagi J, Yoshinaga K, Yoshinaga-Kiriake A, Gotoh N, Ikeda I, Nakagawa K. Dietary triacylglycerol hydroperoxide is not absorbed, yet it induces the formation of other triacylglycerol hydroperoxides in the gastrointestinal tract. Redox Biol 2022; 57:102471. [PMID: 36137475 PMCID: PMC9493066 DOI: 10.1016/j.redox.2022.102471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/07/2022] [Indexed: 11/07/2022] Open
Abstract
The in vivo presence of triacylglycerol hydroperoxide (TGOOH), a primary oxidation product of triacylglycerol (TG), has been speculated to be involved in various diseases. Thus, considerable attention has been paid to whether dietary TGOOH is absorbed from the intestine. In this study, we performed the lymph duct-cannulation study in rats and analyzed the level of TGOOH in lymph following administration of a TG emulsion containing TGOOH. As we successfully detected TGOOH from the lymph, we hypothesized that this might be originated from the intestinal absorption of dietary TGOOH [hypothesis I] and/or the in situ formation of TGOOH [hypothesis II]. To determine the validity of these hypotheses, we then performed another cannulation study using a TG emulsion containing a deuterium-labeled TGOOH (D2-TGOOH) that is traceable in vivo. After administration of this emulsion to rats, we clearly detected unlabeled TGOOH instead of D2-TGOOH from the lymph, indicating that TGOOH is not absorbed from the intestine but is more likely to be produced in situ. By discriminating the isomeric structures of TGOOH present in lymph, we predicted the mechanism by which the intake of dietary TGOOH triggers oxidative stress (e.g., via generation of singlet oxygen) and induces in situ formation of TGOOH. The results of this study hereby provide a foothold to better understand the physiological significance of TGOOH on human health.
Collapse
Affiliation(s)
- Takumi Takahashi
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Shunji Kato
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan; J-Oil Mills Innovation Laboratory, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Junya Ito
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Naoki Shimizu
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Isabella Supardi Parida
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Mayuko Itaya-Takahashi
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Masayoshi Sakaino
- J-Oil Mills Innovation Laboratory, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Jun Imagi
- J-Oil Mills Innovation Laboratory, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Kazuaki Yoshinaga
- Faculty of Food and Agricultural Sciences, Fukushima University, Fukushima, Japan
| | - Aya Yoshinaga-Kiriake
- Department of Life Science, Graduate School of Engineering Science, Akita University, Akita, Japan
| | - Naohiro Gotoh
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, Tokyo, Japan
| | - Ikuo Ikeda
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Kiyotaka Nakagawa
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan; J-Oil Mills Innovation Laboratory, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan.
| |
Collapse
|
21
|
Neutrophils in Intestinal Inflammation: What We Know and What We Could Expect for the Near Future. GASTROINTESTINAL DISORDERS 2022. [DOI: 10.3390/gidisord4040025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Neutrophils are short-lived cells that play a crucial role in inflammation. As in other tissues, these polymorphonuclear phagocytes are involved in the intestinal inflammatory response, on the one hand, contributing to the activation and recruitment of other immune cells, but on the other hand, facilitating intestinal mucosa repair by releasing mediators that aid in the resolution of inflammation. Even though these responses are helpful in physiological conditions, excessive recruitment of activated neutrophils in the gut correlates with increased mucosal damage and severe symptoms in patients with inflammatory bowel disease (IBD) and pre-clinical models of colitis. Thus, there is growing interest in controlling their biology to generate novel therapeutic approaches capable of reducing exacerbated intestinal inflammation. However, the beneficial and harmful effects of neutrophils on intestinal inflammation are still controversial. With this review, we summarise and discuss the most updated literature showing how neutrophils (and neutrophil extracellular traps) contribute to developing and resolving intestinal inflammation and their putative use as therapeutic targets.
Collapse
|
22
|
Xie Y, Fontenot L, Chupina Estrada A, Nelson B, Wang J, Shih DQ, Ho W, Mattai SA, Rieder F, Jensen DD, Bunnett NW, Koon HW. Elafin Reverses Intestinal Fibrosis by Inhibiting Cathepsin S-Mediated Protease-Activated Receptor 2. Cell Mol Gastroenterol Hepatol 2022; 14:841-876. [PMID: 35840034 PMCID: PMC9425040 DOI: 10.1016/j.jcmgh.2022.06.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 12/10/2022]
Abstract
BACKGROUND & AIMS More than half of Crohn's disease patients develop intestinal fibrosis-induced intestinal strictures. Elafin is a human protease inhibitor that is down-regulated in the stricturing intestine of Crohn's disease patients. We investigated the efficacy of elafin in reversing intestinal fibrosis and elucidated its mechanism of action. METHODS We developed a new method to mimic a stricturing Crohn's disease environment and induce fibrogenesis using stricturing Crohn's disease patient-derived serum exosomes to condition fresh human intestinal tissues and primary stricturing Crohn's disease patient-derived intestinal fibroblasts. Three mouse models of intestinal fibrosis, including SAMP1/YitFc mice, Salmonella-infected mice, and trinitrobenzene sulfonic acid-treated mice, were also studied. Elafin-Eudragit FS30D formulation and elafin-overexpressing construct and lentivirus were used. RESULTS Elafin reversed collagen synthesis in human intestinal tissues and fibroblasts pretreated with Crohn's disease patient-derived serum exosomes. Proteome arrays identified cathepsin S as a novel fibroblast-derived pro-fibrogenic protease. Elafin directly suppressed cathepsin S activity to inhibit protease-activated receptor 2 activity and Zinc finger E-box-binding homeobox 1 expression, leading to reduced collagen expression in intestinal fibroblasts. Elafin overexpression reversed ileal fibrosis in SAMP1/YitFc mice, cecal fibrosis in Salmonella-infected mice, and colonic fibrosis in trinitrobenzene sulfonic acid-treated mice. Cathepsin S, protease-activated receptor 2 agonist, and zinc finger E-box-binding homeobox 1 overexpression abolished the anti-fibrogenic effect of elafin in fibroblasts and all 3 mouse models of intestinal fibrosis. Oral elafin-Eudragit FS30D treatment abolished colonic fibrosis in trinitrobenzene sulfonic acid-treated mice. CONCLUSIONS Elafin suppresses collagen synthesis in intestinal fibroblasts via cathepsin S-dependent protease-activated receptor 2 inhibition and decreases zinc finger E-box-binding homeobox 1 expression. The reduced collagen synthesis leads to the reversal of intestinal fibrosis. Thus, modified elafin may be a therapeutic approach for intestinal fibrosis.
Collapse
Affiliation(s)
- Ying Xie
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California,Department of Gastroenterology, The First Hospital of China Medical University, Shenyang City, Liaoning Province, China
| | - Lindsey Fontenot
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Andrea Chupina Estrada
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Becca Nelson
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Jiani Wang
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California,Department of Gastroenterology, The First Hospital of China Medical University, Shenyang City, Liaoning Province, China
| | - David Q. Shih
- F. Widjaja Foundation, Inflammatory Bowel & Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Wendy Ho
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - S. Anjani Mattai
- Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Florian Rieder
- Department of Gastroenterology, Hepatology, and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Dane D. Jensen
- Bluestone Center for Clinical Research, New York University College of Dentistry, New York, New York
| | - Nigel W. Bunnett
- Department of Molecular Pathobiology, Department of Neuroscience and Physiology, Neuroscience Institute, New York University, New York, New York
| | - Hon Wai Koon
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California.
| |
Collapse
|
23
|
Preclinical evaluation of [11C]GW457427 as a tracer for neutrophil elastase. Nucl Med Biol 2022; 106-107:62-71. [DOI: 10.1016/j.nucmedbio.2022.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/22/2021] [Accepted: 01/03/2022] [Indexed: 11/19/2022]
|
24
|
Jacenik D, Fichna J, Małecka-Wojciesko E, Mokrowiecka A. Protease-Activated Receptors - Key Regulators of Inflammatory Bowel Diseases Progression. J Inflamm Res 2022; 14:7487-7497. [PMID: 35002281 PMCID: PMC8721023 DOI: 10.2147/jir.s335502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis and course of inflammatory bowel diseases are related to both immune system disorders and dysfunction of colon permeability. Moreover, co-existing diseases in patients with Crohn's disease and ulcerative colitis are identified. Currently, there are some therapeutic strategies that affect the function of cytokine/s causing inflammation in the intestinal wall. However, additional approaches which target other components of inflammatory bowel diseases pathogenesis are still needed. Accumulating evidence suggests that proteases and protease-activated receptors seem to be responsible for colitis progression. Experimental and observational studies showed alteration of protease-activated receptors expression in the colon of patients with Crohn's disease and ulcerative colitis. Furthermore, it was suggested that the expression of protease-activated receptors correlated with inflammatory bowel diseases activity. Moreover, regulation of protease-activated receptors seems to be responsible for the modulation of colitis and clinical manifestation of inflammatory bowel diseases. In this review, we present the current state of knowledge about the contribution of protease-activated receptors to Crohn's disease and ulcerative colitis and its implications for diagnosis and treatment.
Collapse
Affiliation(s)
- Damian Jacenik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Ewa Małecka-Wojciesko
- Department of Digestive Tract Diseases, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Anna Mokrowiecka
- Department of Digestive Tract Diseases, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
25
|
Craig CF, Filippone RT, Stavely R, Bornstein JC, Apostolopoulos V, Nurgali K. Neuroinflammation as an etiological trigger for depression comorbid with inflammatory bowel disease. J Neuroinflammation 2022; 19:4. [PMID: 34983592 PMCID: PMC8729103 DOI: 10.1186/s12974-021-02354-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/14/2021] [Indexed: 02/06/2023] Open
Abstract
Patients with inflammatory bowel disease (IBD) suffer from depression at higher rates than the general population. An etiological trigger of depressive symptoms is theorised to be inflammation within the central nervous system. It is believed that heightened intestinal inflammation and dysfunction of the enteric nervous system (ENS) contribute to impaired intestinal permeability, which facilitates the translocation of intestinal enterotoxins into the blood circulation. Consequently, these may compromise the immunological and physiological functioning of distant non-intestinal tissues such as the brain. In vivo models of colitis provide evidence of increased blood–brain barrier permeability and enhanced central nervous system (CNS) immune activity triggered by intestinal enterotoxins and blood-borne inflammatory mediators. Understanding the immunological, physiological, and structural changes associated with IBD and neuroinflammation may aid in the development of more tailored and suitable pharmaceutical treatment for IBD-associated depression.
Collapse
Affiliation(s)
- Colin F Craig
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia
| | - Rhiannon T Filippone
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia
| | - Rhian Stavely
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia.,Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Joel C Bornstein
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Australia
| | - Vasso Apostolopoulos
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia.,Immunology Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia
| | - Kulmira Nurgali
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia. .,Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia. .,Regenerative Medicine and Stem Cells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia. .,Institute for Health and Sport, Victoria University, Level 4 Research Labs, Western Centre for Health Research and Education, Sunshine Hospital, 176 Furlong Road, St Albans, VIC, 3021, Australia.
| |
Collapse
|
26
|
Al Ojaimi Y, Blin T, Lamamy J, Gracia M, Pitiot A, Denevault-Sabourin C, Joubert N, Pouget JP, Gouilleux-Gruart V, Heuzé-Vourc'h N, Lanznaster D, Poty S, Sécher T. Therapeutic antibodies - natural and pathological barriers and strategies to overcome them. Pharmacol Ther 2021; 233:108022. [PMID: 34687769 PMCID: PMC8527648 DOI: 10.1016/j.pharmthera.2021.108022] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 02/06/2023]
Abstract
Antibody-based therapeutics have become a major class of therapeutics with over 120 recombinant antibodies approved or under review in the EU or US. This therapeutic class has experienced a remarkable expansion with an expected acceleration in 2021-2022 due to the extraordinary global response to SARS-CoV2 pandemic and the public disclosure of over a hundred anti-SARS-CoV2 antibodies. Mainly delivered intravenously, alternative delivery routes have emerged to improve antibody therapeutic index and patient comfort. A major hurdle for antibody delivery and efficacy as well as the development of alternative administration routes, is to understand the different natural and pathological barriers that antibodies face as soon as they enter the body up to the moment they bind to their target antigen. In this review, we discuss the well-known and more under-investigated extracellular and cellular barriers faced by antibodies. We also discuss some of the strategies developed in the recent years to overcome these barriers and increase antibody delivery to its site of action. A better understanding of the biological barriers that antibodies have to face will allow the optimization of antibody delivery near its target. This opens the way to the development of improved therapy with less systemic side effects and increased patients' adherence to the treatment.
Collapse
Affiliation(s)
- Yara Al Ojaimi
- UMR 1253, iBrain, Inserm, 37000 Tours, France; University of Tours, 37000 Tours, France
| | - Timothée Blin
- University of Tours, 37000 Tours, France; UMR 1100, CEPR, Inserm, 37000 Tours, France
| | - Juliette Lamamy
- University of Tours, 37000 Tours, France; GICC, EA7501, 37000 Tours, France
| | - Matthieu Gracia
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier F-34298, France
| | - Aubin Pitiot
- University of Tours, 37000 Tours, France; UMR 1100, CEPR, Inserm, 37000 Tours, France
| | | | - Nicolas Joubert
- University of Tours, 37000 Tours, France; GICC, EA7501, 37000 Tours, France
| | - Jean-Pierre Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier F-34298, France
| | | | | | - Débora Lanznaster
- UMR 1253, iBrain, Inserm, 37000 Tours, France; University of Tours, 37000 Tours, France
| | - Sophie Poty
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier F-34298, France
| | - Thomas Sécher
- University of Tours, 37000 Tours, France; UMR 1100, CEPR, Inserm, 37000 Tours, France
| |
Collapse
|
27
|
Dos Santos Ramos A, Viana GCS, de Macedo Brigido M, Almeida JF. Neutrophil extracellular traps in inflammatory bowel diseases: Implications in pathogenesis and therapeutic targets. Pharmacol Res 2021; 171:105779. [PMID: 34298111 DOI: 10.1016/j.phrs.2021.105779] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/04/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are the two main forms of inflammatory bowel disease (IBD). Among the various immune cells involved in IBD, neutrophils are the first to infiltrate and appear to contribute to the impairment of the epithelial barrier, destruction of tissues by oxidative and proteolytic damage, as well as to the perpetuation of inflammation by the release of cytokines and chemokines associated with pro-inflammatory effects. In addition to basic effector mechanisms, such as phagocytosis and chemotaxis, neutrophils can also form extracellular traps (NETs), which is made up of a mesh-like structure - which contains its chromatin (DNA + histones) together with granules and enzymes, such as myeloperoxidase (MPO) and neutrophilic elastase (NE) - and that acts as a trap that can result in the death of extracellular pathogens and/or can promote tissue damage. Recent evidence indicates that NETs also play an important and significant role in the pathogenesis of IBD. Previous studies have reported increased levels of NETs in tissue and serum samples from patients with IBD, as well as in experimental colitis. In this review, we discuss current knowledge about the formation of NETs and their role in the pathophysiology of IBD, pointing out potential mechanisms by which NETs promote tissue damage, as well as their involvement in complications associated with IBD. In addition, we propose potential targets for therapy to regulate the production of NETs, making it possible to expand the current spectrum of therapies for IBD.
Collapse
Affiliation(s)
- Anderson Dos Santos Ramos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil.
| | | | | | - Juliana Franco Almeida
- Department of Cellular Biology, University of Brasilia, Brasilia, Brazil; Department of Cellular and Molecular Biology, Federal University of Paraíba, Paraíba, Brazil.
| |
Collapse
|
28
|
Interplay between Extracellular Matrix and Neutrophils in Diseases. J Immunol Res 2021; 2021:8243378. [PMID: 34327245 PMCID: PMC8302397 DOI: 10.1155/2021/8243378] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/03/2021] [Indexed: 12/17/2022] Open
Abstract
The extracellular matrix (ECM) is a highly dynamic and complex network structure, which exists in almost all tissues and is the microenvironment that cells rely on for survival. ECM interacts with cells to regulate diverse functions, including differentiation, proliferation, and migration. Neutrophils are the most abundant immune cells in circulation and play key roles in orchestrating a complex series of events during inflammation. Neutrophils can also mediate ECM remodeling by providing specific matrix-remodeling enzymes (such as neutrophil elastase and metalloproteinases), generating neutrophil extracellular traps, and releasing exosomes. In turn, ECM can remodel the inflammatory microenvironment by regulating the function of neutrophils, which drives disease progression. Both the presence of ECM and the interplay between neutrophils and their extracellular matrices are considered an important and outstanding mechanistic aspect of inflammation. In this review, the importance of ECM will be considered, together with the discussion of recent advances in understanding the underlying mechanisms of the intricate interplay between ECM and neutrophils. A better comprehension of immune cell-matrix reciprocal dependence has exciting implications for the development of new therapeutic options for neutrophil-associated infectious and inflammatory diseases.
Collapse
|
29
|
Dudzińska E, Strachecka A, Gil-Kulik P, Kocki J, Bogucki J, Shemedyuk N, Gryzinska M. Influence of the Treatment Used in Inflammatory Bowel Disease on the Protease Activities. Int J Gen Med 2020; 13:1633-1642. [PMID: 33380821 PMCID: PMC7767739 DOI: 10.2147/ijgm.s267036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/10/2020] [Indexed: 11/24/2022] Open
Abstract
Introduction There is growing evidence that intestinal proteases have a role in the pathogenesis of gastrointestinal inflammatory diseases. Inflammatory bowel disease (IBD), which includes Crohn’s disease (CD) and ulcerative colitis (UC), has an additional source of proteases represented by infiltrated and activated inflammatory cells. The aim of our study was to determine proteolytic system activity in patients with CD and UC. We limited the number of proteases tested by determining proteases active in acidic, neutral and alkaline pH. Materials and Methods The study included 40 patients with IBD – 20 CD patients and 20 UC patients. The control group consisted of 20 healthy subjects. Among the 20 CD patients, 17 were treated with aminosalicylates, 14 with azathioprine, and 4 with corticosteroids, while 8 patients were undergoing biological treatment. Among the 20 UC patients, 19 were treated with aminosalicylates, 8 with azathioprine, and 3 with corticosteroids. The total protein concentration was assayed by the Lowry method. The optimal pH was assayed in pH from 2.2 to 12.8, separated by 0.2 intervals. Proteolytic activities were determined against different substrates (gelatine, haemoglobin, ovalbumin, albumin, cytochrome C, and casein), and haemoglobin was the optimal substrate. Protease activities were determined according to Anson method. Determination of the activities of natural inhibitors of acidic, neutral and alkaline proteases is based on the Lee and Lin method. Results Decreases were observed in the activities of acid proteases (pH 5), alkaline proteases (pH 7), and neutral proteases (pH 7.6 and 8.6) in the groups of CD patients in remission in comparison with the active phase. In the group of patients with biologically treated CD patients, acid protease activity (pH 5.0) was lower than in CD patients not receiving biological treatment. Activities of neutral (pH 7.0) and alkaline (pH 7.6 and 8.6) proteases in the plasma of patients with UC in remission were lower in comparison to the active phase. Activities of acid (pH 5.0) and alkaline (8.6) protease inhibitors were higher in CD patients in the active phase in comparison to remission. In UC patients with exacerbation of the disease, the activity of alkaline (pH 8.6) protease inhibitors was increased compared to remission. Conclusion 1. Our research may suggest that the immunomodulatory treatment used in IBD, aimed at reducing the level of leukocytes and reduction of inflammation, may contribute to a reduction in protease activity. 2. The decrease of protease activities in patients with CD and UC in remission may be a marker suggesting the patients’ response to the treatment.
Collapse
Affiliation(s)
- Ewa Dudzińska
- Chair of Public Health, Medical University of Lublin, Lublin 20-093, Poland
| | - Aneta Strachecka
- Subdepartment of General and Molecular Genetics, Institute of Biological Basis of Animal Production, University of Life Sciences in Lublin, Lublin 20-950, Poland
| | - Paulina Gil-Kulik
- Chair of Medical Genetics, Department of Clinical Genetics, Medical University of Lublin, Lublin 20-080, Poland
| | - Janusz Kocki
- Chair of Medical Genetics, Department of Clinical Genetics, Medical University of Lublin, Lublin 20-080, Poland
| | - Jacek Bogucki
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University, Lublin 20-093, Poland
| | - Natalya Shemedyuk
- Department Biotechnology and Radiology, Stepan Gzhytskyi National University of Veterinary Medicine and Biotechnologies, Lviv 79010, Ukraine
| | - Magdalena Gryzinska
- Subdepartment of General and Molecular Genetics, Institute of Biological Basis of Animal Production, University of Life Sciences in Lublin, Lublin 20-950, Poland
| |
Collapse
|