1
|
Wang X, Bao H, Wang Y, Wang Y, Guo C, Wu Y, Xu Y, Li Y. Innovative peptide therapeutics targeting IL17RA to regulate inflammatory responses. Sci Rep 2025; 15:8542. [PMID: 40121226 PMCID: PMC11929782 DOI: 10.1038/s41598-025-92915-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
Interleukin-17 receptor A (IL17RA) is a critical mediator of pro-inflammatory cytokine signaling and a key immune checkpoint in autoimmune diseases. While monoclonal antibodies targeting IL17RA have demonstrated clinical efficacy, their high costs, complexity in production, and lack of oral bioavailability present significant limitations. In response to these challenges, we developed AL-8(0), a novel peptide specifically designed to inhibit the IL17A-IL17RA signaling pathway. AL-8(0) was synthesized with high purity and systematically evaluated for its binding affinity and anti-inflammatory activity. Biophysical and cellular assays confirmed the peptide's strong affinity for IL17RA and its ability to inhibit inflammatory cytokine production in IL17RA-expressing monocyte-macrophages and keratinocytes. Moreover, its anti-inflammatory effects were com-parable to IL17RA-targeting monoclonal antibodies and were dependent on IL17RA expression, as demonstrated by experiments using IL17RA-deficient cells. These results underscore AL-8(0)'s potential as a targeted therapeutic for autoimmune diseases, offering a peptide-based alternative with lower antigenicity, improved scalability, and potential for oral administration. This study lays the groundwork for further development of AL-8(0) and similar peptides as innovative treatments for inflammatory disorders driven by the IL17A-IL17RA pathway.
Collapse
Affiliation(s)
- Xinmin Wang
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
| | - Hang Bao
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
| | - Yuya Wang
- Inner Mongolia Autonomous Region People's Hospital, Hohhot, 010010, China
| | - Yalu Wang
- The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014010, China
| | - Cheng Guo
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
| | - Yanning Wu
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
| | - Yongbin Xu
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
| | - Yali Li
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China.
| |
Collapse
|
2
|
Saviano A, Apta B, Tull S, Pezhman L, Fatima A, Sevim M, Mete A, Chimen M, Schettino A, Marigliano N, McGettrick HM, Iqbal AJ, Maione F, Rainger GE. PEPITEM, its tripeptide pharmacophores and their peptidomimetic analogues regulate the inflammatory response through parenteral and topical dosing in models of peritonitis and psoriasis. Pharmacol Res 2025; 213:107624. [PMID: 39855372 DOI: 10.1016/j.phrs.2025.107624] [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: 11/28/2024] [Revised: 01/02/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
PEPITEM is an immune-modulatory peptide that effectively regulates inflammation and mitigates immune-mediated inflammatory diseases (IMIDs). Here, we identify two independently active tripeptide pharmacophores within PEPITEM and engineered peptidomimetics with enhanced pharmacodynamic properties. These peptidomimetics regulate T-cell trafficking in vitro and reduce T-cell, neutrophil and macrophage numbers in the inflamed peritoneal cavity in vivo. In a plaque psoriasis model, topical administration reduced disease severity, inflammation and immune cell infiltration, while regulating cytokine release in macrophages and fibroblasts, as well as keratinocyte proliferation. Th1 and Th17 cell abundance, along with their cytokines, was reduced in secondary lymphoid organs. This expanded functional repertoire of PEPITEM and its derivatives provides innovative tools for countering immune and stromal cell-induced pathology in IMIDs. Moreover, by identifying significantly smaller tripeptide pharmacophores of 14 amino acid PEPITEM, we may be able to deliver substantial financial advantages in synthesis and formulation. The order of magnitude increase in efficacy observed for some peptidomimetics may deliver agents with enhanced pharmacological characteristics compared to the parent PEPITEM sequence. Taken together with other reports on the efficacy of PEPITEM, this study paves the way for the development and translation of a novel class of anti-inflammatory agents which may have utility in a broad range of autoimmune and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, Naples 80131, Italy.
| | - Bonita Apta
- Department of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Samantha Tull
- Department of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Laleh Pezhman
- Department of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Areeba Fatima
- Department of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Mustafa Sevim
- Department of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Antonio Mete
- Medsyndesign Ltd, Advanced Technology Innovation Centre, 5 Oakwood Drive, Loughborough LE11 3QF, UK.
| | - Myriam Chimen
- Department of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK.
| | - Anna Schettino
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, Naples 80131, Italy.
| | - Noemi Marigliano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, Naples 80131, Italy.
| | - Helen M McGettrick
- Department of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK.
| | - Asif J Iqbal
- Department of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, Naples 80131, Italy.
| | - G Ed Rainger
- Department of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
3
|
Duarte LG, Laurindo LF, Bishayee A, Casarcia N, Detregiachi CRP, Otoboni AMM, de Alvares Goulart R, Catharin VMCS, Baldi E, Catharin VCS, Guiguer EL, Sanches Silva A, Barbalho SM, Bishayee A. Mango (Mangifera indica L.) By-products in Food Processing and Health Promotion. Nutr Rev 2025:nuae214. [PMID: 39903188 DOI: 10.1093/nutrit/nuae214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025] Open
Abstract
The edible and nonedible parts of the mango (Mangifera indica L.) contain vitamins, phytocompounds, fiber, and fatty acids. This review highlights the uses of mango by-products in the food industry and their effects on human health. The literature offers many new possibilities for the usage of mango secondary products in the food industry, such as the production of functional foods and bakery products, in addition to the potential for extraction of antioxidants and enzymes. Furthermore, due to their antioxidant and anti-inflammatory properties, the consumption of various mango by-products, in the form of peel and leaf (powder or extract), can improve glycemia, plasma lipid levels, satiety, and endothelial function, suggesting that these compounds can prevent or improve various risk factors for cardiovascular complications and metabolic syndrome. Clinical trials show that the discarded parts of mango fruits and leaves can be used to treat diabetes mellitus, obesity, and cardiovascular disorders. Moreover, mango by-products can be utilized to improve the functional characteristics of foods, may be incorporated as fat replacers, and have the potential to leverage agribusiness and reduce environmental damage resulting from the disposal of discarded materials, in addition to reducing waste and the complex chain of environmental damage. Mango by-products also have the potential to produce nutraceutical food items. The use of new technologies can bring to light the production of numerous products made from by-products, contributing to the development of industrial functional foods. In addition, products for the pharmaceutical and cosmetics industries may also be developed. Nutraceutical and pharmaceutical products could have lower prices and could, therefore, be used by low-income populations. The utilization of mango by-products meets the current trend and growing market for better and healthier products. However, more clinical trials are necessary to evaluate the effectiveness of mango by-products on human health, and new technologies can improve industrial applications.
Collapse
Affiliation(s)
- Lidiane Gonsalves Duarte
- Department of Biochemistry and Nutrition, School of Food and Technology of Marilia (FATEC), Marilia, São Paulo 17500-000, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília, São Paulo 17519-030, Brazil
| | | | - Nicolette Casarcia
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, United States
| | - Claudia Rucco P Detregiachi
- Department of Biochemistry and Pharmacology, Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília, Sao Paulo 17012-150, Brazil
| | - Alda Maria M Otoboni
- Department of Biochemistry and Nutrition, School of Food and Technology of Marilia (FATEC), Marilia, São Paulo 17500-000, Brazil
| | - Ricardo de Alvares Goulart
- Department of Biochemistry and Pharmacology, Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília, Sao Paulo 17012-150, Brazil
| | - Virgínia Maria Cavallari Strozze Catharin
- Department of Biochemistry and Pharmacology, Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília, Sao Paulo 17012-150, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, São Paulo 17012-150, Brazil
| | - Edgar Baldi
- Department of Biochemistry and Pharmacology, Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília, Sao Paulo 17012-150, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, São Paulo 17012-150, Brazil
| | - Vitor Cavallari Strozze Catharin
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, São Paulo 17012-150, Brazil
| | - Elen Landgraf Guiguer
- Department of Biochemistry and Nutrition, School of Food and Technology of Marilia (FATEC), Marilia, São Paulo 17500-000, Brazil
- Department of Biochemistry and Pharmacology, Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília, Sao Paulo 17012-150, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, São Paulo 17012-150, Brazil
| | - Ana Sanches Silva
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Study in Animal Science (CECA), University of Oporto, 4501-401 Oporto, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Sandra Maria Barbalho
- Department of Biochemistry and Nutrition, School of Food and Technology of Marilia (FATEC), Marilia, São Paulo 17500-000, Brazil
- Department of Biochemistry and Pharmacology, Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília, Sao Paulo 17012-150, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, São Paulo 17012-150, Brazil
- Medical School of Marília, Research Coordinator, Hospital Beneficente UNIMAR, Marilia, São Paulo, Brazil
| | - Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, United States
| |
Collapse
|
4
|
Mahroum N, Elsalti A, Al Shawaf M, Darkhabani M, Alwani A, Seida R, Ertas MT, Simsek AG, Awad M, Habra M, Alrifaai MA, Bogdanos D, Shoenfeld Y. Artificial intelligence meets the world experts; updates and novel therapies in autoimmunity - The 14th international congress on autoimmunity 2024 (AUTO14), Ljubljana. Autoimmun Rev 2025; 24:103698. [PMID: 39571671 DOI: 10.1016/j.autrev.2024.103698] [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: 06/21/2024] [Revised: 11/16/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024]
Abstract
The bi-annual international congress on autoimmunity is a huge opportunity for the medical community to discuss the latest updates in the field. During the 14th congress 2024 (AUTO14) in Ljubljana, artificial intelligence (AI) occupied special attention due to its recent and ongoing unequivocal role in various medical fields including autoimmunity. For instance, through a challenging debate between world-experts and the most popular AI bot used (ChatGPT), several clinical cases including a case of vasculitis were discussed in the plenary sessions. ChatGPT agreed with the clinical decisions made by the experts nevertheless, the bot added additional aspects related to the specific case. In this regard, ChatGPT emphasized the need for osteoporosis prophylaxis in a patient planned to be treated with systemic steroids for a long time. Furthermore, AUTO14 included the newest updates on most autoimmune disorders, distributed among tens of sessions. Among others, infection and autoimmunity, the sequalae of the pandemic of COVID-19, as well as COVID-19 vaccines were discussed as well. Due to the high numbers of the works presented, and for ensuring that important updates are not missed; we divided our paper into sections. The subtitles throughout the paper correspond to different sessions of the congress, all presenting new updates in the field. A figure aiding in navigating throughout the paper was also provided.
Collapse
Affiliation(s)
- Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey.
| | - Abdulrahman Elsalti
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Maisam Al Shawaf
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Mohammad Darkhabani
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Abdulrahman Alwani
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ravend Seida
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | | | | | - Mustafa Awad
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Mona Habra
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | | | - Dimitrios Bogdanos
- Department of Rheumatology and Clinical Immunology, University Hospital of Larisa, School of Medicine, University of Thessaly, Larissa, Greece
| | - Yehuda Shoenfeld
- Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel; Reichman University, Herzliya, Israel
| |
Collapse
|
5
|
Li T, Liu XM, Yang M, Wu YQ, Liu W, Mao B, Jiang HL. Efficacy and Safety of Zihua Wenfei Zhisou Granule in Treatment of Postinfectious Cough: A Multicenter, Randomized, Double-Blind, Placebo-Controlled, Phase II Clinical Trial. Chin J Integr Med 2025; 31:3-10. [PMID: 39576445 DOI: 10.1007/s11655-024-3918-y] [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] [Accepted: 10/11/2023] [Indexed: 12/28/2024]
Abstract
OBJECTIVE To evaluate the efficacy and safety of Chinese medicine (CM) Zihua Wenfei Zhisou Granule (ZWZG) in postinfectious cough (PIC) patients with CM syndrome of wind-cold invading Fei (Lung, WCIF). METHODS This is a multicenter, randomized, double-blind, parallel-group, placebo-controlled phase II clinical trial. PIC patients with WCIF syndrome were recruited from the Respiratory Departments in 6 hospitals across China between March 2019 and December 2020. Eligible patients were randomly assigned to group A (ZWZG-matched placebo 15 g), group B (active ZWZG 15 g), and group C (active ZWZG 10 g plus ZWZG-matched placebo 5 g) in a 1:1:1 ratio. All medications were taken orally 3 times daily for 14 consecutive days. The primary outcomes were cough relief rate and cough disappearance rate. The secondary outcomes included time to cough relief, time to cough disappearance, and changes in cough symptom score (CSS), cough Visual Analog Scale (VAS) value, Cough-Specific Quality of Life Questionnaire (CQLQ) score, and CM syndrome score from baseline (day 0) to post-treatment (day 14). Adverse events (AEs) in each group were recorded. RESULTS A total of 198 patients were included in the full analysis set (FAS) and safety analysis set (SS), while 183 were enrolled in the per-protocol analysis set (PPS). In the FAS population, the cough relief rate was 47.76%, 90.77% and 84.85% in groups A, B, and C, respectively; while the cough disappearance rate was 31.34%, 72.31% and 68.18%, respectively. The cough relief rates and cough disappearance rates in groups B and C were significantly higher than group A (P<0.0001). Both the median time to cough relief and cough disappearance in groups B and C were shorter than group A (P<0.0001). Compared with group A, groups B and C showed significantly greater improvements from baseline to post-treatment in CSS during daytime and nighttime as well as VAS (P<0.05). There were no significant differences in changes from baseline to post-treatment in CQLQ and CM syndrome scores among 3 groups (P>0.05). Results in the PPS population were consistent with those in the FAS population. Groups B and C showed lower incidence in AEs than group A (P<0.05), while there was no significant difference between groups B and C (P>0.05). No drug-related severe AEs were reported. CONCLUSIONS ZWZG can increase cough disappearance rate and cough relief rate; and it is beneficial in shortening cough duration and reducing cough severity and frequency in patients suffering from PIC. It is safe and generally well tolerated. (Registration No. ChiCTR1900022078).
Collapse
Affiliation(s)
- Ting Li
- Division of Pulmonary Medicine, Department of Internal Medicine, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xue-Mei Liu
- Division of Pulmonary Medicine, Department of Internal Medicine, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mei Yang
- Division of Pulmonary Medicine, Department of Internal Medicine, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yan-Qing Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei Liu
- Division of Pulmonary Medicine, Department of Internal Medicine, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bing Mao
- Division of Pulmonary Medicine, Department of Internal Medicine, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong-Li Jiang
- Division of Pulmonary Medicine, Department of Internal Medicine, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
6
|
Iannuzzo F, Schiano E, Maisto M, Schettino A, Marigliano N, Saviano A, Abo Mansour A, Iqbal AJ, Maione F, Tenore GC, Novellino E. Effect of Proanthocyanidins from Grape Seed Extract on Benign Prostatic Hyperplasia. Nutrients 2024; 17:73. [PMID: 39796507 PMCID: PMC11723264 DOI: 10.3390/nu17010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/23/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Benign prostatic hyperplasia (BPH) is one of the most common chronic diseases affecting the urinary tract that occurs mainly in men over 40 years of age. Among the natural therapies, proanthocyanidins (PACs), which can treat a wide range of immune-mediated inflammatory diseases (IMIDs), have been shown to play an important role in the treatment of pathologies concerning prostate health. In this regard, the present study aimed to evaluate the different bioactivities of a grape seed extract (GSE), rich in polymeric PACs, and its version processed under alkaline conditions (ATGSE), characterized by a higher content of oligomeric PACs, in an animal model of BPH induced by subcutaneous injection of testosterone (1 mg/mouse). METHODS These latter were divided into a control group (vehicle, olive oil), a BPH group (testosterone 1 mg/mouse), and four treatment groups treated with GSE (500 mg/kg) and ATGSE (125, 250, 500 mg/kg) by oral gavage. At the experimental endpoint (4 weeks), hematological and biochemical analyses of blood and tissues were performed. RESULTS Data showed that oral administration of ATGSE (250 mg/kg) was significantly more effective than GSE in reducing prostate (p ≤ 0.0001) and seminal vesicle (p ≤ 0.0001) weight. Moreover, ATGSE exhibited enhanced effectiveness in significantly reducing PSA levels (p ≤ 0.0001 vs. GSE) and the expression of key pro-inflammatory cyto-chemokines in prostate and seminal vesicles homogenates. CONCLUSIONS These findings pave the way for the clinical application of ATGSE as a nutraceutical and/or functional food.
Collapse
Affiliation(s)
- Fortuna Iannuzzo
- Department of Pharmacy, University of Chieti-Pescara G. D’Annunzio, 66100 Chieti, Italy;
| | - Elisabetta Schiano
- Inventia Biotech—Healthcare Food Research Center s.r.l., Strada Statale Sannitica KM 20.700, 81020 Caserta, Italy;
| | - Maria Maisto
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy;
| | - Anna Schettino
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy; (A.S.); (N.M.); (A.S.); (A.J.I.)
| | - Noemi Marigliano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy; (A.S.); (N.M.); (A.S.); (A.J.I.)
| | - Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy; (A.S.); (N.M.); (A.S.); (A.J.I.)
| | - Adel Abo Mansour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 62521, Saudi Arabia;
| | - Asif Jilani Iqbal
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy; (A.S.); (N.M.); (A.S.); (A.J.I.)
- Department of Cardiovascular Sciences, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy; (A.S.); (N.M.); (A.S.); (A.J.I.)
| | - Gian Carlo Tenore
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy;
| | - Ettore Novellino
- Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy;
| |
Collapse
|
7
|
Xie Q, Sun J, Sun M, Wang Q, Wang M. Perturbed microbial ecology in neuromyelitis optica spectrum disorder: Evidence from the gut microbiome and fecal metabolome. Mult Scler Relat Disord 2024; 92:105936. [PMID: 39418776 DOI: 10.1016/j.msard.2024.105936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Neuromyelitis optica spectrum disorder (NMOSD) is a central nervous system inflammatory demyelinating immune-mediated ailment, which is influenced by genetic, epigenetic, and environmental elements. The escalating incidence of NMOSD in recent years implies alterations in environmental risk factors. Recent research has established a correlation between gut microbiomes and the development of NMOSD. METHODS Metagenomic shotgun sequencing and gas chromatography-mass spectrometry (GC-MS) were employed to assess alterations of the structure and function in the fecal microbiome, as well as levels of short-chain fatty acids (SCFAs) in fecal and blood samples, among individuals with neuromyelitis optica spectrum disorder (NMOSD) during the acute phase (n = 25), the remission phase (n = 11), and a group of healthy controls (HCs) (n = 24). We further explored the correlation between gut microbiota and the pathogenesis of NMOSD through fecal microbiota transplantation (FMT). The gut microbiome from human donors diagnosed with NMOSD or HCs was transplanted into germ-free mice, followed by an analysis of the alterations in the structure and functionality of the transplanted mice's gut microbiome. Additionally, the impact of microbiome transfer on the immunity and spinal cord of germ-free mice was assessed through various techniques, including ELISA, flow cytometry, western blot, histopathology, and transcriptome sequencing. RESULTS (1) At the taxonomic levels of genus and species, there were significant differences in the α-diversity of the microbiome between HCs and NMOSD patients in the acute phase, with NMOSD patients having higher species diversity. (2) In the acute phase, the gut microbiota of NMOSD patients was characterized by Ruminococcaceae_unclassified, Campylobacter, Parabacteroides, Lactobacillus, Akkermansia, Streptococcus oralis, Clostridium leptum, Clostridium asparagiforme, Firmicutes bacterium CAG 238, and Lactobacillus fermentum. (3) The relative abundances of Coprobacter, Turicimonas, Gemmiger, Enterobacter, Roseburia sp.CAG 471, Veillonella tobetsuensis, Proteobacteria bacterium CAG 139, Ruminococcus bicirculans, Lactococcus lactis, Flavonifractor plautii, and Streptococcus cristatus were notably lower in patients experiencing remission compared to NMOSD patients in the acute phase, On the other hand, the relative abundances of Flavonifractor (P = 0.049) and Clostridium aldenense (P = 0.049) were significantly higher. Following medication, the gut microbiome distribution in NMOSD patients during remission closely resembled that of healthy controls (HCs). (4) Compared with HCs, acetate levels in the feces of patients with NMOSD in the acute phase were significantly lower. (5) In addition, we transplanted feces from NMOSD patients into germ-free mice and revealed a significant increase in the levels of IL-6, IL-17A, and IL-23 in the blood of mice belonging to the NMOSD fecal transplantation (NFMT) group. Additionally, the IL-10 level exhibited a significant reduction. Moreover, the proportion of Th17 cells displayed a significant increase, while the proportion of Treg cells exhibited a significant decrease in the spleens of NFMT mice. CONCLUSION Patients in the acute phase of neuromyelitis optica spectrum disorder (NMOSD) exhibited imbalances in their gut microbiota and a deficiency in short-chain fatty acids (SCFAs). Following drug treatment, the composition of intestinal microbes in NMOSD patients during the remission phase closely resembled that of the healthy control population. The FMT experiment provided evidence of the significant association between intestinal flora and the pathogenesis of NMOSD. Consequently, investigating gut microbiota and identifying novel microbial markers hold promise for the diagnosis and treatment of NMOSD patients.
Collapse
Affiliation(s)
- QinFang Xie
- Department of Neurology, Lanzhou University Second Hospital, Cuiyingmen 82, Chengguan District, Lanzhou, Gansu 730030, China
| | - Jing Sun
- Department of Neurology, Lanzhou University Second Hospital, Cuiyingmen 82, Chengguan District, Lanzhou, Gansu 730030, China
| | - MengJiao Sun
- Department of Neurology, Lanzhou University Second Hospital, Cuiyingmen 82, Chengguan District, Lanzhou, Gansu 730030, China
| | - Qi Wang
- Department of Neurology, Lanzhou University Second Hospital, Cuiyingmen 82, Chengguan District, Lanzhou, Gansu 730030, China.
| | - ManXia Wang
- Department of Neurology, Lanzhou University Second Hospital, Cuiyingmen 82, Chengguan District, Lanzhou, Gansu 730030, China.
| |
Collapse
|
8
|
Samukha V, Fantasma F, D’Urso G, Colarusso E, Schettino A, Marigliano N, Chini MG, Saviano G, De Felice V, Lauro G, Maione F, Bifulco G, Casapullo A, Iorizzi M. Chemical Profiling of Polar Lipids and the Polyphenolic Fraction of Commercial Italian Phaseolus Seeds by UHPLC-HRMS and Biological Evaluation. Biomolecules 2024; 14:1336. [PMID: 39456269 PMCID: PMC11505683 DOI: 10.3390/biom14101336] [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: 09/09/2024] [Revised: 10/13/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
The common bean (Phaseolus vulgaris L.) is one of the oldest food crops in the world. In this study, the ultra-high-performance liquid chromatography high-resolution mass spectrometry (UHPLC-MS/MS) technique was used to characterize the polar lipid composition and polyphenolic fraction of five bean varieties commonly consumed in Italy: Cannellino (PVCA), Controne (PVCO), Borlotti (PVBO), Stregoni (PVST), and Vellutina (PVVE). Lipid content represents a minor fraction of the whole metabolome in dry beans, and little is known about their polar lipids, which could be potentially bioactive components. Thirty-three compounds were detected through UHPLC-MS/MS, including oxylipins, phospholipids, N-acyl glycerolipids, and several fatty acids. The dichloromethane extracts were subjected to principal component analysis (PCA), with the results showing greater differentiation for the Borlotti variety. Moreover, 27 components belonging to different polyphenol classes, such as phenolic acids, flavonoids, catechins, anthocyanins and their glycosides, and some saponins, were identified in the hydroalcoholic seed extracts. In addition, the mineral content of the beans was determined. Considering the high number of compounds in the five apolar seed extracts, all samples were examined to determine their in vitro inhibitory activity against the enzyme cyclooxygenase-2 (COX-2), which is inducible in inflammatory cells and mediates inflammatory responses. Only PVCO showed the best inhibition of the COX-2 enzyme with an IC50 = 31.15 ± 2.16 µg/mL. In light of these results, the potential anti-inflammatory properties of PVCO were evaluated in the LPS-stimulated murine macrophage cell line J774A.1. Herein, we demonstrate, for the first time, that PVCO at 30 µg/mL can significantly reduce the release of TNF-α, with a less significant anti-inflammatory effect being observed in terms of IL-6 release.
Collapse
Affiliation(s)
- Vadym Samukha
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Isernia, Italy; (V.S.); (F.F.); (G.S.); (V.D.F.); (M.I.)
| | - Francesca Fantasma
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Isernia, Italy; (V.S.); (F.F.); (G.S.); (V.D.F.); (M.I.)
| | - Gilda D’Urso
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (G.D.); (E.C.); (G.L.); (G.B.)
| | - Ester Colarusso
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (G.D.); (E.C.); (G.L.); (G.B.)
| | - Anna Schettino
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy; (A.S.); (N.M.); (F.M.)
| | - Noemi Marigliano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy; (A.S.); (N.M.); (F.M.)
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Isernia, Italy; (V.S.); (F.F.); (G.S.); (V.D.F.); (M.I.)
| | - Gabriella Saviano
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Isernia, Italy; (V.S.); (F.F.); (G.S.); (V.D.F.); (M.I.)
| | - Vincenzo De Felice
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Isernia, Italy; (V.S.); (F.F.); (G.S.); (V.D.F.); (M.I.)
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (G.D.); (E.C.); (G.L.); (G.B.)
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy; (A.S.); (N.M.); (F.M.)
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (G.D.); (E.C.); (G.L.); (G.B.)
| | - Agostino Casapullo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (G.D.); (E.C.); (G.L.); (G.B.)
| | - Maria Iorizzi
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Isernia, Italy; (V.S.); (F.F.); (G.S.); (V.D.F.); (M.I.)
| |
Collapse
|
9
|
Shi C, Li Y, You Z, Tian Y, Zhu X, Xu H, Yang M, Zhang Y, Dong R, Quan H, Shang Y, Li X. Mangiferin Ameliorates CCl 4-Triggered Acute Liver Injury by Inhibiting Inflammatory Response and Oxidative Stress: Involving the Nrf2-ARE Pathway. J Inflamm Res 2024; 17:7081-7097. [PMID: 39380664 PMCID: PMC11460351 DOI: 10.2147/jir.s476288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024] Open
Abstract
Purpose Acute liver injury (ALI) is characterized by inflammation and oxidative stress (OS). Although mangiferin (MGF) has antioxidant and anti-inflammatory effects, its role in ALI remains unclear. Accordingly, we investigated the MGF molecular mechanism in carbon tetrachloride (CCl4)-induced ALI in vivo and in vitro. Materials and Methods The CCl4 was utilized to induce ALI in mice. In vivo, the therapeutic effects of MGF on CCl4-induced liver injury were evaluated through biochemical assays and histomorphological analysis. Additionally, immunohistochemistry, immunofluorescence, ELISA and Western blotting were further applied to explore the mechanism. In vitro, The CCK-8 assay and flow cytometry were employed to investigate the protective effects of MGF against CCl4-induced toxicity in HepG2 cells, while mitochondrial reactive oxygen species levels and Western blotting were used to explore the biological effects and molecular mechanisms. Results MGF treatment resulted in a reduction in serum levels of AST and ALT, diminished concentrations of TNF-α, IL-6, and IL-1β in liver tissue, and concurrently decreased cellular apoptosis. Furthermore, MGF pretreatment enhanced the activity of SOD and GSH while concurrently diminishing the MDA production. This study further demonstrated the upregulation of Nrf2, NQO1, and HO-1 protein expression levels, as well as the downregulation of p-p65 protein expression levels. In vitro investigations revealed that the mitigation of CCl4-induced inflammation and OS by MGF was mediated via the Nrf2- antioxidant response element (ARE) pathway, which was disrupted by ML385 in HepG2 cells. Conclusion CCl4 can induce liver injury, while treatment with MGF mitigates ALI by inhibiting oxidative stress, inflammation, and apoptosis. The protective mechanism of MGF is mediated by the Nrf2-ARE pathway activation.
Collapse
Affiliation(s)
- Caixing Shi
- School of Basic Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Yueyao Li
- College of Integrated Chinese and Western Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Zhidong You
- School of Nursing, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Yiran Tian
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Xiaoyu Zhu
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Hao Xu
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Menghan Yang
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Yutong Zhang
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Rui Dong
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Huirong Quan
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Yongyi Shang
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Xiaojin Li
- School of Basic Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| |
Collapse
|
10
|
Leão LPMDO, Neto AK, de Jesus Nicácio K, Lavorato SN, Leite FB, Teixeira KC, Murgu M, de Paula ACC, Soares MG, Chagas-Paula DA, Dias DF. Novel Synthesized Benzophenone Thiazole Hybrids Exhibited Ex Vivo and In Silico Anti-Inflammatory Activity. Chem Biol Drug Des 2024; 104:e14634. [PMID: 39424371 DOI: 10.1111/cbdd.14634] [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: 06/27/2024] [Revised: 08/24/2024] [Accepted: 09/09/2024] [Indexed: 10/21/2024]
Abstract
Novel benzophenone-thiazole hybrids with different substituents were synthesized and evaluated for anti-inflammatory activity using an ex vivo human whole-blood assay. All hybrids (3c and 5a-h) showed significant anti-inflammatory activity via prostaglandin E2 (PGE2) release inhibition. Moreover, 5c (82.8% of PGE2 inhibition), 5e (83.1% of PGE2 inhibition), and 5h (82.1% of PGE2 inhibition) were comparable to the reference drugs. Molecular docking revealed potential preferable binding to the active sites of cyclooxygenase 2 (COX-2) and microsomal prostaglandin E synthase-1 (mPGES-1) enzymes. This study provides the first evidence that benzophenone-thiazole hybrids may also dock in mPGES-1, a new attractive anti-inflammatory drug target, besides providing promising ex vivo anti-inflammatory activity. Thus, the novel hybrids are promising anti-inflammatory lead compounds and highlight the significance of optimal substituent selection in the design of potent PGE2 inhibitors.
Collapse
Affiliation(s)
| | | | | | - Stefânia Neiva Lavorato
- Center of Biological Sciences and Health, Federal University of Western Bahia, Barreiras, Bahia, Brazil
| | - Fernanda Brito Leite
- Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | | | | | - Ana Cláudia Chagas de Paula
- Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Marisi Gomes Soares
- Institute of Chemistry, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | | | | |
Collapse
|
11
|
Wang S, Liu W, Wei B, Wang A, Wang Y, Wang W, Gao J, Jin Y, Lu H, Ka Y, Yue Q. Traditional herbal medicine: Therapeutic potential in acute gouty arthritis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118182. [PMID: 38621464 DOI: 10.1016/j.jep.2024.118182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/27/2024] [Accepted: 04/08/2024] [Indexed: 04/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute gouty arthritis (AGA) is characterized by a rapid inflammatory reaction caused by the build-up of monosodium urate (MSU) crystals in the tissues surrounding the joints. This condition often associated with hyperuricemia (HUA), is distinguished by its symptoms of intense pain, active inflammation, and swelling of the joints. Traditional approaches in AGA management often fall short of desired outcomes in clinical settings. However, recent ethnopharmacological investigations have been focusing on the potential of Traditional Herbal Medicine (THM) in various forms, exploring their therapeutic impact and targets in AGA treatment. AIM OF THE REVIEW This review briefly summarizes the current potential pharmacological mechanisms of THMs - including active ingredients, extracts, and prescriptions -in the treatment of AGA, and discusses the relevant potential mechanisms and molecular targets in depth. The objective of this study is to offer extensive information and a reference point for the exploration of targeted AGA treatment using THMs. MATERIALS AND METHODS This review obtained scientific publications focused on in vitro and in vivo studies of anti-AGA THMs conducted between 2013 and 2023. The literature was collected from various journals and electronic databases, including PubMed, Elsevier, ScienceDirect, Web of Science, and Google Scholar. The retrieval and analysis of relevant articles were guided by keywords such as "acute gouty arthritis and Chinese herbal medicine," "acute gouty arthritis herbal prescription," "acute gouty arthritis and immune cells," "acute gouty arthritis and inflammation," "acute gouty arthritis and NOD-like receptor thermoprotein domain associated protein 3 (NLRP3)," "acute gouty arthritis and miRNA," and "acute gouty arthritis and oxidative stress." RESULTS We found that AGA has a large number of therapeutic targets, highlighting the effectiveness the potential of THMs in AGA treatment through in vitro and in vivo studies. THMs and their active ingredients can mitigate AGA symptoms through a variety of therapeutic targets, such as influencing macrophage polarization, neutrophils, T cells, natural killer (NK) cells, and addressing factors like inflammation, NLRP3 inflammasome, signaling pathways, oxidative stress, and miRNA multi-target interactions. The anti-AGA properties of THMs, including their active components and prescriptions, were systematically summarized and categorized based on their respective therapeutic targets. CONCLUSION phenolic, flavonoid, terpenoid and alkaloid compounds in THMs are considered the key ingredients to improve AGA. THMs and their active ingredients achieve enhanced efficacy through interactions with multiple targets, of which NLRP3 is a main therapeutic target. Nonetheless, given the intricate composition of traditional Chinese medicine (TCM), additional research is required to unravel the underlying mechanisms and molecular targets through which THMs alleviate AGA.
Collapse
Affiliation(s)
- Siwei Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Wei Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China.
| | - Bowen Wei
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Aihua Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Yiwen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Wen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Jingyue Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Yue Jin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Hang Lu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Yuxiu Ka
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Qingyun Yue
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| |
Collapse
|
12
|
Ma Z, Zeng P, Feng H, Ni L. Network pharmacology and molecular docking to explore the treatment potential and molecular mechanism of Si-Miao decoction against gouty arthritis. Medicine (Baltimore) 2024; 103:e38221. [PMID: 39259129 PMCID: PMC11142817 DOI: 10.1097/md.0000000000038221] [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: 08/17/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 09/12/2024] Open
Abstract
Gouty arthritis (GA) is a common metabolic rheumatological disease. Si-Miao decoction has therapeutic effects on GA. In our study, we investigated the mechanism of Si-Miao decoction against GA using network pharmacology and molecular docking analytical methods. The Traditional Chinese Medicine Systems Pharmacology Database was used as the basis for screening the main targets and agents of the Si-Miao decoction, and the Genecards, OMIM, and Drugbank databases were used to screen GA-related targets. They were analyzed using Venn with the drug targets to obtain the intersection targets. We used Cytoscape 3.9.1 to draw the "Drugs-Compounds-Targets" network and the String database for creative protein-protein interaction networks of target genes and filtered core targets. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes were used to analyze the core targets. Molecular docking was performed using AutoDockTools to predict the binding capacity between nuclear targets and active components in the Si-Miao decoction. A total of 50 chemically active components containing 53 common targets of Si-Miao decoction anti-GA and 53 potential drug target proteins were identified. Core targets, namely, TNF, STAT3, SRC, PPARG, TLR4, PTGS2, MMP9, RELA, TGFB1, and SIRT1, were obtained through PPI network analysis. GO and KEGG analyses showed that the mechanism of anti-GA in Si-Miao decoction may proceed by regulating biological processes such as inflammatory factor levels, cell proliferation, apoptosis, and lipid and glucose metabolism, and modulating the NOD-like receptor signaling pathway, IL-17 signaling pathway, TNF signaling pathway, NF-kappa B signaling pathway, and Toll-like receptor signaling pathway. We further screened the core targets, including PTGS2, MMP9, and PPAGR, as receptor proteins based on their degree value and molecular docking with the main active compounds in Si-Miao decoction, and found that baicalein had high affinity. In conclusion, Si-Miao decoction, through anti-inflammatory, apoptosis-regulating, and anti-oxidative stress action mechanisms in the treatment of GA.
Collapse
Affiliation(s)
- Zebing Ma
- Orthopedics (Orthopedic Group), The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Hunan University of Chinese Medicine, Changsha, China
| | - Peng Zeng
- Orthopedics (Orthopedic Group), The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Haibo Feng
- Orthopedics (Orthopedic Group), The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Lili Ni
- Orthopedics (Orthopedic Group), The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
13
|
Saviano A, Schettino A, Iaccarino N, Mansour AA, Begum J, Marigliano N, Raucci F, Romano F, Riccardi G, Mitidieri E, d'Emmanuele di Villa Bianca R, Bello I, Panza E, Smimmo M, Vellecco V, Rimmer P, Cheesbrough J, Zhi Z, Iqbal TH, Pieretti S, D'Amore VM, Marinelli L, La Pietra V, Sorrentino R, Costa L, Caso F, Scarpa R, Cirino G, Randazzo A, Bucci M, McGettrick HM, Iqbal AJ, Maione F. A reverse translational approach reveals the protective roles of Mangifera indica in inflammatory bowel disease. J Autoimmun 2024; 144:103181. [PMID: 38522129 DOI: 10.1016/j.jaut.2024.103181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/05/2024] [Accepted: 02/10/2024] [Indexed: 03/26/2024]
Abstract
Inflammatory bowel diseases (IBDs) are chronic intestinal disorders often characterized by a dysregulation of T cells, specifically T helper (Th) 1, 17 and T regulatory (Treg) repertoire. Increasing evidence demonstrates that dietary polyphenols from Mangifera indica L. extract (MIE, commonly known as mango) mitigate intestinal inflammation and splenic Th17/Treg ratio. In this study, we aimed to dissect the immunomodulatory and anti-inflammatory properties of MIE using a reverse translational approach, by initially using blood from an adult IBD inception cohort and then investigating the mechanism of action in a preclinical model of T cell-driven colitis. Of clinical relevance, MIE modulates TNF-α and IL-17 levels in LPS spiked sera from IBD patients as an ex vivo model of intestinal barrier breakdown. Preclinically, therapeutic administration of MIE significantly reduced colitis severity, pathogenic T-cell intestinal infiltrate and intestinal pro-inflammatory mediators (IL-6, IL-17A, TNF-α, IL-2, IL-22). Moreover, MIE reversed colitis-induced gut permeability and restored tight junction functionality and intestinal metabolites. Mechanistic insights revealed MIE had direct effects on blood vascular endothelial cells, blocking TNF-α/IFN-γ-induced up-regulation of COX-2 and the DP2 receptors. Collectively, we demonstrate the therapeutic potential of MIE to reverse the immunological perturbance during the onset of colitis and dampen the systemic inflammatory response, paving the way for its clinical use as nutraceutical and/or functional food.
Collapse
Affiliation(s)
- Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Anna Schettino
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Nunzia Iaccarino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Adel Abo Mansour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Jenefa Begum
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Noemi Marigliano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Federica Raucci
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Francesca Romano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Gelsomina Riccardi
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | | | - Ivana Bello
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Elisabetta Panza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Martina Smimmo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Valentina Vellecco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Peter Rimmer
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK; Department of Gastroenterology, Queen Elizabeth Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - Jonathan Cheesbrough
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK; Department of Gastroenterology, Birmingham Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Zhaogong Zhi
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Tariq H Iqbal
- Department of Gastroenterology, Queen Elizabeth Hospital Birmingham NHS Foundation Trust, Birmingham, UK; Institute of Microbiology and Infection (IMI), College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2WB, UK
| | - Stefano Pieretti
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Vincenzo Maria D'Amore
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Luciana Marinelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Valeria La Pietra
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Raffaella Sorrentino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Luisa Costa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, via S. Pansini 5, 80131, Naples, Italy
| | - Francesco Caso
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, via S. Pansini 5, 80131, Naples, Italy
| | - Raffaele Scarpa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, via S. Pansini 5, 80131, Naples, Italy
| | - Giuseppe Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Antonio Randazzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Helen Michelle McGettrick
- Institute of Inflammation and Ageing (IIA), College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2WB, UK
| | - Asif Jilani Iqbal
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy; Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy.
| |
Collapse
|
14
|
Zi X, Su R, Su R, Wang H, Li B, Gao C, Li X, Wang C. Elevated serum IL-2 and Th17/Treg imbalance are associated with gout. Clin Exp Med 2024; 24:9. [PMID: 38240927 PMCID: PMC10799120 DOI: 10.1007/s10238-023-01253-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 12/18/2023] [Indexed: 01/22/2024]
Abstract
Gout is considered an auto-inflammatory disorder, and the immunological drivers have not been fully unraveled. This study compared the peripheral lymphocyte and CD4+T cell subsets, and cytokines in gout and healthy controls (HCs) to explore the contributions of T helper 17 (Th17) cells, T regulatory (Treg) cells and cytokines to the pathogenesis of gout. We enrolled 126 gout patients (53 early-onset gout with age of first presentation < 40 years, and 73 late-onset gout with age of first presentation ≥ 40 years) and 77 HCs. Percentage and absolute numbers of peripheral lymphocyte and CD4+T cell subpopulations in each group were detected by flow cytometry. The serum cytokine levels were determined by flow cytometric bead array. For circulating CD4+T cell subsets, Th17/Treg ratio was significantly higher in early-onset gout, late-onset gout and gout without tophus than HCs; Th17 cells were significantly elevated in early-onset gout and gout without tophus, while the percentage of Treg cells was significantly decreased in early-onset and late-onset gout. Additionally, gout patients had significantly higher cytokines levels (including IL-2, IL-4, IL-6, IL-10, IL-17, IFN-γ, and TNF-α) than HCs; IL-2 levels were positively correlated with Treg cells and negatively correlated with ESR. ROC analysis showed that disease duration, CRP and fibrinogen, had moderate predictive performances for tophus in gout (the AUCs were 0.753, 0.703 and 0.701, respectively). Our study suggests that early-onset and late-onset gout differ in Th17/Treg imbalance, which in early-onset gout is due to elevated Th17 cells and in late-onset gout is due to decreased Treg cells. And increased serum cytokine levels, especially IL-2, may play an essential role in that. Restoring Th17/Treg balance may be a crucial way to improve the prognosis of gout patients.
Collapse
Affiliation(s)
- Xiaoyu Zi
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Ronghui Su
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Rui Su
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Hui Wang
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Baochen Li
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital/Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaofeng Li
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Caihong Wang
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China.
| |
Collapse
|
15
|
Kumar NS, Reddy N, Kumar H, Vemireddy S. Immunomodulatory Plant Natural Products as Therapeutics against Inflammatory Skin Diseases. Curr Top Med Chem 2024; 24:1013-1034. [PMID: 38485678 DOI: 10.2174/0115680266277952240223120435] [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: 10/09/2023] [Revised: 12/30/2023] [Accepted: 01/11/2024] [Indexed: 07/16/2024]
Abstract
Frequently occurring inflammatory skin conditions such as psoriasis, dermatitis, acne, including skin cancer, wounds and other disorders arising out of premature skin aging, deteriorate skin health and adversely impact human life. Even though several synthetic compounds have evolved for treating these skin conditions, natural-product-based therapeutics are gaining popularity with growing evidence of their efficacy and safety for treating skin disorders. Many of these inflammatory skin diseases have underlying disturbances in our immune system and immunomodulatory natural products provide solutions for their effective treatment and aid in understanding the underlying mechanism of such inflammatory skin conditions. Based on this premise, the present review summarizes the possible application of plant-derived immunomodulatory compositions and single molecules for treating inflammatory skin conditions. In vitro, in vivo and mechanistic studies reported the application of selected plant-derived natural products for the treatment of inflammatory skin disorders including, cancer and infections. Several online databases including PubMed, Google Scholar, and Science Direct have been searched for gathering the information covered in this review. Empirical studies demonstrated that most of these natural compounds exhibited therapeutic properties through their immunomodulatory and anti-inflammatory potential supplemented often with anti-microbial, anti-neoplastic, and anti- oxidant activities. Overall, plant-based natural products discussed here are capable of modulating the immune system to minimize or completely suppress the pro-inflammatory markers, scavenge free radicals (ROS), prevent bacteria, fungal, and virus-derived skin infections and often regress skin cancer through the induction of apoptosis. The challenges and opportunities associated with the application of plant-based immunomodulators for skin applications and their safety considerations are also discussed here. The present study indicated that immunomodulatory plant natural products being biologically validated ligands against various biological targets manifested in inflammatory skin diseases, offer an effective, safe and affordable treatment for such disorders affecting skin health. However, further clinical evaluations are needed to substantiate these findings.
Collapse
Affiliation(s)
- Nikhila Sampath Kumar
- Department of Dermatology, Venereology and Leprosy, Kamineni Institute of Medical Sciences (KIMS), Narketpalli, Nalagonda District, Hyderabad, 500 007, Telangana, India
| | - Navaneetha Reddy
- Department of Dermatology, Venereology and Leprosy, Kamineni Institute of Medical Sciences (KIMS), Narketpalli, Nalagonda District, Hyderabad, 500 007, Telangana, India
| | - Halmuthur Kumar
- Vaccine Immunology Laboratory, Indian Institute of Chemical Technology, Hyderabad-500 007, India
| | - Sravanthi Vemireddy
- Vaccine Immunology Laboratory, Indian Institute of Chemical Technology, Hyderabad-500 007, India
| |
Collapse
|
16
|
Choudhary S, Khan S, Rustagi S, Rajpal VR, Khan NS, Kumar N, Thomas G, Pandey A, Hamurcu M, Gezgin S, Zargar SM, Khan MK. Immunomodulatory Effect of Phytoactive Compounds on Human Health: A Narrative Review Integrated with Bioinformatics Approach. Curr Top Med Chem 2024; 24:1075-1100. [PMID: 38551050 DOI: 10.2174/0115680266274272240321065039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 02/08/2024] [Accepted: 02/16/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Immunomodulation is the modification of immune responses to control disease progression. While the synthetic immunomodulators have proven efficacy, they are coupled with toxicity and other adverse effects, and hence, the efforts were to identify natural phytochemicals with immunomodulatory potential. OBJECTIVE To understand the immunomodulatory properties of various phytochemicals and investigate them in Echinacea species extracts using an in silico approach. METHODOLOGY Several scientific database repositories were searched using different keywords: "Phytochemicals," "Alkaloids," "Polyphenols," "Flavonoids," "Lectins," "Glycosides," "Tannins," "Terpenoids," "Sterols," "Immunomodulators," and "Human Immune System" without any language restriction. Additionally, the study specifically investigated the immunomodulatory properties of Echinacea species extracts using gene expression analysis of GSE12259 from NCBI-GEO through the Bioconductor package GEOquery and limma. RESULTS A total of 182 studies were comprehensively analyzed to understand immunomodulatory phytochemicals. The in silico analysis highlighted key biological processes (positive regulation of cytokine production, response to tumor necrosis factor) and molecular functions (cytokine receptor binding, receptor-ligand activity, and cytokine activity) among Echinacea species extracts contributing to immune responses. Further, it also indicated the association of various metabolic pathways, i.e., pathways in cancer, cytokine-cytokine receptor interaction, NF-kappa B, PI3K-Akt, TNF, MAPK, and NOD-like receptor signaling pathways, with immune responses. The study revealed various hub targets, including CCL20, CCL4, GCH1, SLC7A11, SOD2, EPB41L3, TNFAIP6, GCLM, EGR1, and FOS. CONCLUSION The present study presents a cumulative picture of phytochemicals with therapeutic benefits. Additionally, the study also reported a few novel genes and pathways in Echinacea extracts by re-analyzing GSE 12259 indicating its anti-inflammatory, anti-viral, and immunomodulatory properties.
Collapse
Affiliation(s)
| | - Sheeba Khan
- Department of Food Technology, Sam Higginbottom University of Agriculture Technology and Sciences, Prayagraj, 21107, India
| | - Shivani Rustagi
- Department of Food Processing and Technology, Gautam Buddha University, Greater Noida, 201312, India
| | - Vijay Rani Rajpal
- Department of Botany, Hansraj College, University of Delhi, Delhi, 110007, India
| | - Noor Saba Khan
- ICMR-National Institute of Pathology, New Delhi, 110091, India
| | - Neeraj Kumar
- ICMR-National Institute of Pathology, New Delhi, 110091, India
| | - George Thomas
- Department of Molecular and Cellular Engineering, Sam Higginbottom University of Agriculture Technology and Sciences, Prayagraj, 21107, India
| | - Anamika Pandey
- Department of Soil Science and Plant Nutrition, Faculty of Agriculture, Selcuk University, Konya, 42079, Turkey
| | - Mehmet Hamurcu
- Department of Soil Science and Plant Nutrition, Faculty of Agriculture, Selcuk University, Konya, 42079, Turkey
| | - Sait Gezgin
- Department of Soil Science and Plant Nutrition, Faculty of Agriculture, Selcuk University, Konya, 42079, Turkey
| | - Sajad Majeed Zargar
- Proteomics Laboratory, Division of Plant Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190025, India
| | - Mohd Kamran Khan
- Department of Soil Science and Plant Nutrition, Faculty of Agriculture, Selcuk University, Konya, 42079, Turkey
| |
Collapse
|
17
|
Saviano A, Manosour AA, Raucci F, Merlino F, Marigliano N, Schettino A, Wahid M, Begum J, Filer A, Manning JE, Casillo GM, Piccolo M, Ferraro MG, Marzano S, Russomanno P, Bellavita R, Irace C, Amato J, Alfaifi M, Rimmer P, Iqbal T, Pieretti S, Vellecco V, Caso F, Costa L, Giacomelli R, Scarpa R, Cirino G, Bucci M, McGettrick HM, Grieco P, Iqbal AJ, Maione F. New biologic (Ab-IPL-IL-17) for IL-17-mediated diseases: identification of the bioactive sequence (nIL-17) for IL-17A/F function. Ann Rheum Dis 2023; 82:1415-1428. [PMID: 37580108 PMCID: PMC10579190 DOI: 10.1136/ard-2023-224479] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/07/2023] [Indexed: 08/16/2023]
Abstract
OBJECTIVES Interleukin (IL) 17s cytokines are key drivers of inflammation that are functionally dysregulated in several human immune-mediated inflammatory diseases (IMIDs), such as rheumatoid arthritis (RA), psoriasis and inflammatory bowel disease (IBD). Targeting these cytokines has some therapeutic benefits, but issues associated with low therapeutic efficacy and immunogenicity for subgroups of patients or IMIDs reduce their clinical use. Therefore, there is an urgent need to improve the coverage and efficacy of antibodies targeting IL-17A and/or IL-17F and IL-17A/F heterodimer. METHODS AND RESULTS Here, we initially identified a bioactive 20 amino acid IL-17A/F-derived peptide (nIL-17) that mimics the pro-inflammatory actions of the full-length proteins. Subsequently, we generated a novel anti-IL-17 neutralising monoclonal antibody (Ab-IPL-IL-17) capable of effectively reversing the pro-inflammatory, pro-migratory actions of both nIL-17 and IL-17A/F. Importantly, we demonstrated that Ab-IPL-IL-17 has less off-target effects than the current gold-standard biologic, secukinumab. Finally, we compared the therapeutic efficacy of Ab-IPL-IL-17 with reference anti-IL-17 antibodies in preclinical murine models and samples from patients with RA and IBD. We found that Ab-IPL-IL-17 could effectively reduce clinical signs of arthritis and neutralise elevated IL-17 levels in IBD patient serum. CONCLUSIONS Collectively, our preclinical and in vitro clinical evidence indicates high efficacy and therapeutic potency of Ab-IPL-IL-17, supporting the rationale for large-scale clinical evaluation of Ab-IPL-IL-17 in patients with IMIDs.
Collapse
Affiliation(s)
- Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Adel Abo Manosour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Federica Raucci
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Francesco Merlino
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Noemi Marigliano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Anna Schettino
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Mussarat Wahid
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Jenefa Begum
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Andrew Filer
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Julia E Manning
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | | | - Marialuisa Piccolo
- BioChemLab, Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Maria Grazia Ferraro
- BioChemLab, Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Simona Marzano
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | | | - Rosa Bellavita
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Carlo Irace
- BioChemLab, Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Jussara Amato
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Mohammed Alfaifi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Peter Rimmer
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Gastroenterology, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Tariq Iqbal
- Department of Gastroenterology, Queen Elizabeth Hospital Birmingham, Birmingham, UK
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Stefano Pieretti
- Department of Drug Research and Evaluation, Istituto Superiore di Sanità, Roma, Italy
| | | | - Francesco Caso
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Luisa Costa
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Roberto Giacomelli
- Fondazione Policlinico Universitario, and Research Unit of Immuno-Rheumatology, Department of Medicine and Surgery, Campus Bio-Medico University, Via Alvaro del Portillo, 200, 00128 Roma, Italy, and Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy, Roma, Italy
| | - Raffaele Scarpa
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Giuseppe Cirino
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | | | - Helen M McGettrick
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Paolo Grieco
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Asif Jilani Iqbal
- Department of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| |
Collapse
|
18
|
Liu W, Peng J, Wu Y, Ye Z, Zong Z, Wu R, Li H. Immune and inflammatory mechanisms and therapeutic targets of gout: An update. Int Immunopharmacol 2023; 121:110466. [PMID: 37311355 DOI: 10.1016/j.intimp.2023.110466] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/15/2023]
Abstract
Gout is an autoimmune disease characterized by acute or chronic inflammation and damage to bone joints induced due to the precipitation of monosodium urate (MSU) crystals. In recent years, with the continuous development of animal models and ongoing clinical investigations, more immune cells and inflammatory factors have been found to play roles in gouty inflammation. The inflammatory network involved in gout has been discovered, providing a new perspective from which to develop targeted therapy for gouty inflammation. Studies have shown that neutrophil macrophages and T lymphocytes play important roles in the pathogenesis and resolution of gout, and some inflammatory cytokines, such as those in the interleukin-1 (IL-1) family, have been shown to play anti-inflammatory or proinflammatory roles in gouty inflammation, but the mechanisms underlying their roles are unclear. In this review, we explore the roles of inflammatory cytokines, inflammasomes and immune cells in the course of gout development and the research status of therapeutic drugs used for inflammation to provide insights into future targeted therapy for gouty inflammation and the direction of gout pathogenesis research.
Collapse
Affiliation(s)
- Wenji Liu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, 330006 Nanchang, China; The Second Clinical Medical College of Nanchang University, 330006 Nanchang, China
| | - Jie Peng
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, 330006 Nanchang, China; The Second Clinical Medical College of Nanchang University, 330006 Nanchang, China
| | - Yixin Wu
- Queen Mary College of Nanchang University, 330006 Nanchang, China
| | - Zuxiang Ye
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, 330006 Nanchang, China; The Second Clinical Medical College of Nanchang University, 330006 Nanchang, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, 1 MinDe Road, 330006 Nanchang, China
| | - Rui Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, 330006 Nanchang, China.
| | - Hui Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, 330006 Nanchang, China.
| |
Collapse
|
19
|
Wang J, Xian M, Cao H, Wu L, Zhou L, Ma Y, Fan L, Lin L, Li G, Huang Q, Huang SK, Xiao X. Prophylactic and therapeutic potential of magnolol-loaded PLGA-PEG nanoparticles in a chronic murine model of allergic asthma. Front Bioeng Biotechnol 2023; 11:1182080. [PMID: 37214308 PMCID: PMC10192565 DOI: 10.3389/fbioe.2023.1182080] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
Magnolol is a chemically defined and active polyphenol extracted from magnolia plants possessing anti-allergic activity, but its low solubility and rapid metabolism dramatically hinder its clinical application. To improve the therapeutic effects, magnolol-encapsulated polymeric poly (DL-lactide-co-glycolide)-poly (ethylene glycol) (PLGA-PEG) nanoparticles were constructed and characterized. The prophylactic and therapeutic efficacy in a chronic murine model of OVA-induced asthma and the mechanisms were investigated. The results showed that administration of magnolol-loaded PLGA-PEG nanoparticles significantly reduced airway hyperresponsiveness, lung tissue eosinophil infiltration, and levels of IL-4, IL-13, TGF-β1, IL-17A, and allergen-specific IgE and IgG1 in OVA-exposed mice compared to their empty nanoparticles-treated mouse counterparts. Magnolol-loaded PLGA-PEG nanoparticles also significantly prevented mouse chronic allergic airway mucus overproduction and collagen deposition. Moreover, magnolol-encapsulated PLGA-PEG nanoparticles showed better therapeutic effects on suppressing allergen-induced airway hyperactivity, airway eosinophilic inflammation, airway collagen deposition, and airway mucus hypersecretion, as compared with magnolol-encapsulated poly (lactic-co-glycolic acid) (PLGA) nanoparticles or magnolol alone. These data demonstrate the protective effect of magnolol-loaded PLGA-PEG nanoparticles against the development of allergic phenotypes, implicating its potential usefulness for the asthma treatment.
Collapse
Affiliation(s)
- Junyi Wang
- Shenzhen Key Laboratory of Allergy and Immunology, Guangdong Provincial Standardization Allergen Engineering Research Center, State Key Laboratory of Respiratory Disease Shenzhen University Division, Institute of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Mo Xian
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hui Cao
- Shenzhen Key Laboratory of Allergy and Immunology, Guangdong Provincial Standardization Allergen Engineering Research Center, State Key Laboratory of Respiratory Disease Shenzhen University Division, Institute of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pulmonary and Critical Care Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Lei Wu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Libo Zhou
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yihe Ma
- Shenzhen Key Laboratory of Allergy and Immunology, Guangdong Provincial Standardization Allergen Engineering Research Center, State Key Laboratory of Respiratory Disease Shenzhen University Division, Institute of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pulmonary and Critical Care Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Long Fan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Lin Lin
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Guoping Li
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Qinmiao Huang
- Department of Pulmonary and Critical Care Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Shau-Ku Huang
- Shenzhen Key Laboratory of Allergy and Immunology, Guangdong Provincial Standardization Allergen Engineering Research Center, State Key Laboratory of Respiratory Disease Shenzhen University Division, Institute of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pulmonary and Critical Care Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiaojun Xiao
- Shenzhen Key Laboratory of Allergy and Immunology, Guangdong Provincial Standardization Allergen Engineering Research Center, State Key Laboratory of Respiratory Disease Shenzhen University Division, Institute of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pulmonary and Critical Care Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
20
|
Vellecco V, Saviano A, Raucci F, Casillo GM, Mansour AA, Panza E, Mitidieri E, Femminella GD, Ferrara N, Cirino G, Sorrentino R, Iqbal AJ, d'Emmanuele di Villa Bianca R, Bucci M, Maione F. Interleukin-17 (IL-17) triggers systemic inflammation, peripheral vascular dysfunction, and related prothrombotic state in a mouse model of Alzheimer's disease. Pharmacol Res 2023; 187:106595. [PMID: 36470548 DOI: 10.1016/j.phrs.2022.106595] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/08/2022]
Abstract
Alzheimer's disease (AD) is one of the most prevalent forms of neurodegenerative disorders. Previously, we have shown that in vivo administration of an IL-17 neutralizing antibody (IL-17Ab) rescues amyloid-β-induced neuro-inflammation and memory impairment, demonstrating the pivotal role of IL-17 in AD-derived cognitive deficit. Recently, AD has been recognized as a more intriguing pathology affecting vascular networks and platelet function. However, not much is known about peripheral vascular inflammation and how pro-inflammatory circulating cells/mediators could affect peripheral vessels' function. This study aimed to evaluate whether IL-17Ab treatment could also impact peripheral AD features, such as systemic inflammation, peripheral vascular dysfunction, and related pro-thrombotic state in a non-genetic mouse model of AD. Mice were injected intracerebroventricularly with Aβ1-42 peptide (3 μg/3 μl). To evaluate the systemic/peripheral protective profile of IL-17Ab, we used an intranasal administration of IL-17Ab (1 μg/10 μl) at 5, 12, and 19 days after Aβ1-42 injection. Circulating Th17/Treg cells and related cyto-chemokines, haematological parameters, vascular/endothelial reactivity, platelets and coagulation function in mice were evaluated. IL-17Ab treatment ameliorates the systemic/peripheral inflammation, immunological perturbance, vascular/endothelial impairment and pro-thrombotic state, suggesting a key role for this cytokine in fostering inflammatory processes that characterize the multifaced aspects of AD.
Collapse
Affiliation(s)
- Valentina Vellecco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Federica Raucci
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Gian Marco Casillo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Adel Abo Mansour
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Elisabetta Panza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Grazia Daniela Femminella
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy.
| | - Nicola Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; Istituti Clinici Scientifici ICS-Maugeri, Telese Terme, BN, Italy.
| | - Giuseppe Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnologies, School of Medicine, University of Naples, Federico II, Via Pansini, 5, 80131 Naples, Italy.
| | - Asif Jilani Iqbal
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy; Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | | | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| |
Collapse
|