1
|
Qapaja T, Abu-Rumaileh M, Alsabbagh Alchirazi K, Gharaibeh A, Naser A, Hamid O, Alayan D, Regueiro M. Biologics and Oral Small Molecules Are Not Associated With Increased Major Adverse Cardiovascular Events or Venous Thromboembolism in Inflammatory Bowel Disease. Inflamm Bowel Dis 2024:izae267. [PMID: 39536156 DOI: 10.1093/ibd/izae267] [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: 06/02/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Biologics and oral small molecules (OSM) effectively treat inflammatory bowel disease (IBD), but some are linked to higher risks of major adverse cardiovascular events (MACE) and venous thromboembolism (VTE). This study evaluates the MACE and VTE risks in IBD patients treated with biologics or OSM. METHODS Using the TrinNetX multi-institutional database, we examined MACE and VTE in adult IBD patients on biologics and compared them to IBD patients not on biologics. We also compared IBD patients on OSM to those not on OSM. We performed 1:1 propensity score matching. MACE (myocardial infarction [MI], stroke, and all-cause mortality) and VTE were assessed from 30 days to 3 years after drug prescription. RESULTS After matching, IBD patients on biologics had reduced risk of MI, stroke, and all-cause mortality at 1 year, compared to those not on biologics (P < .05). No significant difference in VTE was observed (P = .5). At 3 years, biologic-treated patients had lower risks of MI, stroke, all-cause mortality, and VTE (P < .05). Inflammatory bowel disease patients on OSM showed no significant differences in MI, stroke, or VTE at 1 and 3 years, but had lower all-cause mortality (P < .05). In older IBD patients with at least 1 cardiovascular risk factor, OSM usage showed no significant difference in MI, stroke, or VTE risk compared to nonusers; however, all-cause mortality was decreased at 3 years (P < .05). CONCLUSIONS Inflammatory bowel disease patients treated with biologics or OSM were not at increased risk of MACE or VTE. Although further studies and longer follow-up periods are needed to confirm these findings, our results provide reassurance regarding the safety of these medications in IBD.
Collapse
Affiliation(s)
- Thabet Qapaja
- Division of Hospital Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | - Ahmad Gharaibeh
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Ahmad Naser
- Department of Internal Medicine, Jacobi Medical Center, New York, NY, USA
| | - Osama Hamid
- Department of Gastroenterology, University of Texas Southwestern, Dallas, TX, USA
| | - Dina Alayan
- Department of Pulmonary and Critical Care, MetroHealth Medical Center, Cleveland, OH, USA
| | - Miguel Regueiro
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
2
|
Alipouriani A, Erozkan K, Schabl L, Sancheti H, Sebastian S, Wong SY, Tozer P, Cohen BL, Holubar SD. TOpClass Class 4 Perineal Crohn's Disease: A Systematic Review and Meta-analysis of Perineal Wound Complication After Proctectomy in Crohn's Patients. Inflamm Bowel Dis 2024:izae198. [PMID: 39418126 DOI: 10.1093/ibd/izae198] [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: 05/11/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Nonhealing perineal wounds have been reported to be common after proctectomy for Crohn's disease (CD). We performed a systematic review and meta-analysis of perineal wound healing after proctectomy for CD and assessed the risk factors for nonhealing. METHODS A comprehensive literature search was conducted in PubMed, Embase, and Scopus databases from 2010 to 2023, and articles reporting perineal wound healing rates after proctectomy for CD were included. Data on study characteristics and proportion of healed wounds, and risk factors, were extracted. Random-effects meta-analysis was performed to estimate the pooled proportion and 95% CIs using the "meta" package in R. Heterogeneity was assessed using the I2 statistic. RESULTS We identified 501 articles, of which 252 remained after de-duplication. After screening, 4 retrospective cohort studies involving 333 patients were included. Across the 4 studies, the pooled proportion of completely healed perineal wounds at 6 months was 65% (95% CI 52%-80%), and 70% (95% CI 60%-83%) at 12 months. Significant heterogeneity was found between studies (I2 = 86% at 6 months). Three studies examined risk factors for impaired healing after proctectomy. One study identified preoperative perineal sepsis as the only independent factor associated with impaired healing (P = .001) on multivariable analysis. In 1 study, male sex, shorter time from diversion to proctectomy, and higher preoperative C-reactive protein levels were all associated with delayed healing in univariate analysis. Another study found that close rectal dissection was associated with significantly lower healing rates than total mesorectal excision (P = .01). Prior use of tumor necrosis factor inhibitors was not associated with wound healing outcomes. CONCLUSIONS This meta-analysis revealed complete perineal healing in only 70% of patients 12 months after proctectomy for CD. This highlights knowledge gaps, including the identification of modifiable risk factors and methods for preventing or as rescue therapy, such as vacuum-assisted closure and flap reconstruction, for nonhealing perineal wounds after proctectomy for CD. Poor perineal wound healing outcomes are likely related to imperfectly understood underlying inflammatory dysregulation and systemically impaired wound healing in patients with CD.
Collapse
Affiliation(s)
- Ali Alipouriani
- Department of Colon and Rectal Surgery, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Kamil Erozkan
- Department of Colon and Rectal Surgery, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Lucas Schabl
- Department of Colon and Rectal Surgery, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Himani Sancheti
- Department of Colon and Rectal Surgery, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Shaji Sebastian
- Department of Gastroenterology, IBD Unit, Hull University Teaching Hospitals, Hull, UK
| | - Serre-Yu Wong
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Phil Tozer
- Department of Colon and Rectal Surgery, St. Mark's Hospital, London, UK
| | - Benjamin L Cohen
- Department of Gastroenterology and Hepatology, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Stefan D Holubar
- Department of Colon and Rectal Surgery, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
3
|
Suri C, Pande B, Sahu T, Sahithi LS, Verma HK. Revolutionizing Gastrointestinal Disorder Management: Cutting-Edge Advances and Future Prospects. J Clin Med 2024; 13:3977. [PMID: 38999541 PMCID: PMC11242723 DOI: 10.3390/jcm13133977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/22/2024] [Accepted: 06/29/2024] [Indexed: 07/14/2024] Open
Abstract
In recent years, remarkable strides have been made in the management of gastrointestinal disorders, transforming the landscape of patient care and outcomes. This article explores the latest breakthroughs in the field, encompassing innovative diagnostic techniques, personalized treatment approaches, and novel therapeutic interventions. Additionally, this article emphasizes the use of precision medicine tailored to individual genetic and microbiome profiles, and the application of artificial intelligence in disease prediction and monitoring. This review highlights the dynamic progress in managing conditions such as inflammatory bowel disease, gastroesophageal reflux disease, irritable bowel syndrome, and gastrointestinal cancers. By delving into these advancements, we offer a glimpse into the promising future of gastroenterology, where multidisciplinary collaborations and cutting-edge technologies converge to provide more effective, patient-centric solutions for individuals grappling with gastrointestinal disorders.
Collapse
Affiliation(s)
- Chahat Suri
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
- Lung Health and Immunity, Helmholtz Zentrum Munich, IngolstädterLandstraße 1, 85764 Oberschleißheim, 85764 Munich, Germany
| | - Babita Pande
- Department of Physiology, All India Institute of Medical Science, Raipur 492099, India
| | - Tarun Sahu
- Department of Physiology, All India Institute of Medical Science, Raipur 492099, India
| | | | - Henu Kumar Verma
- Lung Health and Immunity, Helmholtz Zentrum Munich, IngolstädterLandstraße 1, 85764 Oberschleißheim, 85764 Munich, Germany
| |
Collapse
|
4
|
Yang Y, Zhao C, Yang Z, Du C, Chang Z, Wen X, Zhang X, Liu Y, Hu L, Gao Z. Myeloid-derived growth factor ameliorates dextran sodium sulfate-induced colitis by regulating macrophage polarization. J Mol Med (Berl) 2024; 102:875-886. [PMID: 38695882 PMCID: PMC11213757 DOI: 10.1007/s00109-024-02447-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/19/2024] [Accepted: 04/15/2024] [Indexed: 06/29/2024]
Abstract
Inflammatory bowel disease (IBD) is characterized by inflammatory conditions in the gastrointestinal tract. According to reports, IBD prevalence is increasing globally, with heavy economic and physical burdens. Current IBD clinical treatment is limited to pharmacological methods; therefore, new strategies are needed. Myeloid-derived growth factor (MYDGF) secreted by bone marrow-derived mononuclear macrophages has beneficial effects in multiple inflammatory diseases. To this end, the present study aimed to establish an experimental IBD mouse model using dextran sulfate sodium in drinking water. MYDGF significantly alleviated DSS-induced colitis, suppressed lymphocyte infiltration, restored epithelial integrity in mice, and decreased apoptosis in the colon tissue. Moreover, the number of M1 macrophages was decreased and that of M2 macrophages was increased by the action of MYDGF. In MYDGF-treated mice, the NF-κB and MAPK pathways were partially inhibited. Our findings indicate that MYDGF could mitigate DSS-induced mice IBD by reducing inflammation and restoring epithelial integrity through regulation of intestinal macrophage polarization via NF-κB and MAPK pathway inhibition. KEY MESSAGES: MYDGF alleviated DSS-induced acute colitis. MYDGF maintains colon epithelial barrier integrity and relieves inflammation. MYDGF regulates colon macrophage polarization. MYDGF partially inhibited the activation of NF-κB and MAPK pathway.
Collapse
Affiliation(s)
- Yang Yang
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Conghui Zhao
- Department of Pathology, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Zi Yang
- Department of Endodontics, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Conglin Du
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology and Beijing Laboratory of Oral Health, Beijing, 100050, China
| | - Zhichao Chang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology and Beijing Laboratory of Oral Health, Beijing, 100050, China
| | - Xin Wen
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology and Beijing Laboratory of Oral Health, Beijing, 100050, China
| | - Xiujuan Zhang
- Nephrology Department, Zhucheng People's Hospital, Shandong, 262200, China
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, 100050, China.
| | - Liang Hu
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
| | - Zhenhua Gao
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
5
|
Nguyen OTP, Misun PM, Hierlemann A, Lohasz C. A Versatile Intestine-on-Chip System for Deciphering the Immunopathogenesis of Inflammatory Bowel Disease. Adv Healthc Mater 2024; 13:e2302454. [PMID: 38253407 PMCID: PMC11468350 DOI: 10.1002/adhm.202302454] [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: 07/30/2023] [Revised: 12/21/2023] [Indexed: 01/24/2024]
Abstract
The multifactorial nature of inflammatory bowel disease (IBD) necessitates reliable and practical experimental models to elucidate its etiology and pathogenesis. To model the intestinal microenvironment at the onset of IBD in vitro, it is important to incorporate relevant cellular and noncellular components before inducing stepwise pathogenic developments. A novel intestine-on-chip system for investigating multiple aspects of IBD's immunopathogenesis is presented. The system includes an array of tight and polarized barrier models formed from intestinal epithelial cells on an in-vivo-like subepithelial matrix within one week. The dynamic remodeling of the subepithelial matrix by cells or their secretome demonstrates the physiological relevance of the on-chip barrier models. The system design enables introduction of various immune cell types and inflammatory stimuli at specific locations in the same barrier model, which facilitates investigations of the distinct roles of each cell type in intestinal inflammation development. It is showed that inflammatory behavior manifests in an upregulated expression of inflammatory markers and cytokines (TNF-α). The neutralizing effect of the anti-inflammatory antibody Infliximab on levels of TNF-α and its inducible cytokines could be explicitly shown. Overall, an innovative approach to systematically developing a microphysiological system to comprehend immune-system-mediated disorders of IBD and to identify new therapeutic strategies is presented.
Collapse
Affiliation(s)
- Oanh T. P. Nguyen
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Patrick M. Misun
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Andreas Hierlemann
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Christian Lohasz
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| |
Collapse
|
6
|
Chen M, Lan H, Jin K, Chen Y. Responsive nanosystems for targeted therapy of ulcerative colitis: Current practices and future perspectives. Drug Deliv 2023; 30:2219427. [PMID: 37288799 PMCID: PMC10405869 DOI: 10.1080/10717544.2023.2219427] [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: 01/30/2023] [Revised: 05/15/2023] [Accepted: 05/20/2023] [Indexed: 06/09/2023] Open
Abstract
The pharmacological approach to treating gastrointestinal diseases is suffering from various challenges. Among such gastrointestinal diseases, ulcerative colitis manifests inflammation at the colon site specifically. Patients suffering from ulcerative colitis notably exhibit thin mucus layers that offer increased permeability for the attacking pathogens. In the majority of ulcerative colitis patients, the conventional treatment options fail in controlling the symptoms of the disease leading to distressing effects on the quality of life. Such a scenario is due to the failure of conventional therapies to target the loaded moiety into specific diseased sites in the colon. Targeted carriers are needed to address this issue and enhance the drug effects. Conventional nanocarriers are mostly readily cleared and have nonspecific targeting. To accumulate the desired concentration of the therapeutic candidates at the inflamed area of the colon, smart nanomaterials with responsive nature have been explored recently that include pH responsive, reactive oxygen species responsive (ROS), enzyme responsive and thermo - responsive smart nanocarrier systems. The formulation of such responsive smart nanocarriers from nanotechnology scaffolds has resulted in the selective release of therapeutic drugs, avoiding systemic absorption and limiting the undesired delivery of targeting drugs into healthy tissues. Recent advancements in the field of responsive nanocarrier systems have resulted in the fabrication of multi-responsive systems i.e. dual responsive nanocarriers and derivitization that has increased the biological tissues and smart nanocarrier's interaction. In addition, it has also led to efficient targeting and significant cellular uptake of the therapeutic moieties. Herein, we have highlighted the latest status of the responsive nanocarrier drug delivery system, its applications for on-demand delivery of drug candidates for ulcerative colitis, and the prospects are underpinned.
Collapse
Affiliation(s)
- Min Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yun Chen
- Department of Colorectal Surgery, Xinchang People’s Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang, Zhejiang, China
| |
Collapse
|
7
|
Ibing S, Cho JH, Böttinger EP, Ungaro RC. Second-Line Biologic Therapy Following Tumor Necrosis Factor Antagonist Failure: A Real-World Propensity Score-Weighted Analysis. Clin Gastroenterol Hepatol 2023; 21:2629-2638. [PMID: 36787837 PMCID: PMC11531069 DOI: 10.1016/j.cgh.2023.01.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 01/05/2023] [Accepted: 01/31/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND& AIMS Tumor necrosis factor (TNF) antagonists often are used as first-line medications to treat moderate to severe inflammatory bowel disease (IBD), but many patients do not achieve or maintain response. Our aim was to compare the effectiveness of second-line treatments (ustekinumab, vedolizumab, or a second TNF antagonist) after TNF antagonist exposure in patients with Crohn's disease (CD) and ulcerative colitis (UC) from 2 electronic health records-based cohorts. METHODS We identified patients with prior TNF antagonist exposure who switched to a different biologic in the Mount Sinai Health System (MSHS) electronic health records (CD, n = 527; UC, n = 165) and the Study of a Prospective Adult Research Cohort (SPARC) from the Inflammatory Bowel Disease Plexus Program of the Crohn's & Colitis Foundation (CD, n = 412; UC, n = 129). Treatment failure was defined as the composite of any IBD-related surgery, IBD-related hospitalization, new prescription of oral/intravenous corticosteroids, or need to switch to a third biologic agent. Time-to-event analysis was conducted with inverse probability of treatment-weighted data. RESULTS Overall, treatment failure occurred in 85% of MSHS and 72% of SPARC CD patients. In SPARC, the likelihood of treatment failure was significantly lower with ustekinumab compared with vedolizumab as second-line treatment (adjusted hazard ratio, 0.66; 95% CI, 0.54-0.82; P < .001), a trend confirmed in MSHS (adjusted hazard ratio, 0.89; 95% CI, 0.77-1.04; P = .15). In both cohorts, the superiority of ustekinumab compared with vedolizumab was shown when considering treatment failure as prescription of steroids or a third biologic agent. In UC, no differences between second-line treatment groups were identified. CONCLUSIONS In 2 independent real-world cohort settings, second-line therapy in CD with ustekinumab after TNF antagonist treatment failure was associated with a lower likelihood of treatment failure than second-line vedolizumab.
Collapse
Affiliation(s)
- Susanne Ibing
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, New York; Digital Health Center, Hasso Plattner Institute, University of Potsdam, Potsdam, Germany
| | - Judy H Cho
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Erwin P Böttinger
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, New York; Digital Health Center, Hasso Plattner Institute, University of Potsdam, Potsdam, Germany
| | - Ryan C Ungaro
- The Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
8
|
Roco-Videla Á, Villota-Arcos C, Pino-Astorga C, Mendoza-Puga D, Bittner-Ortega M, Corbeaux-Ascui T. Intermittent Fasting and Reduction of Inflammatory Response in a Patient with Ulcerative Colitis. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1453. [PMID: 37629743 PMCID: PMC10456230 DOI: 10.3390/medicina59081453] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023]
Abstract
Ulcerative colitis is an inflammatory disease that affects the colon, generating a crisis period associated with diarrhea and ulcerations. Stress plays a pivotal role in modulating the inflammatory response and aggravating progression. Different studies have shown that fasting reduces inflammation markers, and intermittent fasting decreases inflammatory markers such as IL-2, IL-6, and RCP. Goal: To evaluate the impact of intermittent fasting on a patient diagnosed with ulcerative colitis. A female patient underwent intermittent fasting (10/14) for eight weeks. Clinical tests were performed for blood count, RCP, biochemical profile, glycemia, and T4/TSH levels. Fecal calprotectin was determined. Clinical exams were assessed before and after intermittent fasting. Inflammation markers, such as CRP and calprotectin, were significantly reduced after eight weeks of intermittent fasting. The patient reported feeling better and was seizure-free during the following months when she continued fasting intermittently. Intermittent fasting allowed for a reduction in inflammation markers.
Collapse
Affiliation(s)
- Ángel Roco-Videla
- Facultad de Salud y Ciencias Sociales, Universidad de las Américas, Providencias, Santiago 7500975, Chile
| | - Claudio Villota-Arcos
- Escuela de Nutrición y Dietética, Facultad de Ciencias de la Salud, Universidad Bernardo O’Higgins, Santiago 8370993, Chile;
- Laboratorio de Microbiologia y Biotecnologia Oral, Facultad de Ciencias de la Vida, Universidad Andres Bello, Echaurren 237, Piso 6, Santiago 8370133, Chile;
| | - Carolina Pino-Astorga
- Escuela de Nutrición y Dietética, Facultad de Ciencias de la Salud, Universidad Bernardo O’Higgins, Santiago 8370993, Chile;
| | - Daniela Mendoza-Puga
- Integramédica, Parte de BUPA, Director Médico ITR-IBB-ITA, Cerro Colorado 5240, Las Condes, Santiago 7560995, Chile;
| | - Mauricio Bittner-Ortega
- Laboratorio de Microbiologia y Biotecnologia Oral, Facultad de Ciencias de la Vida, Universidad Andres Bello, Echaurren 237, Piso 6, Santiago 8370133, Chile;
| | | |
Collapse
|
9
|
Zhu Y, Wang Y, Xia G, Zhang X, Deng S, Zhao X, Xu Y, Chang G, Tao Y, Li M, Li H, Huang X, Chan HF. Oral Delivery of Bioactive Glass-Loaded Core-Shell Hydrogel Microspheres for Effective Treatment of Inflammatory Bowel Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207418. [PMID: 37092589 PMCID: PMC10288274 DOI: 10.1002/advs.202207418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/13/2023] [Indexed: 05/03/2023]
Abstract
Resolving inflammation and promoting intestinal tissue regeneration are critical for inflammatory bowel disease (IBD) treatment. Bioactive glass (BG) is a clinically approved bone graft material and has been shown to modulate inflammatory response, but it is unknown whether BG can be applied to treat IBD. Here, it is reported that BG attenuates pro-inflammatory response of lipopolysaccharide (LPS)-stimulated macrophages and hence reduces inflammatory damage to intestinal organoids in vitro. In addition, zein/sodium alginate-based core-shell microspheres (Zein/SA/BG) are developed for oral delivery of BG, which helps prevent premature dissolution of BG in the stomach. The results show that Zein/SA/BG protects BG from a gastric-simulated environment while dissolved in an intestinal-simulated environment. When administered to acute and chronic colitis mice model, Zein/SA/BG significantly reduces intestinal inflammation, promotes epithelial tissue regeneration, and partially restores microbiota homeostasis. These findings are the first to reveal the therapeutic efficacy of BG against IBD, which may provide a new therapeutic approach at low cost for effective IBD treatment.
Collapse
Affiliation(s)
- Yanlun Zhu
- Key Laboratory for Regenerative Medicine of the Ministry of Education of ChinaSchool of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SAR999077China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongShatinHong Kong SAR999077China
| | - Yiwei Wang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine600 Yishan RdShanghai200233China
| | - Guanggai Xia
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine600 Yishan RdShanghai200233China
| | - Xuerao Zhang
- Key Laboratory for Regenerative Medicine of the Ministry of Education of ChinaSchool of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SAR999077China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongShatinHong Kong SAR999077China
| | - Shuai Deng
- Key Laboratory for Regenerative Medicine of the Ministry of Education of ChinaSchool of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SAR999077China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongShatinHong Kong SAR999077China
- Cell Therapy and Cell Drugs of Luzhou Key LaboratorySchool of PharmacySouthwest Medical UniversityLuzhouSichuan646000China
| | - Xiaoyu Zhao
- Key Laboratory for Regenerative Medicine of the Ministry of Education of ChinaSchool of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SAR999077China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongShatinHong Kong SAR999077China
| | - Yanteng Xu
- Laboratory of Biomaterials and Translational MedicineCenter for NanomedicineThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Guozhu Chang
- Key Laboratory for Regenerative Medicine of the Ministry of Education of ChinaSchool of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SAR999077China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongShatinHong Kong SAR999077China
| | - Yu Tao
- Laboratory of Biomaterials and Translational MedicineCenter for NanomedicineThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational MedicineCenter for NanomedicineThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
- Guangdong Provincial Key Laboratory of Liver DiseaseGuangzhou510630China
| | - Haiyan Li
- Chemical and Environmental EngineeringSchool of EngineeringRMIT University124 La Trobe StMelbourneVIC3000Australia
| | - Xinyu Huang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine600 Yishan RdShanghai200233China
| | - Hon Fai Chan
- Key Laboratory for Regenerative Medicine of the Ministry of Education of ChinaSchool of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SAR999077China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongShatinHong Kong SAR999077China
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics999077Hong Kong SARChina
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkHong Kong SAR999077China
| |
Collapse
|
10
|
Pagnini C, Di Paolo MC, Urgesi R, Pallotta L, Fanello G, Graziani MG, Delle Fave G. Safety and Potential Role of Lactobacillus rhamnosus GG Administration as Monotherapy in Ulcerative Colitis Patients with Mild-Moderate Clinical Activity. Microorganisms 2023; 11:1381. [PMID: 37374884 DOI: 10.3390/microorganisms11061381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/30/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Probiotics are microorganisms that confer benefits to the host, and, for this reason, they have been proposed in several pathologic states. Specifically, probiotic bacteria have been investigated as a therapeutic option in ulcerative colitis (UC) patients, but clinical results are dishomogeneous. In particular, many probiotic species with different therapeutic schemes have been proposed, but no study has investigated probiotics in monotherapy in adequate trials for the induction of remission. Lactobacillus rhamnosus GG (LGG) is the more intensively studied probiotic and it has ideal characteristics for utilization in UC patients. The aim of the present study is to investigate the clinical efficacy and safety of LGG administration in an open trial, delivered in monotherapy at two different doses, in UC patients with mild-moderate disease. The UC patients with mild-moderate disease activity (Partial Mayo score ≥ 2) despite treatment with oral mesalamine were included. The patients stopped oral mesalamine and were followed up for one month, then were randomized to receive LGG supplement at dose of 1.2 or 2.4 × 1010 CFU/day for one month. At the end of the study, the clinical activity was evaluated and compared to that at the study entrance (efficacy). Adverse events were recorded (safety). The primary end-point was clinical improvement (reduction in the Partial Mayo score) and no serious adverse events, while the secondary end-points were the evaluation of different efficacies and safeties between the two doses of LGG. The patients with disease flares dropped out of the study and went back to standard therapy. The efficacy data were analyzed in an intention-to-treat (ITT) and per-protocol (PP) analysis. Out of the 76 patients included in the study, 75 started the probiotic therapy (n = 38 and 37 per group). In the ITT analysis, 32/76 (42%) responded to treatment, 21/76 (28%) remained stable, and 23/76 (30%) had a worsening of their clinical condition; 55 (72%) completed the treatment and were analyzed in a PP analysis: 32/55 (58%) had a clinical response, 21 (38%) remained stable, and 2 (4%) had a light worsening of their clinical condition (p < 0.0001). Overall, 37% of the patients had a disease remission. No severe adverse event was recorded, and only one patient stopped therapy due to obstinate constipation. No difference in the clinical efficacy and safety has been recorded between groups treated with different doses of LGG. The present prospective clinical trial demonstrates, for the first time, that LGG in monotherapy is safe and effective for the induction of remission in UC patients with mild-moderate disease activity (ClinicalTrials.gov identifier: NCT04102852).
Collapse
Affiliation(s)
- Cristiano Pagnini
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Maria Carla Di Paolo
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Riccardo Urgesi
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Lorella Pallotta
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Gianfranco Fanello
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Maria Giovanna Graziani
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Gianfranco Delle Fave
- Department of Gastroenterology, "Sapienza" University of Rome, 00185 Rome, Italy
- Onlus "S. Andrea", 00199 Rome, Italy
| |
Collapse
|
11
|
Michaels M, Madsen KL. Immunometabolism and microbial metabolites at the gut barrier: Lessons for therapeutic intervention in inflammatory bowel disease. Mucosal Immunol 2023; 16:72-85. [PMID: 36642380 DOI: 10.1016/j.mucimm.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 01/15/2023]
Abstract
The concept of immunometabolism has emerged recently whereby the repolarizing of inflammatory immune cells toward anti-inflammatory profiles by manipulating cellular metabolism represents a new potential therapeutic approach to controlling inflammation. Metabolic pathways in immune cells are tightly regulated to maintain immune homeostasis and appropriate functional specificity. Because effector and regulatory immune cell populations have different metabolic requirements, this allows for cellular selectivity when regulating immune responses based on metabolic pathways. Gut microbes have a major role in modulating immune cell metabolic profiles and functional responses through extensive interactions involving metabolic products and crosstalk between gut microbes, intestinal epithelial cells, and mucosal immune cells. Developing strategies to target metabolic pathways in mucosal immune cells through the modulation of gut microbial metabolism has the potential for new therapeutic approaches for human autoimmune and inflammatory diseases, such as inflammatory bowel disease. This review will give an overview of the relationship between metabolic reprogramming and immune responses, how microbial metabolites influence these interactions, and how these pathways could be harnessed in the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Margret Michaels
- University of Alberta, Department of Medicine, Edmonton, Alberta, Canada
| | - Karen L Madsen
- University of Alberta, Department of Medicine, Edmonton, Alberta, Canada; IMPACTT: Integrated Microbiome Platforms for Advancing Causation Testing & Translation, Edmonton, Alberta, Canada.
| |
Collapse
|
12
|
Tavares EDA, Guerra GCB, da Costa Melo NM, Dantas-Medeiros R, da Silva ECS, Andrade AWL, de Souza Araújo DF, da Silva VC, Zanatta AC, de Carvalho TG, de Araújo AA, de Araújo-Júnior RF, Zucolotto SM. Toxicity and Anti-Inflammatory Activity of Phenolic-Rich Extract from Nopalea cochenillifera (Cactaceae): A Preclinical Study on the Prevention of Inflammatory Bowel Diseases. PLANTS (BASEL, SWITZERLAND) 2023; 12:594. [PMID: 36771677 PMCID: PMC9921826 DOI: 10.3390/plants12030594] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/04/2023] [Accepted: 01/11/2023] [Indexed: 06/18/2023]
Abstract
Phenolic compounds have been scientifically recognized as beneficial to intestinal health. The cactus Nopalea cochenillifera, used as anti-inflammatory in traditional medicine, is a rich source of these bioactive compounds. The present study aimed to investigate the phytochemical profile of N. cochenillifera extract and evaluate its acute toxicity and anti-inflammatory effect on 2,4-dinitrobenzenesulfonic acid (DNBS)-induced colitis in rats. The total phenolic content per gram of dry extract was 67.85 mg. Through HPLC-IES-MSn, a total of 25 compounds such as saccharides, organic acids, phenolic acids and flavonoids were characterized. The dose of 2000 mg/kg of extract by an oral route showed no signs of toxicity, mortality or significant changes in biochemical and hematological parameters. Regarding intestinal anti-inflammatory effects, animals were treated with three different doses of extract or sulfasalazine. Macroscopic analysis of the colon indicated that the extract decreased the disease activity index. Levels of IL-1β and TNF-α decreased, IL-10 increased and MDA and MPO enzyme levels decreased when compared with the control group. In addition, a down-regulation of MAPK1/ERK2 and NF-κB p65 pathway markers in colon tissue was observed. The epithelial integrity was improved according to histopathological and immunohistological analysis. Thus, the extract provided strong preclinical evidence of being effective in maintaining the remission of colitis.
Collapse
Affiliation(s)
- Emanuella de Aragão Tavares
- Graduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Gerlane Coelho Bernardo Guerra
- Graduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
- Department of Biophysics and Pharmacology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
- Graduate Program in Pharmaceutical Science, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, Brazil
| | - Nadja Maria da Costa Melo
- Graduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Renato Dantas-Medeiros
- Graduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | | | - Anderson Wilbur Lopes Andrade
- Graduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | | | - Valéria Costa da Silva
- Graduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Ana Caroline Zanatta
- Department of Biomolecular Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto, São Paulo University, São Paulo, Ribeirão Preto 14040-903, Brazil
| | - Thaís Gomes de Carvalho
- Program Degree in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, Brazil
| | - Aurigena Antunes de Araújo
- Department of Biophysics and Pharmacology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
- Graduate Program in Pharmaceutical Science, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, Brazil
- Program Degree in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, Brazil
| | - Raimundo Fernandes de Araújo-Júnior
- Graduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
- Program Degree in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, Brazil
- Cancer and Inflammation Research Laboratory, Morphology Department, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Silvana Maria Zucolotto
- Graduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
- Graduate Program in Pharmaceutical Science, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, Brazil
| |
Collapse
|
13
|
Astorga J, Gasaly N, Dubois-Camacho K, De la Fuente M, Landskron G, Faber KN, Urra FA, Hermoso MA. The role of cholesterol and mitochondrial bioenergetics in activation of the inflammasome in IBD. Front Immunol 2022; 13:1028953. [PMID: 36466902 PMCID: PMC9716353 DOI: 10.3389/fimmu.2022.1028953] [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] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/26/2022] [Indexed: 10/15/2023] Open
Abstract
Inflammatory Bowel Disease (IBD) is characterized by a loss of intestinal barrier function caused by an aberrant interaction between the immune response and the gut microbiota. In IBD, imbalance in cholesterol homeostasis and mitochondrial bioenergetics have been identified as essential events for activating the inflammasome-mediated response. Mitochondrial alterations, such as reduced respiratory complex activities and reduced production of tricarboxylic acid (TCA) cycle intermediates (e.g., citric acid, fumarate, isocitric acid, malate, pyruvate, and succinate) have been described in in vitro and clinical studies. Under inflammatory conditions, mitochondrial architecture in intestinal epithelial cells is dysmorphic, with cristae destruction and high dynamin-related protein 1 (DRP1)-dependent fission. Likewise, these alterations in mitochondrial morphology and bioenergetics promote metabolic shifts towards glycolysis and down-regulation of antioxidant Nuclear erythroid 2-related factor 2 (Nrf2)/Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) signaling. Although the mechanisms underlying the mitochondrial dysfunction during mucosal inflammation are not fully understood at present, metabolic intermediates and cholesterol may act as signals activating the NLRP3 inflammasome in IBD. Notably, dietary phytochemicals exhibit protective effects against cholesterol imbalance and mitochondrial function alterations to maintain gastrointestinal mucosal renewal in vitro and in vivo conditions. Here, we discuss the role of cholesterol and mitochondrial metabolism in IBD, highlighting the therapeutic potential of dietary phytochemicals, restoring intestinal metabolism and function.
Collapse
Affiliation(s)
- Jessica Astorga
- Laboratory of Innate Immunity, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Naschla Gasaly
- Laboratory of Innate Immunity, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, Groningen, Netherlands
| | - Karen Dubois-Camacho
- Laboratory of Innate Immunity, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Metabolic Plasticity and Bioenergetics, Program of Molecular and Clinical Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Marjorie De la Fuente
- Laboratory of Biomedicine Research, School of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Glauben Landskron
- Laboratory of Biomedicine Research, School of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, Groningen, Netherlands
| | - Félix A. Urra
- Laboratory of Metabolic Plasticity and Bioenergetics, Program of Molecular and Clinical Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Marcela A. Hermoso
- Laboratory of Innate Immunity, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
14
|
Uchiyama K, Takagi T, Mizushima K, Asaeda K, Kajiwara M, Kashiwagi S, Minagawa Y, Hotta Y, Tanaka M, Inoue K, Dohi O, Okayama T, Yoshida N, Katada K, Kamada K, Ishikawa T, Yasuda H, Konishi H, Kishimoto M, Naito Y, Itoh Y. Investigation on the Inhibitory Effect of Wnt-5a on Colonic Mucosal Inflammation in Patients with Ulcerative Colitis. Dig Dis Sci 2022; 67:4760-4769. [PMID: 35590045 DOI: 10.1007/s10620-022-07537-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 03/23/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Recent progress in ulcerative colitis (UC) treatment has been remarkable, and various medications have been applied. However, some patients with UC are refractory to treatment and convert to surgery. AIM To investigate the role of colonic mucosal Wnt-5a expression in the pathogenesis of UC and the effect of bioactive Wnt-5a peptide on colitis in mice. METHODS Wnt-5a peptide was intraperitoneally administered to mice every day from the beginning of dextran sulfate sodium (DSS) treatment. The severity of colitis was evaluated based on body weight change, colonic length, and histological scores. Colonic mucosal TNF-α and KC mRNA expression levels were measured. This study included 70 patients with UC in clinical remission. Wnt-5a, TNFα, and IL-8 mRNA expression in the rectal mucosa were measured by quantitative real-time polymerase chain reaction using biopsy materials. Wnt-5a mRNA expression levels were compared between patients who relapsed and those in remission. We examined the correlation of Wnt-5a expression with TNF-α and IL-8 expression. RESULTS Wnt-5a peptide significantly attenuated the severity of DSS-induced colitis. Moreover, mucosal TNF-α and KC mRNA expression were significantly suppressed by Wnt-5a peptide treatment. Wnt-5a mRNA levels were significantly lower in patients with subsequent relapse than in those who remained in remission. Mucosal Wnt-5a was inversely correlated with TNF α and IL-8 expression. CONCLUSION Wnt-5a peptide suppressed colitis in mice, and decreased Wnt-5a expression was strongly associated with relapse in patients with UC. Wnt-5a may have an inhibitory effect on mucosal inflammation in UC, and Wnt-5a peptide could be a new therapeutic strategy.
Collapse
Affiliation(s)
- Kazuhiko Uchiyama
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Tomohisa Takagi
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan.
- Department for Medical Innovation and Translational Medical Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan.
| | - Katsura Mizushima
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kohei Asaeda
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Mariko Kajiwara
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Saori Kashiwagi
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yuki Minagawa
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yuma Hotta
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Makoto Tanaka
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Ken Inoue
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Osamu Dohi
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Tetsuya Okayama
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Naohisa Yoshida
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kazuhiro Katada
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kazuhiro Kamada
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hiroaki Yasuda
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hideyuki Konishi
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Mitsuo Kishimoto
- Department of Surgical Pathology, Kyoto City Hospital, Kyoto, 604-8845, Japan
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
15
|
Yasmin F, Sahito AM, Mir SL, Khatri G, Shaikh S, Gul A, Hassan SA, Koritala T, Surani S. Electrical neuromodulation therapy for inflammatory bowel disease. World J Gastrointest Pathophysiol 2022; 13:128-142. [PMID: 36187600 PMCID: PMC9516456 DOI: 10.4291/wjgp.v13.i5.128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 02/19/2022] [Accepted: 07/18/2022] [Indexed: 02/08/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an inflammatory disease of the gastrointestinal (GI) tract. It has financial and quality of life impact on patients. Although there has been a significant advancement in treatments, a considerable number of patients do not respond to it or have severe side effects. Therapeutic approaches such as electrical neuromodulation are being investigated to provide alternate options. Although bioelectric neuromodulation technology has evolved significantly in the last decade, sacral nerve stimulation (SNS) for fecal incontinence remains the only neuromodulation protocol commonly utilized use for GI disease. For IBD treatment, several electrical neuromodulation techniques have been studied, such as vagus NS, SNS, and tibial NS. Several animal and clinical experiments were conducted to study the effectiveness, with encouraging results. The precise underlying mechanisms of action for electrical neuromodulation are unclear, but this modality appears to be promising. Randomized control trials are required to investigate the efficacy of intrinsic processes. In this review, we will discuss the electrical modulation therapy for the IBD and the data pertaining to it.
Collapse
Affiliation(s)
- Farah Yasmin
- Department of Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Abdul Moiz Sahito
- Department of Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Syeda Lamiya Mir
- Department of Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Govinda Khatri
- Department of Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Somina Shaikh
- Department of Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Ambresha Gul
- Department of Medicine, People’s University of Medical and Health Sciences, Nawabshah 67480, Pakistan
| | - Syed Adeel Hassan
- Department of Medicine, University of Louisville, Louiseville, KY 40292, United States
| | - Thoyaja Koritala
- Department of Medicine, Mayo Clinic, Rochester, NY 55902, United States
| | - Salim Surani
- Department of Medicine, Texas A&M University, College Station, TX 77843, United States
- Department of Anesthesiology, Mayo Clinic, Rochester, MN 55902, United States
| |
Collapse
|
16
|
Targeting Lineage-Specific Transcription Factors and Cytokines of the Th17/Treg Axis by Novel 1,3,4-Oxadiazole Derivatives of Pyrrolo[3,4-d]pyridazinone Attenuates TNBS-Induced Experimental Colitis. Int J Mol Sci 2022; 23:ijms23179897. [PMID: 36077306 PMCID: PMC9456461 DOI: 10.3390/ijms23179897] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 11/25/2022] Open
Abstract
The pharmacotherapy of inflammatory bowel disease (IBD) is still not fully effective and safe. Attempts to search for new IBD drugs remain an incessant research aim. One of the novel approaches is targeting the developmental pathway molecules and effector cytokines of Th17/Treg axis. This study aimed to elucidate the impact of new pyrrolo[3,4-d]pyridazinone derivatives, compounds 7b, 10b, or 13b, on the course of experimental colitis in rats and to assess whether these new compounds may influence Th17/Treg axis. Rats were pretreated with studied compounds intragastrically before intrarectal administration of 2,4,6-trinitrobenzenesulfonic acid used for colitis induction. Body weight loss, disease activity index, colon index, and colon tissue damage were analyzed to evaluate the severity of colitis. The colonic levels of RORγt, STAT3, CCR6, Foxp3, IL-6, IL-10, IL-17, TNF-α, IL-23, and PGE2 were assessed. Pretreatment with compounds 7b and 13b alleviated the severity of colitis and concomitantly counteracted the increased levels of RORγt, STAT3, CCR6, IL-6, IL-17, IL-23, TNF-α, and PGE2. The beneficial effect of compounds 7b and 13b may be due to the decrease in the levels of Th17-specific transcription factors and cytokines. The studied compounds might therefore constitute a promising therapeutic strategy in Th17/Treg imbalance-driven inflammatory conditions such as IBD.
Collapse
|
17
|
Roblin X, Serone A, Yoon OK, Zhuo, L, Grant E, Woo J, Liu J, Galien R, D’Haens G. Effects of JAK1-Preferential Inhibitor Filgotinib on Circulating Biomarkers and Whole Blood Genes/Pathways of Patients With Moderately to Severely Active Crohn's Disease. Inflamm Bowel Dis 2022; 28:1207-1218. [PMID: 34741617 PMCID: PMC9340524 DOI: 10.1093/ibd/izab253] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Pro-inflammatory cytokines are dysregulated in Crohn's disease (CD) and could serve as surrogate markers to improve diagnostic and therapeutic approaches, potentially addressing an unmet need. We profiled circulating biomarkers and whole blood transcriptional pathway activity to identify those associated with CD using data from the phase 2 FITZROY study with filgotinib, an oral preferential janus kinase-1 inhibitor. METHODS Patients with serum and whole blood samples taken from the induction period were included. Serum cytokines were measured (ELISA), whole blood RNA sequenced, and stool samples taken to measure fecal calprotectin (FC). Spearman's Rank correlations were assessed between biomarkers and baseline disease activity; post-treatment endoscopic improvement was measured by the Simplified Endoscopy Score for CD (SES-CD), FC and the Crohn's Disease Activity Index. Effect of filgotinib on circulating biomarkers was also evaluated. RESULTS Serum biomarkers (n = 168) and whole blood RNA sequencing (n = 104) were assessed. Moderate correlation between serum analytes with SES-CD and FC was noted; most highly correlated were acute phase proteins CRP (rho = 0.35 [SES-CD] and 0.47 [FC]), serum amyloid A (rho = 0.40 and 0.39, respectively) and pro-inflammatory cytokines interleukin (IL)-6 (rho = 0.31 and 0.30, respectively), IL-22 (rho = 0.36 and 0.35, respectively), and oncostatin M (rho = 0.35 and 0.33, respectively). Filgotinib treatment was associated with reduction of many candidate biomarkers, particularly in patients with treatment response. Early changes in IL-6 and IL-10 may be prognostic for endoscopic response. CONCLUSIONS Several circulating factors with potential as CD activity biomarkers were identified. Larger studies are necessary to investigate the best utility of these markers for CD.
Collapse
Affiliation(s)
- Xavier Roblin
- Gastroenterology Unit, University Hospital of Saint Etienne, Saint-Priest-en-Jarez, France
| | | | | | | | | | - Jacky Woo
- Gilead Sciences, Inc., Foster City, CA, USA
| | | | | | - Geert D’Haens
- Inflammatory Bowel Disease Centre, Academic Medical Centre, 1105 AZ Amsterdam, Netherlands
| |
Collapse
|
18
|
Choi MG, Ye BD, Yang SK, Shim TS, Jo KW, Park SH. The Risk of Tuberculosis in Patients With Inflammatory Bowel Disease Treated With Vedolizumab or Ustekinumab in Korea. J Korean Med Sci 2022; 37:e107. [PMID: 35411727 PMCID: PMC9001185 DOI: 10.3346/jkms.2022.37.e107] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/10/2022] [Indexed: 11/23/2022] Open
Abstract
The present study investigated the risk of active tuberculosis in patients with inflammatory bowel disease (IBD) treated with vedolizumab or ustekinumab, in actual clinical settings in a country with an intermediate tuberculosis burden. The medical records of 238 patients with IBD who received vedolizumab or ustekinumab were retrospectively reviewed at a tertiary referral center in South Korea. All patients had ≥ 3 months of follow-up duration and underwent a latent tuberculosis infection screening test before initiation of the administration of these drugs. Of the 238 patients enrolled, 181 had Crohn's disease, and 57 had ulcerative colitis. During the median 18.7 months of follow-up, active tuberculosis did not develop in any patient treated with vedolizumab or ustekinumab. Therefore, we concluded that the risk of tuberculosis appears to be low in patients with IBD treated with vedolizumab or ustekinumab in South Korea.
Collapse
Affiliation(s)
- Myeong Geun Choi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mokdong Hospital, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Byong Duk Ye
- Department of Gastroenterology and Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Suk-Kyun Yang
- Department of Gastroenterology and Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Tae Sun Shim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung-Wook Jo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Sang Hyoung Park
- Department of Gastroenterology and Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
19
|
Shrestha N, Xu Y, Prévost JRC, McCartney F, Brayden D, Frédérick R, Beloqui A, Préat V. Impact of PEGylation on an antibody-loaded nanoparticle-based drug delivery system for the treatment of inflammatory bowel disease. Acta Biomater 2022; 140:561-572. [PMID: 34923097 DOI: 10.1016/j.actbio.2021.12.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 12/01/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022]
Abstract
Nanoparticle-based oral drug delivery systems have the potential to target inflamed regions in the gastrointestinal tract by specifically accumulating at disrupted colonic epithelium. But, delivery of intact protein drugs at the targeted site is a major challenge due to the harsh gastrointestinal environment and the protective mucus layer. Biocompatible nanoparticles engineered to target the inflamed colonic tissue and efficiently penetrate the mucosal layer can provide a promising approach for orally delivering monoclonal antibodies to treat inflammatory bowel disease. The study aims to develop mucus-penetrating nanoparticles composed of poly(lactic-co-glycolic acid, PLGA) polymers with two different polyethylene glycol (PEG) chain lengths (2 kDa and 5kDa) to encapsulate monoclonal antibody against tumor necrosis factor-α (TNF-α). The impact of different PEG chain lengths on the efficacy of the nanosystems was evaluated in vitro, ex vivo, and in vivo. Both PLGA-PEG2k and PLGA-PEG5k nanoparticles successfully encapsulated the antibody and significantly reduced TNF-α secretion from activated macrophages and intestinal epithelial cells. However, only antibody-loaded PLGA-PEG2k nanoparticles were able to alleviate the experimental acute colitis in mice demonstrated by improved colon weight/length ratio, histological score, and reduced tissue-associated myeloperoxidase activity and expression of proinflammatory cytokine TNF-α levels compared with the control group. The results suggest that despite having no significant differences in the in vitro cell-based assays, PEG chain length has a significant impact on the in vivo performance of the mucus penetrating nanoparticles. Overall, PLGA-PEG2k nanoparticles were presented as a promising oral delivery system for targeted antibody delivery to treat inflammatory bowel disease. STATEMENT OF SIGNIFICANCE: There is an unmet therapeutic need for oral drug delivery systems for safe and effective antibody therapy of inflammatory bowel disease. Therefore, we have developed PEGylated PLGA-based nanoparticulate drug delivery systems for oral targeted delivery of anti-TNF-α antibody as a potential alternative treatment strategy. The PEG chain length did not affect encapsulation efficiency or interaction with mucin in vitro but resulted in differences in in vitro release profile and in vivo efficacy study. We demonstrated the superiority of anti-TNF-α mAb-PLGA-PEG2k over mAb-PLGA-PEG5k nanoparticles to effectively exhibit anti-inflammatory responses in an acute murine colitis model. These nanoparticle-based formulations may be adjusted to encapsulate other drugs that could be applied to a number of disorders at different mucosal surfaces.
Collapse
Affiliation(s)
- Neha Shrestha
- Université catholique de Louvain, Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Brussels 1200, Belgium.
| | - Yining Xu
- Université catholique de Louvain, Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Brussels 1200, Belgium
| | - Julien R C Prévost
- Université catholique de Louvain, Medicinal Chemistry, Louvain Drug Research Institute, Brussels 1200, Belgium
| | - Fiona McCartney
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield Dublin 4, Ireland
| | - David Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield Dublin 4, Ireland
| | - Raphaël Frédérick
- Université catholique de Louvain, Medicinal Chemistry, Louvain Drug Research Institute, Brussels 1200, Belgium
| | - Ana Beloqui
- Université catholique de Louvain, Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Brussels 1200, Belgium
| | - Véronique Préat
- Université catholique de Louvain, Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Brussels 1200, Belgium.
| |
Collapse
|
20
|
Wang CPJ, Byun MJ, Kim SN, Park W, Park HH, Kim TH, Lee JS, Park CG. Biomaterials as therapeutic drug carriers for inflammatory bowel disease treatment. J Control Release 2022; 345:1-19. [DOI: 10.1016/j.jconrel.2022.02.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 12/13/2022]
|
21
|
Wei W, Zhang Y, Li R, Cao Y, Yan X, Ma Y, Zhang Y, Yang M, Zhang M. Oral Delivery of Pterostilbene by L-Arginine-Mediated “Nano-Bomb” Carrier for the Treatment of Ulcerative Colitis. Int J Nanomedicine 2022; 17:603-616. [PMID: 35177902 PMCID: PMC8843770 DOI: 10.2147/ijn.s347506] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/25/2022] [Indexed: 12/23/2022] Open
Affiliation(s)
- Wei Wei
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Xi’an No.1 Hospital, Shaanxi Institute of Ophthalmology, Shaanxi Key Laboratory of Ophthalmology, Clinical Research Center for Ophthalmology Diseases of Shaanxi Province, First Affiliated Hospital of Northwestern University, Xi’an, Shaanxi, People’s Republic of China
| | - Yujie Zhang
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Runqing Li
- Department of Radiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Yameng Cao
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Xiangji Yan
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Yana Ma
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Yuanyuan Zhang
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Mei Yang
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Correspondence: Mei Yang; Mingzhen Zhang, Email ;
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| |
Collapse
|
22
|
Yang M, Yang C, Zhang Y, Yan X, Ma Y, Zhang Y, Cao Y, Xu Q, Tu K, Zhang M. Orally pH-activated "nano-bomb" carrier combined with berberine by regulating gene silencing and gut microbiota for site-specific treatment of ulcerative colitis. Biomater Sci 2022; 10:1053-1067. [DOI: 10.1039/d1bm01765a] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Ulcerative colitis (UC) is a chronic, relapsing inflammatory bowel disease that features colonic epithelial barrier dysfunction and gut dysbiosis. Preclinical studies demonstrated that inhibiting the overexpression of CD98 via small...
Collapse
|
23
|
Kim J, Yoon H, Kim N, Lee KM, Jung SA, Choi CH, Kim ES, Jung Y, Eun CS, Kim TO, Kang SB, Kim YS, Seo GS, Lee CK, Im JP, Park SJ, Park DI, Ye BD. Clinical Outcomes and Response Predictors of Vedolizumab Induction Treatment for Korean Patients With Inflammatory Bowel Diseases Who Failed Anti-TNF Therapy: A KASID Prospective Multicenter Cohort Study. Inflamm Bowel Dis 2021; 27:1931-1941. [PMID: 33501935 DOI: 10.1093/ibd/izaa361] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND We investigated the real-life effectiveness and safety of vedolizumab (VDZ) induction therapy among Korean patients with Crohn disease (CD) or ulcerative colitis (UC) for whom anti-tumor necrosis factor therapy previously failed. METHODS Adult patients who started VDZ induction therapy at 16 centers were prospectively enrolled in the Korean VDZ nationwide registry. The coprimary outcomes were clinical remission, defined as a Crohn's Disease Activity Index score <150 points and a partial Mayo score ≤2 points with a combined rectal bleeding and stool frequency subscore ≤1 point at week 14 and endoscopic remission defined as a Mayo endoscopic subscore ≤1 point. We also analyzed predictors of clinical remission. RESULTS Between August 2017 and November 2019, a total of 158 patients (80 with CD and 78 with UC) received VDZ induction therapy. Clinical remission rates among patients with CD and patients with UC were 44.1% and 44.0%, respectively. Among patients with UC, the endoscopic remission rate was 32.4%. Clinical response and remission rates showed increasing trends during induction therapy. Multivariable analysis revealed that clinical response at week 6 was the only predictor of clinical remission at week 14 for both patients with CD and patients with UC. Among patients who experienced 1 or more adverse events (n = 71; 44.9%), disease exacerbation (n = 28; 17.7%) was the most common adverse event. CONCLUSIONS Among Korean patients with CD or UC for whom anti-tumor necrosis factor therapy failed, VDZ induction therapy was effective and safe. The early clinical response was associated with clinical remission after VDZ induction therapy.
Collapse
Affiliation(s)
- Jeongseok Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Nayoung Kim
- Department of Biostatistics and Clinical Epidemiology, Asan Medical Center, Seoul, Korea
| | - Kang-Moon Lee
- Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Ae Jung
- Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul, Korea
| | - Chang Hwan Choi
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Eun Soo Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kyungpook National University, School of Medicine, Daegu, Korea
| | - Yunho Jung
- Division of Gastroenterology, Department of Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Chang Soo Eun
- Department of Internal Medicine, Hanyang University Guri Hospital, Guri, Korea
| | - Tae Oh Kim
- Department of Internal Medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Sang-Bum Kang
- Division of Gastroenterology, Department of Internal Medicine, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Korea
| | - You Sun Kim
- Department of Internal Medicine, Seoul Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Geom-Seog Seo
- Department of Internal Medicine, Wonkwang University School of Medicine, Iksan, Korea
| | - Chang Kyun Lee
- Center for Crohn's and Colitis, Department of Gastroenterology, Kyung Hee University College of Medicine, Seoul, Korea
| | - Jong Pil Im
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Soo Jung Park
- Department of Internal Medicine and Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Dong Il Park
- Division of Gastroenterology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Byong Duk Ye
- Department of Gastroenterology and Inflammatory Bowel Disease Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
24
|
Tumor Necrosis Factor's Pathway in Crohn's Disease: Potential for Intervention. Int J Mol Sci 2021; 22:ijms221910273. [PMID: 34638616 PMCID: PMC8508644 DOI: 10.3390/ijms221910273] [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] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022] Open
Abstract
Crohn’s disease (CD) is a chronic disorder characterized by full thickness patchy inflammation of the gastrointestinal tract. The pathogenesis is multifactorial and involves defective innate immune responses, microbiome alterations, and dysregulated activation of the acquired component of mucosal immunity. One of the molecular mediators that is involved at different levels in the initiation and progression of intestinal inflammation characteristic of CD is tumor necrosis factor (TNF). The present manuscript provides a comprehensive review focused on the potential role of TNF in the different phases of CD pathogenesis, particularly in light of its potential clinical implications. Currently available drugs blocking TNF are evaluated and discussed, specifically for open issues that still remain utilizing such therapy. TNF exerts a paramount role in the established phase of intestinal inflammation that characterizes CD patients, and anti-TNF biologics have definitely changed patient management, offering effective and safe options of treatment. Nonetheless, many patients still do not respond to anti-TNF therapy or experience unwanted side-effects. This could partially be due to the role that TNF plays in intestinal homeostasis that is particularly important during the early phase of the inflammatory process. In fact, emerging evidence supporting the dichotomous role of TNF and the identification of molecular markers will guide a more tailored and refined therapy for CD patients in the near future.
Collapse
|
25
|
Heidari N, Abbasi-Kenarsari H, Namaki S, Baghaei K, Zali MR, Ghaffari Khaligh S, Hashemi SM. Adipose-derived mesenchymal stem cell-secreted exosome alleviates dextran sulfate sodium-induced acute colitis by Treg cell induction and inflammatory cytokine reduction. J Cell Physiol 2021; 236:5906-5920. [PMID: 33728664 DOI: 10.1002/jcp.30275] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 12/09/2020] [Accepted: 12/26/2020] [Indexed: 12/16/2022]
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is an inflammatory condition that results in gastrointestinal tract damage. Various factors, including environmental and genetic agents, disrupt the function of the intestinal immune system that can lead to IBD. Mesenchymal stem cells (MSCs) display an immunoregulatory function and demonstrate regenerative potential by paracrine action. In this study, we evaluated the immunomodulatory effects of MSCs' derived exosomes in the acute form of dextran sulfate sodium (DSS)-induced colitis. Exosomes were isolated from adipose-derived MSCs. Acute colitis was induced by DSS. The exosome was used by intraperitoneal injection into mice with acute colitis. Stool consistency, body weight changes, bleeding severity, colon length, and weight were examined. At the experimental endpoint (Day 7), the changes in the colon tissue were evaluated. The level of cytokines of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), interleukin-17 (IL-17), IL-4, IL-12, transforming growth factor-β (TGF-β) and, IL-10, and Treg cells percentage were assayed. Results showed that exosome administration diminished colon shortening, bodyweight loss, bleeding, and colon injury. The levels of IFN-γ, TNF-α, IL-12, and IL-17 were decreased, and the level of TGF-β, IL-4, and IL-10 were increased in lymph node and spleen of mice treated with exosome. Percentages of CD4+ CD25+ Foxp3+ Treg cells were grown in the lymph node and spleen of mice treated with exosomes. Overall, current data suggest that MSC-derived exosome could regulate the Treg population and improves inflammation in DSS-induced acute colitis.
Collapse
Affiliation(s)
- Neda Heidari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hajar Abbasi-Kenarsari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Namaki
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorder Research Center, Research Institute for Gastroenterology and Liver Disease, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Disease, Shahid Beheshti University of Medical Science, Tehran, Iran
| | | | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Antonioli L, Pellegrini C, Fornai M, Benvenuti L, D’Antongiovanni V, Colucci R, Bertani L, Di Salvo C, Semeghini G, La Motta C, Giusti L, Zallocco L, Ronci M, Quattrini L, Angelucci F, Coviello V, Oh WK, Ha QTK, Németh ZH, Haskó G, Blandizzi C. Preclinical Development of FA5, a Novel AMP-Activated Protein Kinase (AMPK) Activator as an Innovative Drug for the Management of Bowel Inflammation. Int J Mol Sci 2021; 22:6325. [PMID: 34199160 PMCID: PMC8231528 DOI: 10.3390/ijms22126325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
Acadesine (ACA), a pharmacological activator of AMP-activated protein kinase (AMPK), showed a promising beneficial effect in a mouse model of colitis, indicating this drug as an alternative tool to manage IBDs. However, ACA displays some pharmacodynamic limitations precluding its therapeutical applications. Our study was aimed at evaluating the in vitro and in vivo effects of FA-5 (a novel direct AMPK activator synthesized in our laboratories) in an experimental model of colitis in rats. A set of experiments evaluated the ability of FA5 to activate AMPK and to compare the efficacy of FA5 with ACA in an experimental model of colitis. The effects of FA-5, ACA, or dexamethasone were tested in rats with 2,4-dinitrobenzenesulfonic acid (DNBS)-induced colitis to assess systemic and tissue inflammatory parameters. In in vitro experiments, FA5 induced phosphorylation, and thus the activation, of AMPK, contextually to the activation of SIRT-1. In vivo, FA5 counteracted the increase in spleen weight, improved the colon length, ameliorated macroscopic damage score, and reduced TNF and MDA tissue levels in DNBS-treated rats. Of note, FA-5 displayed an increased anti-inflammatory efficacy as compared with ACA. The novel AMPK activator FA-5 displays an improved anti-inflammatory efficacy representing a promising pharmacological tool against bowel inflammation.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Carolina Pellegrini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Laura Benvenuti
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Vanessa D’Antongiovanni
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy;
| | - Lorenzo Bertani
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Clelia Di Salvo
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Giorgia Semeghini
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Concettina La Motta
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Laura Giusti
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy;
| | - Lorenzo Zallocco
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Maurizio Ronci
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy;
| | - Luca Quattrini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Francesco Angelucci
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Vito Coviello
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Won-Keun Oh
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Korea; (W.-K.O.); (Q.T.K.H.)
| | - Quy Thi Kim Ha
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Korea; (W.-K.O.); (Q.T.K.H.)
| | - Zoltan H. Németh
- Department of Anesthesiology, Columbia University, New York City, NY 10027, USA; (Z.H.N.); (G.H.)
- Department of Surgery, Morristown Medical Center, Morristown, NJ 07960, USA
| | - Gyorgy Haskó
- Department of Anesthesiology, Columbia University, New York City, NY 10027, USA; (Z.H.N.); (G.H.)
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| |
Collapse
|
27
|
Wang Y, Ye H, Qiao L, Du C, Wei Z, Wang T, Wang J, Liu R, Wang P. Intestinal Anti-Inflammatory Effects of Selenized Ulva pertusa Polysaccharides in a Dextran Sulfate Sodium-Induced Inflammatory Bowel Disease Model. J Med Food 2021; 24:236-247. [PMID: 33739884 DOI: 10.1089/jmf.2020.4787] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The purpose of this study was to examine the alleviative effects of selenized polysaccharides from Ulva pertusa (ulvan-Se) on inflammatory bowel disease (IBD) in mice. The dextran sulfate sodium (DSS)-induced IBD mouse model was used to explore the protective effects of ulvan-Se on the intestinal mechanical and immune barrier. At doses less than 1208 mg/kg·bw ulvan-Se showed no significant damage to Institute of Cancer Research (ICR) mice in an acute toxicity test. The results showed that DSS destroyed the mechanical barrier, which includes epithelial cells, while ulvan-Se promoted mRNA expression of tight junction proteins (zonula occludens protein 1, occludin, and claudin-1) and inhibited the infiltration of white blood cells into the intestines. At 100 mg/kg·bw, ulvan-Se enhanced the antioxidant capacity of mice more effectively than the 50 mg/kg·bw ulvan-Se. Furthermore, ulvan-Se improved the intestinal immune barrier by increasing immunoglobulin A and immunoglobulin M, while regulating the levels of interleukin (IL)-1β, interferon-γ, and IL-4. Oral administration of ulvan-Se also suppressed tumor necrosis factor-α, IL-1β, IL-6, and cyclooxygenase-2 mRNA expression mediated by the nuclear factor kappa B pathway. Taken together, our findings reveal that ulvan-Se could be used as a potential alternative supplement for reducing intestinal inflammation in IBD.
Collapse
Affiliation(s)
- Yifan Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China.,State Environmental Protection Key Laboratory of Estuarine and Coastal Environment, Beijing, China
| | - Han Ye
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Leke Qiao
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Chunying Du
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | | | - Ting Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Jingfeng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Ruizhi Liu
- State Environmental Protection Key Laboratory of Estuarine and Coastal Environment, Beijing, China
| | - Peng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| |
Collapse
|
28
|
Deleu S, Machiels K, Raes J, Verbeke K, Vermeire S. Short chain fatty acids and its producing organisms: An overlooked therapy for IBD? EBioMedicine 2021; 66:103293. [PMID: 33813134 PMCID: PMC8047503 DOI: 10.1016/j.ebiom.2021.103293] [Citation(s) in RCA: 336] [Impact Index Per Article: 112.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/01/2021] [Accepted: 03/07/2021] [Indexed: 02/07/2023] Open
Abstract
The gut microbiome and the intestinal immune system are driving contributors to inflammatory bowel diseases (IBD). Both have an important signalling factor in common: short-chain fatty acids (SCFAs). SCFAs (acetate, propionate and butyrate) are produced by bacterial fermentation in the gut and exert several effects on host metabolism and immune system. This review provides an overview of the current knowledge of these effects, with specific focus on energy metabolism, intestinal barrier, immune system, and disease activity in IBD. To conclude, more research is needed on the cross-feeding mechanisms in the gut microbiome, as well as on the therapeutic potential of SCFAs on different disease models. Also randomized controlled trials and prospective cohort studies should investigate the clinical impact of SCFA administration.
Collapse
Affiliation(s)
- Sara Deleu
- Department of Chronic Diseases, Metabolism & Ageing [CHROMETA], Translational Research Center for Gastrointestinal Disorders [TARGID], KU Leuven, Leuven, Belgium
| | - Kathleen Machiels
- Department of Chronic Diseases, Metabolism & Ageing [CHROMETA], Translational Research Center for Gastrointestinal Disorders [TARGID], KU Leuven, Leuven, Belgium
| | - Jeroen Raes
- Center for Microbiology, VIB, Leuven, Belgium; Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Kristin Verbeke
- Department of Chronic Diseases, Metabolism & Ageing [CHROMETA], Translational Research Center for Gastrointestinal Disorders [TARGID], KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Department of Chronic Diseases, Metabolism & Ageing [CHROMETA], Translational Research Center for Gastrointestinal Disorders [TARGID], KU Leuven, Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium.
| |
Collapse
|
29
|
Chela H, Pasha SB, Wan XF, Ghouri YA. A review on medical management of inflammatory bowel disease during the coronavirus disease 2019 pandemic. J Gastroenterol Hepatol 2021; 36:918-926. [PMID: 32876952 DOI: 10.1111/jgh.15241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/25/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022]
Abstract
The coronavirus disease 2019 pandemic has engulfed the world and is the highlight of medical community at this time. As humanity fights the battle against this virus, questions are arising regarding the appropriate management of at risk patient populations. The immunocompromised cohort is particularly susceptible to this infection, and we have tried to explore the medical management of one such group, which is composed of individuals with inflammatory bowel disease (IBD). There is limited data on the management of IBD during the ongoing pandemic. Several medical societies have put forth suggestions on how to manage immunocompromised patients in order to minimize risk of developing coronavirus disease 2019. This review aims to present available recommendations from experts and provides an insight on preventive and therapeutic strategies that can be implemented for the medical management of patients with IBD. We anticipate that as more information arises, new guidelines will emerge.
Collapse
Affiliation(s)
- Harleen Chela
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Syed Bilal Pasha
- Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Xiu-Feng Wan
- MU Center for Research on Influenza Systems Biology (CRISB), University of Missouri, Columbia, Missouri, USA.,Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA.,Department of Electrical Engineering and Computer Science, College of Engineering, University of Missouri, Columbia, Missouri, USA.,Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA.,MU Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, USA
| | - Yezaz A Ghouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri, USA
| |
Collapse
|
30
|
Bonaz B, Sinniger V, Pellissier S. Therapeutic Potential of Vagus Nerve Stimulation for Inflammatory Bowel Diseases. Front Neurosci 2021; 15:650971. [PMID: 33828455 PMCID: PMC8019822 DOI: 10.3389/fnins.2021.650971] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
The vagus nerve is a mixed nerve, comprising 80% afferent fibers and 20% efferent fibers. It allows a bidirectional communication between the central nervous system and the digestive tract. It has a dual anti-inflammatory properties via activation of the hypothalamic pituitary adrenal axis, by its afferents, but also through a vago-vagal inflammatory reflex involving an afferent (vagal) and an efferent (vagal) arm, called the cholinergic anti-inflammatory pathway. Indeed, the release of acetylcholine at the end of its efferent fibers is able to inhibit the release of tumor necrosis factor (TNF) alpha by macrophages via an interneuron of the enteric nervous system synapsing between the efferent vagal endings and the macrophages and releasing acetylcholine. The vagus nerve also synapses with the splenic sympathetic nerve to inhibit the release of TNF-alpha by splenic macrophages. It can also activate the spinal sympathetic system after central integration of its afferents. This anti-TNF-alpha effect of the vagus nerve can be used in the treatment of chronic inflammatory bowel diseases, represented by Crohn’s disease and ulcerative colitis where this cytokine plays a key role. Bioelectronic medicine, via vagus nerve stimulation, may have an interest in this non-drug therapeutic approach as an alternative to conventional anti-TNF-alpha drugs, which are not devoid of side effects feared by patients.
Collapse
Affiliation(s)
- Bruno Bonaz
- Division of Hepato-Gastroenterology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France.,Grenoble Institute of Neurosciences, Inserm U1216, University Grenoble Alpes, Grenoble, France
| | - Valérie Sinniger
- Division of Hepato-Gastroenterology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France.,Grenoble Institute of Neurosciences, Inserm U1216, University Grenoble Alpes, Grenoble, France
| | - Sonia Pellissier
- Laboratoire Inter-Universitaire de Psychologie Personnalité, Cognition, Changement Social, University Grenoble Alpes, University Savoie Mont Blanc, Grenoble, France
| |
Collapse
|
31
|
Ferenczi S, Solymosi N, Horváth I, Szeőcs N, Grózer Z, Kuti D, Juhász B, Winkler Z, Pankotai T, Sükösd F, Stágel A, Paholcsek M, Dóra D, Nagy N, Kovács KJ, Zanoni I, Szallasi Z. Efficient treatment of a preclinical inflammatory bowel disease model with engineered bacteria. Mol Ther Methods Clin Dev 2021; 20:218-226. [PMID: 33426148 PMCID: PMC7782194 DOI: 10.1016/j.omtm.2020.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 11/12/2020] [Indexed: 01/02/2023]
Abstract
We developed an orally administered, engineered, bacterium-based, RNA interference-mediated therapeutic method to significantly reduce the symptoms in the most frequently used animal model of inflammatory bowel disease. This bacterium-mediated RNA interference strategy was based on the genomically stable, non-pathogenic E. coli MDS42 strain, which was engineered to constitutively produce invasin and the listeriolysin O cytolysin. These proteins enabled the bacteria first to invade the colon epithelium and then degrade in the phagosome. This allowed the delivery of a plasmid encoding small hairpin RNA (shRNA) targeting tumor necrosis factor (TNF) into the cytoplasm of the target cells. The expression levels of TNF and other cytokines significantly decreased upon this treatment in dextran sulfate sodium (DSS)-induced colitis, and the degree of inflammation was significantly reduced. With further safety modifications this method could serve as a safe and side effect-free alternative to biologicals targeting TNF or other inflammatory mediators.
Collapse
Affiliation(s)
- Szilamer Ferenczi
- Institute of Experimental Medicine, Laboratory of Molecular Neuroendocrinology, Budapest, Hungary
- Central European Biosystems, Gödöllő, Hungary
| | - Norbert Solymosi
- Centre for Bioinformatics, University of Veterinary Medicine Budapest, Budapest, Hungary
| | | | | | | | - Dániel Kuti
- Institute of Experimental Medicine, Laboratory of Molecular Neuroendocrinology, Budapest, Hungary
| | - Balázs Juhász
- Institute of Experimental Medicine, Laboratory of Molecular Neuroendocrinology, Budapest, Hungary
| | - Zsuzsanna Winkler
- Institute of Experimental Medicine, Laboratory of Molecular Neuroendocrinology, Budapest, Hungary
| | - Tibor Pankotai
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Farkas Sükösd
- Department of Pathology, Laboratory of Molecular Pathology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Anikó Stágel
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Melinda Paholcsek
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dávid Dóra
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Nándor Nagy
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Krisztina J. Kovács
- Institute of Experimental Medicine, Laboratory of Molecular Neuroendocrinology, Budapest, Hungary
| | - Ivan Zanoni
- Divisions of Immunology and Gastroenterology, Harvard Medical School, Boston Children’s Hospital, Boston, MA, USA
| | - Zoltan Szallasi
- Danish Cancer Society Research Center, Copenhagen, Denmark
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, USA
- 2nd Department of Pathology, MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
| |
Collapse
|
32
|
Pistol GC, Bulgaru CV, Marin DE, Oancea AG, Taranu I. Dietary Grape Seed Meal Bioactive Compounds Alleviate Epithelial Dysfunctions and Attenuates Inflammation in Colon of DSS-Treated Piglets. Foods 2021; 10:foods10030530. [PMID: 33806347 PMCID: PMC7999447 DOI: 10.3390/foods10030530] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/25/2021] [Accepted: 02/27/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammatory Bowel Diseases (IBD) are chronic inflammations associated with progressive degradation of intestinal epithelium and impairment of the local innate immune response. Restoring of epithelial integrity and of the mucosal barrier function, together with modulation of inflammatory and innate immune markers, represent targets for alternative strategies in IBD. The aim of our study was to evaluate the effects of a diet including 8% grape seed meal (GSM), rich in bioactive compounds (polyphenols, polyunsaturated fatty acids (PUFAs), fiber) on the markers of colonic epithelial integrity, mucosal barrier function, pro-inflammatory, and innate immunity in DSS-treated piglets used as animal models of intestinal inflammation. Our results have demonstrated the beneficial effects of bioactive compounds from dietary GSM, exerted at three complementary levels: (a) restoration of the epithelial integrity and mucosal barrier reinforcement by modulation of claudins, Occludin (OCCL) and Zonula-1 (ZO-1) tight junction genes and proteins, myosin IXB (MYO9B) and protein tyrosine phosphatase (PTPN) tight junction regulators and mucin-2 (MUC2) gene; (b) reduction of pro-inflammatory MMP-2 (matrix metalloproteinase-2) and MMP-9 (matrix metalloproteinase-9) genes and activities; and (c) suppression of the innate immune TLR-2 (Toll-like receptor-2) and TLR-4 (Toll-like receptor-4) genes and attenuation of the expression of MyD88 (Myeloid Differentiation Primary Response 88)/MD-2 (Myeloid differentiation factor-2) signaling molecules. These beneficial effects of GSM could further attenuate the transition of chronic colitis to carcinogenesis, by modulating the in-depth signaling mediators belonging to the Wnt pathway.
Collapse
Affiliation(s)
- Gina Cecilia Pistol
- Laboratory of Animal Biology, INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, 077015 Balotesti, Romania; (C.V.B.); (D.E.M.); (I.T.)
- Correspondence: ; Tel.: +40-21-351-2082
| | - Cristina Valeria Bulgaru
- Laboratory of Animal Biology, INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, 077015 Balotesti, Romania; (C.V.B.); (D.E.M.); (I.T.)
| | - Daniela Eliza Marin
- Laboratory of Animal Biology, INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, 077015 Balotesti, Romania; (C.V.B.); (D.E.M.); (I.T.)
| | - Alexandra Gabriela Oancea
- Laboratory of Chemistry and Nutrition Physiology, INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, 077015 Balotesti, Romania;
| | - Ionelia Taranu
- Laboratory of Animal Biology, INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, 077015 Balotesti, Romania; (C.V.B.); (D.E.M.); (I.T.)
| |
Collapse
|
33
|
Franzin M, Stefančič K, Lucafò M, Decorti G, Stocco G. Microbiota and Drug Response in Inflammatory Bowel Disease. Pathogens 2021; 10:211. [PMID: 33669168 PMCID: PMC7919657 DOI: 10.3390/pathogens10020211] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 02/06/2023] Open
Abstract
A mutualistic relationship between the composition, function and activity of the gut microbiota (GM) and the host exists, and the alteration of GM, sometimes referred as dysbiosis, is involved in various immune-mediated diseases, including inflammatory bowel disease (IBD). Accumulating evidence suggests that the GM is able to influence the efficacy of the pharmacological therapy of IBD and to predict whether individuals will respond to treatment. Additionally, the drugs used to treat IBD can modualate the microbial composition. The review aims to investigate the impact of the GM on the pharmacological therapy of IBD and vice versa. The GM resulted in an increase or decrease in therapeutic responses to treatment, but also to biotransform drugs to toxic metabolites. In particular, the baseline GM composition can help to predict if patients will respond to the IBD treatment with biologic drugs. On the other hand, drugs can affect the GM by incrementing or reducing its diversity and richness. Therefore, the relationship between the GM and drugs used in the treatment of IBD can be either beneficial or disadvantageous.
Collapse
Affiliation(s)
- Martina Franzin
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Katja Stefančič
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (K.S.); (G.S.)
| | - Marianna Lucafò
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy;
| | - Giuliana Decorti
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy;
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy;
| | - Gabriele Stocco
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (K.S.); (G.S.)
| |
Collapse
|
34
|
Jowett GM, Norman MDA, Yu TTL, Rosell Arévalo P, Hoogland D, Lust ST, Read E, Hamrud E, Walters NJ, Niazi U, Chung MWH, Marciano D, Omer OS, Zabinski T, Danovi D, Lord GM, Hilborn J, Evans ND, Dreiss CA, Bozec L, Oommen OP, Lorenz CD, da Silva RMP, Neves JF, Gentleman E. ILC1 drive intestinal epithelial and matrix remodelling. NATURE MATERIALS 2021; 20:250-259. [PMID: 32895507 PMCID: PMC7611574 DOI: 10.1038/s41563-020-0783-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 07/23/2020] [Indexed: 05/02/2023]
Abstract
Organoids can shed light on the dynamic interplay between complex tissues and rare cell types within a controlled microenvironment. Here, we develop gut organoid cocultures with type-1 innate lymphoid cells (ILC1) to dissect the impact of their accumulation in inflamed intestines. We demonstrate that murine and human ILC1 secrete transforming growth factor β1, driving expansion of CD44v6+ epithelial crypts. ILC1 additionally express MMP9 and drive gene signatures indicative of extracellular matrix remodelling. We therefore encapsulated human epithelial-mesenchymal intestinal organoids in MMP-sensitive, synthetic hydrogels designed to form efficient networks at low polymer concentrations. Harnessing this defined system, we demonstrate that ILC1 drive matrix softening and stiffening, which we suggest occurs through balanced matrix degradation and deposition. Our platform enabled us to elucidate previously undescribed interactions between ILC1 and their microenvironment, which suggest that they may exacerbate fibrosis and tumour growth when enriched in inflamed patient tissues.
Collapse
Affiliation(s)
- Geraldine M Jowett
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
- Centre for Host Microbiome Interactions, King's College London, London, UK
- Wellcome Trust Cell Therapies and Regenerative Medicine PhD Programme, London, UK
- Centre for Stem Cells & Regenerative Medicine, King's College London, London, UK
| | - Michael D A Norman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Tracy T L Yu
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | | | | | - Suzette T Lust
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Emily Read
- Centre for Host Microbiome Interactions, King's College London, London, UK
- Wellcome Trust Cell Therapies and Regenerative Medicine PhD Programme, London, UK
| | - Eva Hamrud
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
- Wellcome Trust Cell Therapies and Regenerative Medicine PhD Programme, London, UK
- Centre for Stem Cells & Regenerative Medicine, King's College London, London, UK
| | - Nick J Walters
- BioMediTech, Tampere University Tampere Finland, Helsinki, Finland
- Natural Resources Institute Finland, Helsinki, Finland
| | - Umar Niazi
- Guy's and St Thomas' National Health Service Foundation Trust and King's College London National Institute for Health Research Biomedical Research Centre Translational Bioinformatics Platform, Guy's Hospital, London, UK
| | - Matthew Wai Heng Chung
- Centre for Host Microbiome Interactions, King's College London, London, UK
- Wellcome Trust Cell Therapies and Regenerative Medicine PhD Programme, London, UK
- Centre for Stem Cells & Regenerative Medicine, King's College London, London, UK
| | - Daniele Marciano
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Omer S Omer
- School of Immunology and Microbial Sciences, King's College London, London, UK
- Department of Gastroenterology, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| | - Tomasz Zabinski
- Centre for Host Microbiome Interactions, King's College London, London, UK
| | - Davide Danovi
- Centre for Stem Cells & Regenerative Medicine, King's College London, London, UK
| | - Graham M Lord
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jöns Hilborn
- Department of Chemistry, Ångström Laboratory, Uppsala University, Uppsala, Sweden
| | - Nicholas D Evans
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Cécile A Dreiss
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Laurent Bozec
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Oommen P Oommen
- Bioengineering and Nanomedicine Lab, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Ricardo M P da Silva
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
- i3S-Instituto de Investigação e Inovação em Saúde-and INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Joana F Neves
- Centre for Host Microbiome Interactions, King's College London, London, UK.
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK.
| |
Collapse
|
35
|
Ilan Y. Second-Generation Digital Health Platforms: Placing the Patient at the Center and Focusing on Clinical Outcomes. Front Digit Health 2020; 2:569178. [PMID: 34713042 PMCID: PMC8521820 DOI: 10.3389/fdgth.2020.569178] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022] Open
Abstract
Artificial intelligence (AI) digital health systems have drawn much attention over the last decade. However, their implementation into medical practice occurs at a much slower pace than expected. This paper reviews some of the achievements of first-generation AI systems, and the barriers facing their implementation into medical practice. The development of second-generation AI systems is discussed with a focus on overcoming some of these obstacles. Second-generation systems are aimed at focusing on a single subject and on improving patients' clinical outcomes. A personalized closed-loop system designed to improve end-organ function and the patient's response to chronic therapies is presented. The system introduces a platform which implements a personalized therapeutic regimen and introduces quantifiable individualized-variability patterns into its algorithm. The platform is designed to achieve a clinically meaningful endpoint by ensuring that chronic therapies will have sustainable effect while overcoming compensatory mechanisms associated with disease progression and drug resistance. Second-generation systems are expected to assist patients and providers in adopting and implementing of these systems into everyday care.
Collapse
|
36
|
Nanoparticle-based therapeutics of inflammatory bowel diseases: a narrative review of the current state and prospects. JOURNAL OF BIO-X RESEARCH 2020. [DOI: 10.1097/jbr.0000000000000078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
37
|
Paradoxical Psoriasis Induced by Anti-TNFα Treatment: Evaluation of Disease-Specific Clinical and Genetic Markers. Int J Mol Sci 2020; 21:ijms21217873. [PMID: 33114187 PMCID: PMC7660646 DOI: 10.3390/ijms21217873] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/14/2020] [Accepted: 10/21/2020] [Indexed: 02/08/2023] Open
Abstract
Paradoxical psoriasis (PP) may occur during treatment with anti-tumor necrosis factor-alpha (TNF-α) drugs in various chronic immune-mediated diseases, mainly inflammatory bowel diseases (IBD) and psoriasis. In this study, clinical and genetic characteristics of PP arising in IBD and psoriatic patients were investigated to identify disease-specific markers of the paradoxical effect. A total of 161 IBD and psoriatic patients treated with anti-TNF-α drugs were included in the study. Of these patients, 39 developed PP. All patients were characterized for the main clinical–pathologic characteristics and genotyped for six candidate single nucleotide polymorphisms (SNPs) selected for their possible role in PP susceptibility. In IBD patients, the onset of PP was associated with female sex, presence of comorbidities, and use of adalimumab. IBD patients with PP had a higher frequency of the TNF-α rs1799964 rare allele (p = 0.006) compared with cases without the paradoxical effect, and a lower frequency of the human leucocyte antigen (HLA)-Cw06 rs10484554 rare allele (p = 0.03) compared with psoriatic patients with PP. Overall, these findings point to specific clinical and genetic characteristics of IBD patients with PP and provide data showing that genetic variability may be related to the paradoxical effect of anti-TNF-α drugs with possible implications into clinical practice.
Collapse
|
38
|
Andrade AWL, Guerra GCB, de Souza Araújo DF, de Araújo Júnior RF, de Araújo AA, de Carvalho TG, Fernandes JM, Diez-Echave P, Hidalgo-García L, Rodriguez-Cabezas ME, Gálvez J, Zucolotto SM. Anti-Inflammatory and Chemopreventive Effects of Bryophyllum pinnatum (Lamarck) Leaf Extract in Experimental Colitis Models in Rodents. Front Pharmacol 2020; 11:998. [PMID: 32848723 PMCID: PMC7403504 DOI: 10.3389/fphar.2020.00998] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/19/2020] [Indexed: 12/18/2022] Open
Abstract
Inflammatory bowel diseases, mainly ulcerative colitis and Crohn's disease are characterized by chronic inflammation in the intestine. Currently several therapeutic strategies available to treat inflammatory bowel diseases. Though, most treatments can be associated with serious adverse effects what justifies the search for new treatments. In this sense, we highlight the interest in herbal products rich in bioactive compounds which immunomodulatory and antioxidant properties as is the case of Bryophyllum pinnatum (Crassulaceae). This plant is used in traditional medicine in Brazil for treating inflammatory diseases. We hypothesized that hydroethanolic B. pinnatum leaf extract has intestinal anti-inflammatory effects on two experimental colitis models: 2.4-dinitrobenzene sulfonic acid (DNBS) in rats, and dextran sulfate sodium (DSS) in mice. Ultra-fast liquid chromatography method used for the quantification of the main compounds indicated good linearity, specificity, selectivity, precision, robustness and accuracy. The major flavonoids (mg/g of the extract) quantified were: quercetin 3-O-α-L-arabinopyranosyl-(1→2)-α-L-rhamnopyranoside (35.56 ± 0.086 mg/g), kaempferol 3-O-α-L-arabinopyranosyl-(1→2)-α-L-rhamnopyranoside (4.66 ± 0.076 mg/g) and quercetin-3-O-rhamnopyranoside (4.56 ± 0.026 mg/g). The results obtained in the DNBS and DSS models indicate that extract has both chemopreventive and anti-inflammatory effects, observing a significant reduction in the disease activity index score, and less macroscopic and microscopic damage. The extract promoted downregulation of Toll-like receptor and kappa B p65 nuclear factor gene expression, leading to a reduction in pro-inflammatory and oxidative mediators, chemokines, and cell adhesion molecules. This immunomodulatory property was proposed that one of the possible action mechanisms of extract. An improvement in intestinal damage was also associated with a reduction in oxidative stress and infiltration of leukocytes, as evidenced by the reduction in malonaldialdehyde and myeloperoxidase activity and increase in total glutathione in the colonic tissue. Moreover, the extract improved the cytoarchitecture of the colonic tissue and the integrity of the intestinal epithelial barrier by restoring the expression of the proteins associated with mucosa protection. In view of the beneficial effects showed by the B. pinnatum leaf extract in preclinical rodent models of colitis there is the potential to conduct some future clinical studies to ensure safe and effective development of a phytotherapeutic treatment for human inflammatory bowel diseases.
Collapse
Affiliation(s)
- Anderson Wilbur Lopes Andrade
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, Brazil.,Health Science Center, Postgraduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | | | - Raimundo Fernandes de Araújo Júnior
- Postgraduate Program in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil.,Postgraduate Program in Functional and Structural Biology, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil
| | | | - Thaís Gomes de Carvalho
- Postgraduate Program in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil
| | - Júlia Morais Fernandes
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Patrícia Diez-Echave
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), University of Granada, Granada, Spain.,CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Laura Hidalgo-García
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), University of Granada, Granada, Spain.,CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Maria Elena Rodriguez-Cabezas
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), University of Granada, Granada, Spain.,CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Julio Gálvez
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), University of Granada, Granada, Spain.,CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Silvana Maria Zucolotto
- Health Science Center, Postgraduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
39
|
Magro F, Cordeiro G, Dias AM, Estevinho MM. Inflammatory Bowel Disease - Non-biological treatment. Pharmacol Res 2020; 160:105075. [PMID: 32653651 DOI: 10.1016/j.phrs.2020.105075] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022]
Abstract
Nowadays, non-biological treatments remain valuable approaches among the therapeutic armamentarium of inflammatory bowel disease (IBD). Mesalamine is the core treatment of mild‑to‑moderate ulcerative colitis (UC) and corticosteroids are crucial for the induction of remission of moderate‑to‑severe flares in both UC and Crohn's disease (CD). Even approaches as cyclosporine, tacrolimus, azathioprine, methotrexate, and surgery still have a nuclear position as strategies to induce and/or maintain remission in IBD. Due to their particularities and to the accumulated evidence, each of these strategies conquered peculiar roles in the overall IBD strategy, all of them contributing to better outcomes. This review emphasizes the particular roles that non-biological treatments gained over time: recent mesalamine formulations to increase adhesion rates, higher doses of 5-ASA for high-risk patients, MMX technology to improve drug release and attain higher bowel concentrations, cyclosporine as a bridge to vedolizumab, tacrolimus as a potential alternative to thiopurines or infliximab, azathioprine in combination therapy with infliximab and dubious in monotherapy, and surgery as a mean to a "better end".
Collapse
Affiliation(s)
- Fernando Magro
- Clinical Pharmacology Unit, São João Hospital University Centre, Porto, Portugal; Department of Biomedicine, Unit of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal; Service of Gastroenterology, São João Hospital University Centre, Porto, Portugal.
| | - Gonçalo Cordeiro
- Clinical Pharmacology Unit, São João Hospital University Centre, Porto, Portugal
| | - Andreia Martins Dias
- Clinical Pharmacology Unit, São João Hospital University Centre, Porto, Portugal
| | - Maria Manuela Estevinho
- Department of Biomedicine, Unit of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Gastroenterology, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| |
Collapse
|
40
|
Antonioli L, Fornai M, Pellegrini C, Masi S, Puxeddu I, Blandizzi C. Ectopic Lymphoid Organs and Immune-Mediated Diseases: Molecular Basis for Pharmacological Approaches. Trends Mol Med 2020; 26:1021-1033. [PMID: 32600794 DOI: 10.1016/j.molmed.2020.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/21/2020] [Accepted: 06/04/2020] [Indexed: 12/15/2022]
Abstract
Chronic inflammation is the result a persistent increase in the expression of several proinflammatory pathways with impaired inflammatory resolution. Ectopic lymphoid organs (ELOs), untypical lymphoid annexes, emerge during chronic inflammation and contribute to the physiopathology of chronic inflammatory disorders. This review discusses the pathophysiological role of ELOs in the progression of immune-mediated inflammatory diseases (IMIDs), including multiple sclerosis (MS), rheumatoid arthritis (RA), inflammatory bowel disease (IBD), atherosclerosis, and Sjögren syndrome (SSj). The molecular pathways underlying the emergence of ELOs are of interest for the development of novel pharmacological approaches for the management of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy.
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | | | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Ilaria Puxeddu
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
41
|
Silver nanoparticles based on blackcurrant extract show potent anti-inflammatory effect in vitro and in DSS-induced colitis in mice. Int J Pharm 2020; 585:119549. [PMID: 32554032 DOI: 10.1016/j.ijpharm.2020.119549] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022]
Abstract
Silver nanoparticles have been used in a range of applications and although they are already employed in medicine, there are new, promising possibilities for their utilization. We investigated the potential of silver nanoparticles obtained with the use of blackcurrant extract in vitro in the LPS-stimulated RAW264.7 macrophages and in vivo in the murine DSS-induced colitis model. The examined formulations contained particles of 95 nm (Ag95) and 213 nm (Ag213) diameter. In vitro, both formulations inhibited nitric oxide (NO) release. In vivo, the preparations alleviated colitis as evidenced by a decreased macroscopic score and myeloperoxidase activity (indicative of neutrophil infiltration). In both cases, the nanoparticles of larger diameter showed better anti-inflammatory properties. Although further tests are required, our results indicate a plausible new use of silver nanoparticles in inflammatory bowel diseases.
Collapse
|
42
|
Troncone E, Marafini I, Del Vecchio Blanco G, Di Grazia A, Monteleone G. Novel Therapeutic Options for People with Ulcerative Colitis: An Update on Recent Developments with Janus Kinase (JAK) Inhibitors. Clin Exp Gastroenterol 2020; 13:131-139. [PMID: 32440190 PMCID: PMC7211304 DOI: 10.2147/ceg.s208020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022] Open
Abstract
Crohn’s disease (CD) and ulcerative colitis (UC), the main forms of inflammatory bowel disease (IBD) in human beings, are chronic relapsing-remitting disorders of the gastrointestinal tract, which usually require lifelong therapies. For many years, IBD have been managed with corticosteroids, aminosalicylates and immunosuppressants (ie, thiopurines). The advent of biologic therapies (anti-TNF-α agents) has significantly improved the outcome of IBD patients in terms of prolonged clinical remission, corticosteroid sparing, achievement of mucosal healing and prevention of disease-related complications. Nevertheless, primary failure or loss of response to biologics occur in about 50% of patients treated with these drugs. Therefore, the need for new effective treatments for such patients has critically emerged as an urgent priority. With this regard, several small-molecule drugs (SMDs) targeting lymphocyte trafficking (ie, sphingosine-1-phosphate receptor modulators) and the JAK/STAT pathway (eg, tofacitinib) have been recently developed and tested in IBD. In particular, JAK inhibitors are oral compounds characterized by short half-life, low antigenicity and the ability to dampen several pro-inflammatory pathways simultaneously. Tofacitinib, a pan-JAK inhibitor, has shown good efficacy and safety in UC clinical trials and has been recently approved for the treatment of UC patients. In this review, we analyze the main evidence supporting the use of JAK inhibitors in UC and explore the unanswered questions about the use of this class of drug in UC.
Collapse
Affiliation(s)
- Edoardo Troncone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Irene Marafini
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | | | - Antonio Di Grazia
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
43
|
Gorczynski RM, Hoffmann G. Combined IMIG and immune Ig attenuates inflammatory colitis in mice. Int Immunopharmacol 2020; 83:106464. [PMID: 32278130 DOI: 10.1016/j.intimp.2020.106464] [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: 02/13/2020] [Revised: 03/13/2020] [Accepted: 03/29/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Using a combination of homologous and heterologous (mouse/human) polyclonal anti-idiotypic Igs and immune Igs in BALB/c mice we have previously reported attenuation of allergic type responses following OVA immunization. We have now investigated attenuation of an inflammatory colitis in C57BL/6 mice receiving dextran sodium sulfate (DSS) in their drinking water, using additional treatment of DSS-exposed mice with combined human Igs, commercial IVIG (given IM, hence hereafter IMIG) as a source of pooled anti-idiotype Ig, and human anti-Tet as immune Ig. METHODS Acute or chronic colitis was induced by DSS in groups of C57BL/6 mice. Mice also received weekly immunotherapy with im injections of polyclonal immune Ig, polyclonal anti-idiotype Ig, or the combined Igs, for a total of 5 injections, beginning with DSS treatment or after 2 cycles of DSS. Weight loss and mortality were monitored daily, and the extent of colitis was determined further using colonic length measurement, and by ELISA measurement of inflammatory cytokines in supernatants from colonic explant cultures. RESULTS Mice developed colitis in both the acute and chronic models with loss of body weight, shortened colon lengths, and increased expression of inflammatory cytokines in colonic tissue. Loss of body weight, and inflammatory cytokine production, was attenuated only in chronic colitis, and only after combined IMIG and immune Ig treatment, and not in groups receiving only IMIG or immune Ig alone. CONCLUSION Heterologous combinations of polyclonal IMIG and immune Ig can attenuate inflammatory colitis in mice. Given the described efficacy of this treatment for allergic desensitization, we hypothesize this methodology may have widespread clinic utility.
Collapse
Affiliation(s)
- R M Gorczynski
- Universityof Toronto, ON, Canada; Network Immunology, Vancouver, BC, Canada.
| | - G Hoffmann
- Network Immunology, Vancouver, BC, Canada
| |
Collapse
|
44
|
Aminoacyl-tRNA synthetase inhibition activates a pathway that branches from the canonical amino acid response in mammalian cells. Proc Natl Acad Sci U S A 2020; 117:8900-8911. [PMID: 32253314 DOI: 10.1073/pnas.1913788117] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Signaling pathways that sense amino acid abundance are integral to tissue homeostasis and cellular defense. Our laboratory has previously shown that halofuginone (HF) inhibits the prolyl-tRNA synthetase catalytic activity of glutamyl-prolyl-tRNA synthetase (EPRS), thereby activating the amino acid response (AAR). We now show that HF treatment selectively inhibits inflammatory responses in diverse cell types and that these therapeutic benefits occur in cells that lack GCN2, the signature effector of the AAR. Depletion of arginine, histidine, or lysine from cultured fibroblast-like synoviocytes recapitulates key aspects of HF treatment, without utilizing GCN2 or mammalian target of rapamycin complex 1 pathway signaling. Like HF, the threonyl-tRNA synthetase inhibitor borrelidin suppresses the induction of tissue remodeling and inflammatory mediators in cytokine-stimulated fibroblast-like synoviocytes without GCN2, but both aminoacyl-tRNA synthetase (aaRS) inhibitors are sensitive to the removal of GCN1. GCN1, an upstream component of the AAR pathway, binds to ribosomes and is required for GCN2 activation. These observations indicate that aaRS inhibitors, like HF, can modulate inflammatory response without the AAR/GCN2 signaling cassette, and that GCN1 has a role that is distinct from its activation of GCN2. We propose that GCN1 participates in a previously unrecognized amino acid sensor pathway that branches from the canonical AAR.
Collapse
|
45
|
Roosenboom B, van Lochem EG, Meijer J, Smids C, Nierkens S, Brand EC, van Erp LW, Kemperman LG, Groenen MJ, Horjus Talabur Horje CS, Wahab PJ. Development of Mucosal PNAd + and MAdCAM-1 + Venules during Disease Course in Ulcerative Colitis. Cells 2020; 9:cells9040891. [PMID: 32268498 PMCID: PMC7226824 DOI: 10.3390/cells9040891] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/31/2020] [Accepted: 04/03/2020] [Indexed: 12/31/2022] Open
Abstract
PNAd and MAdCAM-1 addressins on venules are of importance in T-cell homing and potential therapeutic targets in ulcerative colitis (UC). Normally, PNAd+ high endothelial venules (HEVs) are only present in lymphoid organs, whereas small numbers of MAdCAM-1+ venules can be seen in non-lymphoid tissue. We aimed to study their presence in the intestinal mucosa of UC patients at diagnosis and during follow-up, and their correlation with disease activity. Colonic biopsy specimens of 378 UC patients were analyzed by immunohistochemistry for CD3, CD20, ERG, MECA-79 (PNAd) and MECA-376 (MAdCAM-1) and compared to healthy controls (HC). The proportion of PNAd+HEVs in UC at diagnosis was 4.9% (IQR 2.0%-8.3%), while none were detected in HC. During follow-up, PNAd+HEVs completely disappeared in remission (n = 93), whereas the proportion in active disease was similar to baseline (n = 285, p = 0.39). The proportion of MAdCAM-1+venules in UC at baseline was 5.8% (IQR 2.6-10.0). During follow-up, the proportion in remission was comparable to diagnosis, but upregulated (7.5% (IQR 4.4-10.9), p = 0.001) in active disease. In conclusion, PNAd+HEVs appear in UC during active inflammation which could thus serve as a marker for disease activity, whereas MAdCAM-1+venules remain present after inflammation is resolved and increase after subsequent flares, reflecting chronicity and potentially serving as a therapeutic target.
Collapse
Affiliation(s)
- Britt Roosenboom
- Crohn & Colitis Centre Rijnstate, Department of Gastroenterology and Hepatology, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands
- Correspondence: ; Tel.: +3188-0058952; Fax: +3188-0057506
| | - Ellen G. van Lochem
- Department of Microbiology and Immunology, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands
| | - Jos Meijer
- Department of Pathology, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands
| | - Carolijn Smids
- Crohn & Colitis Centre Rijnstate, Department of Gastroenterology and Hepatology, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands
| | - Stefan Nierkens
- U-DAIR and Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Eelco C. Brand
- Department of Gastroenterology and Hepatology and Center for Translational Immunology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Liselot W. van Erp
- Crohn & Colitis Centre Rijnstate, Department of Gastroenterology and Hepatology, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands
| | | | - Marcel J.M. Groenen
- Crohn & Colitis Centre Rijnstate, Department of Gastroenterology and Hepatology, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands
| | - Carmen S. Horjus Talabur Horje
- Crohn & Colitis Centre Rijnstate, Department of Gastroenterology and Hepatology, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands
| | - Peter J. Wahab
- Crohn & Colitis Centre Rijnstate, Department of Gastroenterology and Hepatology, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands
| |
Collapse
|
46
|
Tyler CJ, Guzman M, Lundborg LR, Yeasmin S, Perez-Jeldres T, Yarur A, Behm B, Dulai PS, Patel D, Bamias G, Rivera-Nieves J. Inherent Immune Cell Variation Within Colonic Segments Presents Challenges for Clinical Trial Design. J Crohns Colitis 2020; 14:1364-1377. [PMID: 32239151 PMCID: PMC7533898 DOI: 10.1093/ecco-jcc/jjaa067] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Intestinal biopsy sampling during IBD trials represents a valuable adjunct strategy for understanding drug responses at the tissue level. Given the length and distinctive embryonic origins of the proximal and distal colon, we investigated whether inherent regional differences of immune cell composition could introduce confounders when sampling different disease stages, or pre/post drug administration. Here, we capitalise on novel mass cytometry technology to perform deep immunophenotyping of distinct healthy colonic segments, using the limited numbers of biopsies that can be harvested from patients. METHODS Biopsies [2.8 mm] were collected from the caecum, transverse colon, descending colon, and rectum of normal volunteers. Intestinal leukocytes were isolated, stained with a panel of 37 antibodies, and mass cytometry data acquired. RESULTS Site-specific patterns of leukocyte localisation were observed. The proximal colon featured increased CD8+ T cells [particularly resident memory], monocytes, and CD19+ B cells. Conversely, the distal colon and rectum tissues exhibited enrichment for CD4+ T cells and antibody-secreting cells. The transverse colon displayed increased abundance of both γδ T cells and NK cells. Subsets of leukocyte lineages also displayed gradients of expression along the colon length. CONCLUSIONS Our results show an inherent regional immune cell variation within colonic segments, indicating that regional mucosal signatures must be considered when assessing disease stages or the prospective effects of trial drugs on leukocyte subsets. Precise protocols for intestinal sampling must be implemented to allow for the proper interpretation of potential differences observed within leukocyte lineages present in the colonic lamina propria.
Collapse
Affiliation(s)
- Christopher J Tyler
- Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA,San Diego VA Medical Center, San Diego, CA, USA
| | - Mauricio Guzman
- Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA,San Diego VA Medical Center, San Diego, CA, USA
| | - Luke R Lundborg
- Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA,San Diego VA Medical Center, San Diego, CA, USA
| | - Shaila Yeasmin
- Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA,San Diego VA Medical Center, San Diego, CA, USA
| | - Tamara Perez-Jeldres
- Universidad Católica de Chile, Santiago, Chile,Hospital San Borja Arriarán, Santiago, Chile
| | - Andres Yarur
- Division of Gastroenterology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian Behm
- Division of Gastroenterology, University of Virginia, Charlottesville, VI, USA
| | | | - Derek Patel
- San Diego VA Medical Center, San Diego, CA, USA
| | - Giorgos Bamias
- GI Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, Athens, Greece
| | - Jesús Rivera-Nieves
- Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA,San Diego VA Medical Center, San Diego, CA, USA,Corresponding author: Jesús Rivera-Nieves, MD, Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, 9500 Gilman Drive Bldg. BRF-II Rm. 4A32, San Diego, CA 92093-0063. Tel.: 858.534.5495; fax: 858.246.1788;
| |
Collapse
|
47
|
Stankovic B, Dragasevic S, Klaassen K, Kotur N, Srzentic Drazilov S, Zukic B, Sokic Milutinovic A, Milovanovic T, Lukic S, Popovic D, Pavlovic S, Nikcevic G. Exploring inflammatory and apoptotic signatures in distinct Crohn's disease phenotypes: Way towards molecular stratification of patients and targeted therapy. Pathol Res Pract 2020; 216:152945. [PMID: 32279918 DOI: 10.1016/j.prp.2020.152945] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Crohn's disease (CD) is chronic inflammatory bowel disease with different phenotypic characteristics influencing disease prognosis and therapeutic strategies. The aim of this pilot study was to analyze selected inflammatory and apoptotic markers in non-inflamed and inflamed samples of ileal mucosa of non-stricturing/non-penetrating (NS/NP) and stricturing (S) CD mucosal phenotypes in order to characterize their distinct profiles. METHODS From twenty CD patients (9 NS/NP, 11 S) paired non-inflamed and inflamed ileal biopsies were collected and used for analysis of cytokine (TNF and IL6) and apoptotic (Bcl2, Bax, Fas and FasL) genes' expression levels by real-time PCR, while NFκB transcriptional potency was assessed by electromobility gel shift assay. RESULTS Our results demonstrated significant upregulation of TNF and IL6 in inflamed area of both NS/NP (p = 0.03, p = 0.01) and S phenotypes (p = 0.04, p = 0.04), respectively. However, TNF increase was more prominent in NS/NP compared to S inflamed mucosa (p = 0.02). Also, level of proapoptotic Bax was significantly higher in NS/NP compared to S inflamed mucosa (p = 0.01). Opposing transcription potency of NFκB has been detected between two phenotypes: being decreased in NS/NP (p = 0.07) and increased in S (p = 0.1) inflamed compared to non-inflamed mucosa, demonstrating trend towards statistical significance. CONCLUSIONS We found that two distinct CD phenotypes have specific molecular signatures. Obtained results could direct improvement of current and development of new therapeutic strategies based on more specific molecular stratification of CD patients.
Collapse
Affiliation(s)
- Biljana Stankovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia.
| | - Sanja Dragasevic
- Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, Koste Todorovica 2, 11000 Belgrade, Serbia; School of Medicine, University of Belgrade, Dr Subotica 8, 11000 Belgrade, Serbia.
| | - Kristel Klaassen
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia.
| | - Nikola Kotur
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia.
| | - Sanja Srzentic Drazilov
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia.
| | - Branka Zukic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia.
| | - Aleksandra Sokic Milutinovic
- Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, Koste Todorovica 2, 11000 Belgrade, Serbia; School of Medicine, University of Belgrade, Dr Subotica 8, 11000 Belgrade, Serbia.
| | - Tamara Milovanovic
- Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, Koste Todorovica 2, 11000 Belgrade, Serbia; School of Medicine, University of Belgrade, Dr Subotica 8, 11000 Belgrade, Serbia.
| | - Snezana Lukic
- Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, Koste Todorovica 2, 11000 Belgrade, Serbia; School of Medicine, University of Belgrade, Dr Subotica 8, 11000 Belgrade, Serbia.
| | - Dragan Popovic
- Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, Koste Todorovica 2, 11000 Belgrade, Serbia; School of Medicine, University of Belgrade, Dr Subotica 8, 11000 Belgrade, Serbia.
| | - Sonja Pavlovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia.
| | - Gordana Nikcevic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia.
| |
Collapse
|
48
|
Caër C, Wick MJ. Human Intestinal Mononuclear Phagocytes in Health and Inflammatory Bowel Disease. Front Immunol 2020; 11:410. [PMID: 32256490 PMCID: PMC7093381 DOI: 10.3389/fimmu.2020.00410] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/21/2020] [Indexed: 12/18/2022] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a complex immune-mediated disease of the gastrointestinal tract that increases morbidity and negatively influences the quality of life. Intestinal mononuclear phagocytes (MNPs) have a crucial role in maintaining epithelial barrier integrity while controlling pathogen invasion by activating an appropriate immune response. However, in genetically predisposed individuals, uncontrolled immune activation to intestinal flora is thought to underlie the chronic mucosal inflammation that can ultimately result in IBD. Thus, MNPs are involved in fine-tuning mucosal immune system responsiveness and have a critical role in maintaining homeostasis or, potentially, the emergence of IBD. MNPs include monocytes, macrophages and dendritic cells, which are functionally diverse but highly complementary. Despite their crucial role in maintaining intestinal homeostasis, specific functions of human MNP subsets are poorly understood, especially during diseases such as IBD. Here we review the current understanding of MNP ontogeny, as well as the recently identified human intestinal MNP subsets, and discuss their role in health and IBD.
Collapse
Affiliation(s)
- Charles Caër
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mary Jo Wick
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
49
|
Inhibitors Targeting RIPK1/RIPK3: Old and New Drugs. Trends Pharmacol Sci 2020; 41:209-224. [PMID: 32035657 DOI: 10.1016/j.tips.2020.01.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/13/2019] [Accepted: 01/02/2020] [Indexed: 12/26/2022]
Abstract
The scaffolding function of receptor-interacting protein kinase 1 (RIPK1) regulates prosurvival signaling and inflammatory gene expression, while its kinase activity mediates both apoptosis and necroptosis; the latter involving RIPK3 kinase activity. The mutual transition between the scaffold and kinase functions of RIPK1 is regulated by (de)ubiquitylation and (de)phosphorylation. RIPK1-mediated cell death leads to disruption of epithelial barriers and/or release of damage-associated molecular patterns (DAMPs), cytokines, and chemokines, propagating inflammatory and degenerative diseases. Many drug development programs have pursued targeting RIPK1, and to a lesser extent RIPK3 kinase activity. In this review, we classify existing and novel small-molecule drugs based on their pharmacodynamic (PD) type I, II, and III binding mode. Finally, we discuss their applicability and therapeutic potential in inflammatory and degenerative experimental disease models.
Collapse
|
50
|
Graham DB, Xavier RJ. Pathway paradigms revealed from the genetics of inflammatory bowel disease. Nature 2020; 578:527-539. [PMID: 32103191 PMCID: PMC7871366 DOI: 10.1038/s41586-020-2025-2] [Citation(s) in RCA: 401] [Impact Index Per Article: 100.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022]
Abstract
Inflammatory bowel disease (IBD) is a complex genetic disease that is instigated and amplified by the confluence of multiple genetic and environmental variables that perturb the immune-microbiome axis. The challenge of dissecting pathological mechanisms underlying IBD has led to the development of transformative approaches in human genetics and functional genomics. Here we describe IBD as a model disease in the context of leveraging human genetics to dissect interactions in cellular and molecular pathways that regulate homeostasis of the mucosal immune system. Finally, we synthesize emerging insights from multiple experimental approaches into pathway paradigms and discuss future prospects for disease-subtype classification and therapeutic intervention.
Collapse
Affiliation(s)
- Daniel B Graham
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Center for Microbiome Informatics and Therapeutics, MIT, Cambridge, MA, USA.
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Center for Microbiome Informatics and Therapeutics, MIT, Cambridge, MA, USA.
| |
Collapse
|