1
|
Han B, Tang D, Lv X, Fan J, Li S, Zhu H, Zhang J, Xu S, Xu X, Huang Z, Huang Z, Lin G, Zhan L, Lv X. Integrated multi-omics reveal gut microbiota-mediated bile acid metabolism alteration regulating immunotherapy responses to anti-α4β7-integrin in Crohn's disease. Gut Microbes 2024; 16:2310894. [PMID: 38312103 PMCID: PMC10854365 DOI: 10.1080/19490976.2024.2310894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/23/2024] [Indexed: 02/06/2024] Open
Abstract
Gut microbiota and related metabolites are both crucial factors that significantly influence how individuals with Crohn's disease respond to immunotherapy. However, little is known about the interplay among gut microbiota, metabolites, Crohn's disease, and the response to anti-α4β7-integrin in current studies. Our research utilized 2,4,6-trinitrobenzene sulfonic acid to induce colitis based on the humanized immune system mouse model and employed a combination of whole-genome shotgun metagenomics and non-targeted metabolomics to investigate immunotherapy responses. Additionally, clinical cases with Crohn's disease initiating anti-α4β7-integrin therapy were evaluated comprehensively. Particularly, 16S-rDNA gene high-throughput sequencing and targeted bile acid metabolomics were conducted at weeks 0, 14, and 54. We found that anti-α4β7-integrin therapy has shown significant potential for mitigating disease phenotypes in remission-achieving colitis mice. Microbial profiles demonstrated that not only microbial composition but also microbially encoded metabolic pathways could predict immunotherapy responses. Metabonomic signatures revealed that bile acid metabolism alteration, especially elevated secondary bile acids, was a determinant of immunotherapy responses. Especially, the remission mice significantly enriched the proportion of the beneficial Lactobacillus and Clostridium genera, which were correlated with increased gastrointestinal levels of BAs involving lithocholic acid and deoxycholic acid. Moreover, most of the omics features observed in colitis mice were replicated in clinical cases. Notably, anti-α4β7 integrin provided sustained therapeutic benefits in clinical remitters during follow-up, and long-lasting remission was linked to persistent changes in the microbial-related bile acids. In conclusion, gut microbiota-mediated bile acid metabolism alteration could play a crucial role in regulating immunotherapy responses to anti-α4β7-integrin in Crohn's disease. Therefore, the identification of prognostic microbial signals facilitates the advancement of targeted probiotics that activate anti-inflammatory bile acid metabolic pathways, thereby improving immunotherapy responses. The integrated multi-omics established in our research provide valuable insights into potential mechanisms that impact treatment responses in complex diseases.
Collapse
Affiliation(s)
- Bing Han
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Daiyuan Tang
- Postgraduate College, Kunming Medical University, Kunming, China
| | - Xiaodan Lv
- Department of Clinical Experimental Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Junhua Fan
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shiquan Li
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hui Zhu
- Department of Microbiology, Guangxi Medical University, Nanning, China
| | - Jiatong Zhang
- Postgraduate College, Guangxi Medical University, Nanning, China
| | - Shang Xu
- Postgraduate College, Guangxi Medical University, Nanning, China
| | - Xiaofang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ziqian Huang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhixi Huang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guangfu Lin
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lingling Zhan
- Department of Clinical Experimental Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaoping Lv
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
2
|
He R, Zhao S, Cui M, Chen Y, Ma J, Li J, Wang X. Cutaneous manifestations of inflammatory bowel disease: basic characteristics, therapy, and potential pathophysiological associations. Front Immunol 2023; 14:1234535. [PMID: 37954590 PMCID: PMC10637386 DOI: 10.3389/fimmu.2023.1234535] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease typically involving the gastrointestinal tract but not limited to it. IBD can be subdivided into Crohn's disease (CD) and ulcerative colitis (UC). Extraintestinal manifestations (EIMs) are observed in up to 47% of patients with IBD, with the most frequent reports of cutaneous manifestations. Among these, pyoderma gangrenosum (PG) and erythema nodosum (EN) are the two most common skin manifestations in IBD, and both are immune-related inflammatory skin diseases. The presence of cutaneous EIMs may either be concordant with intestinal disease activity or have an independent course. Despite some progress in research on EIMs, for instance, ectopic expression of gut-specific mucosal address cell adhesion molecule-1 (MAdCAM-1) and chemokine CCL25 on the vascular endothelium of the portal tract have been demonstrated in IBD-related primary sclerosing cholangitis (PSC), little is understood about the potential pathophysiological associations between IBD and cutaneous EIMs. Whether cutaneous EIMs are inflammatory events with a commonly shared genetic background or environmental risk factors with IBD but independent of IBD or are the result of an extraintestinal extension of intestinal inflammation, remains unclear. The review aims to provide an overview of the two most representative cutaneous manifestations of IBD, describe IBD's epidemiology, clinical characteristics, and histology, and discuss the immunopathophysiology and existing treatment strategies with biologic agents, with a focus on the potential pathophysiological associations between IBD and cutaneous EIMs.
Collapse
Affiliation(s)
- Ronghua He
- Department of Gastroenterology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Subei Zhao
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingyu Cui
- Department of Gastroenterology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yanhao Chen
- Department of Gastroenterology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jinrong Ma
- Department of Gastroenterology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jintao Li
- Department of Gastroenterology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaodong Wang
- Department of Gastroenterology, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
3
|
Manzanares LD, David J, Ren X, Yalom LK, Piccolo EB, Dehghan Y, David AJ, Hanauer SB, Sumagin R. Atovaquone attenuates experimental colitis by reducing neutrophil infiltration of colonic mucosa. Front Pharmacol 2022; 13:1011115. [PMID: 36313299 PMCID: PMC9614091 DOI: 10.3389/fphar.2022.1011115] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/12/2022] [Indexed: 10/08/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic relapsing disease featuring aberrant accumulation of neutrophils in colonic mucosa and the luminal space. Although significant advances in UC therapy have been made with the development of novel biologics and small molecules targeting immune responses, success of most current therapies is still limited, with significant safety concerns. Thus, there is a need to develop additional safe and effective therapies for the treatment of UC. Antimalarial drugs have been safely used for many years to resolve tissue inflammation and the associated pathologies. Atovaquone is a recent FDA-approved antimalarial drug that has shown anti-viral and tumor-suppressive properties in vitro however, its role in mucosal inflammation has not been evaluated. Using pre-clinical murine DSS-induced colitis model combined with complementary in vivo peritonitis and ex vivo human neutrophil activation and chemotaxis assays we investigated functional and mechanistic impacts of atovaquone on disease resolution and neutrophil trafficking. We demonstrate that atovaquone promotes resolution of DSS-induced murine colitis by reducing neutrophil accumulation in the inflamed colonic mucosa. Mechanistically, we show that atovaquone suppressed induction of CD11b expression in neutrophils, reducing their polarization and migratory ability. Thus, our findings identify a new role of atovaquone in promoting resolution of mucosal inflammation, supporting the idea of potential repurposing of this FDA-approved drug as UC therapeutic.
Collapse
Affiliation(s)
- Laura D. Manzanares
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Joseph David
- Department of Medicine, Gastroenterology and Hepatology University of Arizona College of Medicine, Phoenix, AZ, United States
| | - Xingsheng Ren
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Lenore K. Yalom
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Enzo B. Piccolo
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Yalda Dehghan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Aidan J. David
- College of Arts and Sciences, Case Western Reserve Unviersity, Cleveland, OH, United States
| | - Stephen B. Hanauer
- Department of Medicine, Gastroenterology and Hepatology Northwestern Memorial Hospital, Chicago, IL, United States
| | - Ronen Sumagin
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
4
|
Martina MG, Giorgio C, Allodi M, Palese S, Barocelli E, Ballabeni V, Szpakowska M, Chevigne A, Piet van Hamburg J, Davelaar N, Lubberts E, Bertoni S, Radi M. Discovery of small-molecules targeting the CCL20/CCR6 axis as first-in-class inhibitors for inflammatory bowel diseases. Eur J Med Chem 2022; 243:114703. [DOI: 10.1016/j.ejmech.2022.114703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/03/2022]
|
5
|
Leoncini G, Gentili M, Lusenti E, Caruso L, Calafà C, Migliorati G, Riccardi C, Villanacci V, Ronchetti S. The novel role of glucocorticoid-induced leucine zipper as a marker of mucosal healing in inflammatory bowel diseases. Pharmacol Res 2022; 182:106353. [PMID: 35835370 DOI: 10.1016/j.phrs.2022.106353] [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: 05/11/2022] [Revised: 06/22/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023]
Abstract
Glucocorticoid-induced leucin zipper (GILZ) mediates the effects of glucocorticoids in immune cells, but little is known about its role in both the gastro-intestinal (GI) mucosa and inflammatory bowel diseases (IBD) in humans. To investigate the GILZ protein expression profile in the GI tract, mucosal biopsies from 80 patients were retrospectively enrolled in this study and subdivided into three groups: 1) patients without clinical-endoscopic and histological evidence of IBD; 2) IBD patients; 3) patients with chronic atrophic gastritis (CAG) and Barrett esophagus (BE), both characterized by intestinal metaplasia (IM). GILZ expression was assessed by immunohistochemical and immunofluorescence methods. Our results showed that GILZ protein was strongly expressed in the secretory cells in healthy mucosa. GILZ expression was reduced in goblet cells in active disease, whereas it was restored in quiescent diseases. Conversely, entero-endocrine cells were not involved in such inflammation-driven dynamics, as GILZ expression remained detectable in active disease. Moreover, GILZ was expressed in IM, but was limited to CAG, and was not detected in BE. In summary, GILZ acts as a secretory protein in the GI mucosa in healthy, hyperplastic and metaplastic conditions. Its secretion by goblet cells is mostly affected by neutrophils mucosal infiltration and seems to be directly related to active mucosal inflammation in IBD. Overall, our findings suggest that GILZ is a suitable molecule to be considered as a histological marker of mucosal healing.
Collapse
Affiliation(s)
- Giuseppe Leoncini
- Pathology Unit, Department of Pathology and Laboratory Medicine, ASST del Garda Desenzano del Garda, Brescia, Italy
| | - Marco Gentili
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Eleonora Lusenti
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Laura Caruso
- Pathology Unit, Department of Pathology and Laboratory Medicine, ASST del Garda Desenzano del Garda, Brescia, Italy
| | - Cristina Calafà
- Pathology Unit, Department of Pathology and Laboratory Medicine, ASST del Garda Desenzano del Garda, Brescia, Italy
| | - Graziella Migliorati
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Carlo Riccardi
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Simona Ronchetti
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| |
Collapse
|
6
|
Hu W, Fang T, Chen X. Identification of Differentially Expressed Genes and miRNAs for Ulcerative Colitis Using Bioinformatics Analysis. Front Genet 2022; 13:914384. [PMID: 35719390 PMCID: PMC9201719 DOI: 10.3389/fgene.2022.914384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: Ulcerative colitis (UC) is a chronic inflammatory disease of the intestine whose cause and underlying mechanisms are not fully understood. The aim of this study was to use bioinformatics analysis to identify differentially expressed genes (DEGs) with diagnostic and therapeutic potential in UC.Materials and methods: Three UC datasets (GSE179285, GSE75214, GSE48958) were downloaded from the Gene Expression Omnibus (GEO) database. DEGs between normal and UC tissues were identified using the GEO2R online tool. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of the DEGs were performed using Metascape. Protein-protein interaction network (PPI) analysis and visualization using STRING and Cytoscape. Finally, the miRNA gene regulatory network was constructed by Cytoscape to predict potential microRNAs (miRNAs) associated with DEGs.Results: A total of 446 DEGs were identified, consisting of 309 upregulated genes and 137 downregulated genes. The enriched functions and pathways of the DEGs include extracellular matrix, regulation of cell adhesion, inflammatory response, response to cytokine, monocarboxylic acid metabolic process, response to toxic substance. The analysis of KEGG pathway indicates that the DEGs were significantly enriched in Complement and coagulation cascades, Amoebiasis, TNF signaling pathway, bile secretion, and Mineral absorption. Combining the results of the PPI network and CytoHubba, 9 hub genes including CXCL8, ICAM1, CXCR4, CD44, IL1B, MMP9, SPP1, TIMP1, and HIF1A were selected. Based on the DEG-miRNAs network construction, 7 miRNAs including miR-335-5p, mir-204-5p, miR-93-5p, miR106a-5p, miR-21-5p, miR-146a-5p, and miR-155-5p were identified as potential critical miRNAs.Conclusion: In summary, we identified DEGs that may be involved in the progression or occurrence of UC. A total of 446 DEGs,9 hub genes and 7 miRNAs were identified, which may be considered as biomarkers of UC. Further studies, however, are needed to elucidate the biological functions of these genes in UC.
Collapse
Affiliation(s)
- Weitao Hu
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Taiyong Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xiaoqing Chen
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- *Correspondence: Xiaoqing Chen,
| |
Collapse
|
7
|
Cottle C, Anbazhagan M, Lipat A, Patel M, Porter A, Hogan K, Rajan D, Matthews J, Kugathasan S, Chinnadurai R. Complexity of Secretory Chemokines in Human Intestinal Organoid Cultures Ex Vivo. GASTRO HEP ADVANCES 2022; 1:457-460. [PMID: 35634262 PMCID: PMC9141070 DOI: 10.1016/j.gastha.2022.02.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/09/2022] [Indexed: 12/27/2022]
Affiliation(s)
- C. Cottle
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia
| | - M. Anbazhagan
- Division of Pediatric Gastroenterology, Department of Pediatrics, Emory University School of Medicine & Children’s Healthcare of Atlanta, Atlanta, Georgia
| | - A. Lipat
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia
| | - M. Patel
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia
| | - A.P. Porter
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia
| | - K. Hogan
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia
| | - D. Rajan
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia
| | - J.D. Matthews
- Division of Pediatric Gastroenterology, Department of Pediatrics, Emory University School of Medicine & Children’s Healthcare of Atlanta, Atlanta, Georgia
| | - S. Kugathasan
- Division of Pediatric Gastroenterology, Department of Pediatrics, Emory University School of Medicine & Children’s Healthcare of Atlanta, Atlanta, Georgia
| | - R. Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia
| |
Collapse
|
8
|
Jiang L, Chi C, Yuan F, Lu M, Hu D, Wang L, Liu X. Anti-inflammatory effects of anemonin on acute ulcerative colitis via targeted regulation of protein kinase C-θ. Chin Med 2022; 17:39. [PMID: 35346284 PMCID: PMC8962473 DOI: 10.1186/s13020-022-00599-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/18/2022] [Indexed: 11/10/2022] Open
Abstract
Background Ulcerative colitis (UC) is an inflammatory bowel disease that causes continuous mucosal inflammation. Anemonin is a natural molecule from the Ranunculaceae and Gramineae plants that exerts anti-inflammatory properties. This study aimed to explore the effects and mechanisms of anemonin on UC. Methods C57BL/6 mice were administered dextran sulphate sodium (DSS; 3% [w/v]) to establish an animal model of UC. Mice were treated with an intraperitoneal injection of anemonin. Body weight and the disease activity index (DAI) were recorded. Haematoxylin and eosin staining, RT-qPCR, ELISA, and western blotting were performed to evaluate the histopathological changes and tissue inflammation. HT-29 cells were treated with lipopolysaccharide (LPS) and anemonin. Cell inflammation was evaluated using RT-qPCR and western blotting. The target proteins of anemonin were predicted using bioinformatics analysis and confirmed in vitro and in vivo. Results Anemonin improved DSS-induced body weight loss, shortened colon length, increased DAI, and induced pathological changes in the colon tissue of mice. Anemonin inhibited DSS-induced colon tissue inflammation as the release of IL-1β, TNF-α, and IL-6 was significantly suppressed. Additionally, anemonin attenuated LPS-induced cytokine production in HT-29 cells. PKC-θ was predicted as a target protein of anemonin. Anemonin did not affect PRKCQ gene transcription, but inhibited its translation. PRKCQ overexpression partially reversed the protective effects of anemonin on HT-29 cells. Adeno-associated virus delivery of the PRKCQ vector significantly reversed the protective effects of anemonin on the mouse colon. Conclusions Anemonin has the potential to treat UC. The anti-inflammatory effects of anemonin may be mediated through targeting PKC-θ.
Collapse
Affiliation(s)
- Lu Jiang
- Department of Gastroenterology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No.42 Wenhua west road, Jinan, 250011, Shandong, China.
| | - Chunhua Chi
- Department of Anorectal Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, Shandong, China
| | - Fang Yuan
- Department of Gastrology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, Shandong, China
| | - Meiqi Lu
- Department of Gastroenterology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No.42 Wenhua west road, Jinan, 250011, Shandong, China
| | - Dongqing Hu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, Shandong, China
| | - Lin Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, Shandong, China
| | - Xiaoming Liu
- Department of Geriatrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No.42 Wenhua west road, Jinan, 250011, Shandong, China.
| |
Collapse
|
9
|
Anti-Inflammatory and Immunoregulatory Action of Sesquiterpene Lactones. Molecules 2022; 27:molecules27031142. [PMID: 35164406 PMCID: PMC8839508 DOI: 10.3390/molecules27031142] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/01/2022] [Accepted: 02/06/2022] [Indexed: 01/21/2023] Open
Abstract
Sesquiterpene lactones (SL), characterized by their high prevalence in the Asteraceae family, are one of the major groups of secondary metabolites found in plants. Researchers from distinct research fields, including pharmacology, medicine, and agriculture, are interested in their biological potential. With new SL discovered in the last years, new biological activities have been tested, different action mechanisms (synergistic and/or antagonistic effects), as well as molecular structure–activity relationships described. The review identifies the main sesquiterpene lactones with interconnections between immune responses and anti-inflammatory actions, within different cellular models as well in in vivo studies. Bioaccessibility and bioavailability, as well as molecular structure–activity relationships are addressed. Additionally, plant metabolic engineering, and the impact of sesquiterpene lactone extraction methodologies are presented, with the perspective of biological activity enhancement. Sesquiterpene lactones derivatives are also addressed. This review summarizes the current knowledge regarding the therapeutic potential of sesquiterpene lactones within immune and inflammatory activities, highlighting trends and opportunities for their pharmaceutical/clinical use.
Collapse
|
10
|
Honig G, Larkin PB, Heller C, Hurtado-Lorenzo A. Research-Based Product Innovation to Address Critical Unmet Needs of Patients with Inflammatory Bowel Diseases. Inflamm Bowel Dis 2021; 27:S1-S16. [PMID: 34791292 PMCID: PMC8922161 DOI: 10.1093/ibd/izab230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Indexed: 12/09/2022]
Abstract
Despite progress in recent decades, patients with inflammatory bowel diseases face many critical unmet needs, demonstrating the limitations of available treatment options. Addressing these unmet needs will require interventions targeting multiple aspects of inflammatory bowel disease pathology, including disease drivers that are not targeted by available therapies. The vast majority of late-stage investigational therapies also focus primarily on a narrow range of fundamental mechanisms. Thus, there is a pressing need to advance to clinical stage differentiated investigational therapies directly targeting a broader range of key mechanistic drivers of inflammatory bowel diseases. In addition, innovations are critically needed to enable treatments to be tailored to the specific underlying abnormal biological pathways of patients; interventions with improved safety profiles; biomarkers to develop prognostic, predictive, and monitoring tests; novel devices for nonpharmacological approaches such as minimally invasive monitoring; and digital health technologies. To address these needs, the Crohn's & Colitis Foundation launched IBD Ventures, a venture philanthropy-funding mechanism, and IBD Innovate®, an innovative, product-focused scientific conference. This special IBD Innovate® supplement is a collection of articles reflecting the diverse and exciting research and development that is currently ongoing in the inflammatory bowel disease field to deliver innovative and differentiated products addressing critical unmet needs of patients. Here, we highlight the pipeline of new product opportunities currently advancing at the preclinical and early clinical development stages. We categorize and describe novel and differentiated potential product opportunities based on their potential to address the following critical unmet patient needs: (1) biomarkers for prognosis of disease course and prediction/monitoring of treatment response; (2) restoration of eubiosis; (3) restoration of barrier function and mucosal healing; (4) more effective and safer anti-inflammatories; (5) neuromodulatory and behavioral therapies; (6) management of disease complications; and (7) targeted drug delivery.
Collapse
|
11
|
Giussani P, Prinetti A, Tringali C. The Role of Sphingolipids in Cancer Immunotherapy. Int J Mol Sci 2021; 22:ijms22126492. [PMID: 34204326 PMCID: PMC8234743 DOI: 10.3390/ijms22126492] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 01/04/2023] Open
Abstract
Immunotherapy is now considered an innovative and strong strategy to beat metastatic, drug-resistant, or relapsing tumours. It is based on the manipulation of several mechanisms involved in the complex interplay between cancer cells and immune system that culminates in a form of immune-tolerance of tumour cells, favouring their expansion. Current immunotherapies are devoted enforcing the immune response against cancer cells and are represented by approaches employing vaccines, monoclonal antibodies, interleukins, checkpoint inhibitors, and chimeric antigen receptor (CAR)-T cells. Despite the undoubted potency of these treatments in some malignancies, many issues are being investigated to amplify the potential of application and to avoid side effects. In this review, we discuss how sphingolipids are involved in interactions between cancer cells and the immune system and how knowledge in this topic could be employed to enhance the efficacy of different immunotherapy approaches. In particular, we explore the following aspects: how sphingolipids are pivotal components of plasma membranes and could modulate the functionality of surface receptors expressed also by immune cells and thus their functionality; how sphingolipids are related to the release of bioactive mediators, sphingosine 1-phosphate, and ceramide that could significantly affect lymphocyte egress and migration toward the tumour milieu, in addition regulating key pathways needed to activate immune cells; given the renowned capability of altering sphingolipid expression and metabolism shown by cancer cells, how it is possible to employ sphingolipids as antigen targets.
Collapse
|
12
|
Ma G, Du H, Hu Q, Yang W, Pei F, Xiao H. Health benefits of edible mushroom polysaccharides and associated gut microbiota regulation. Crit Rev Food Sci Nutr 2021; 62:6646-6663. [PMID: 33792430 DOI: 10.1080/10408398.2021.1903385] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Edible mushrooms have been an important part of the human diet for thousands of years, and over 100 varieties have been cultivated for their potential human health benefits. In recent years, edible mushroom polysaccharides (EMPs) have been studied for their activities against obesity, inflammatory bowel disease (IBD), and cancer. Particularly, accumulating evidence on the exact causality between these health risks and specific gut microbiota species has been revealed and characterized, and most of the beneficial health effects of EMPs have been associated with its reversal impacts on gut microbiota dysbiosis. This demonstrates the key role of EMPs in decreasing health risks through gut microbiota modulation effects. This review article compiles and summarizes the latest studies that focus on the health benefits and underlying functional mechanisms of gut microbiota regulation via EMPs. We conclude that EMPs can be considered a dietary source for the improvement and prevention of several health risks, and this review provides the theoretical basis and technical guidance for the development of novel functional foods with the utilization of edible mushrooms.
Collapse
Affiliation(s)
- Gaoxing Ma
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing, People's Republic of China.,Department of Food Science, University of Massachusetts, Amherst, Massachusetts, USA
| | - Hengjun Du
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts, USA
| | - Qiuhui Hu
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing, People's Republic of China
| | - Wenjian Yang
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing, People's Republic of China
| | - Fei Pei
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing, People's Republic of China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts, USA
| |
Collapse
|