1
|
Chen D, Yang J, Ren L, Zheng Z, Jin Z, Wen J, He J, Ding R, Wang J, Lin R, Song Q. Pyroptosis was suppressed by 20-hydroxyecdysone through Lin28b-mediated let-7d in vascular endothelial cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6083-6097. [PMID: 39652175 DOI: 10.1007/s00210-024-03591-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/31/2024] [Indexed: 04/11/2025]
Abstract
20-hydroxyecdysone (20E), a natural polyhydroxylated steroid, has exhibited anti-inflammatory effects across various diseases. This study investigates the potential connection between 20E's anti-inflammatory properties and the RNA-binding protein Lin28b, which is notably upregulated in TNF-α-stimulated endothelial cells. Herein, we discovered that 20E can selectively downregulate Lin28b expression without affecting its paralog Lin28a. Notably, 20E treatment could significantly attenuate pyroptosis, an inflammatory form of programmed cell death, as evidenced by reduced IL-1β and LDH release, and fewer propidium iodide (PI)-positive cells. Subsequent protein analysis revealed that 20E inhibited the enhanced expressions of key pyroptosis-associated proteins, GSDMD, GSDMD-N, and GSDME-N. Besides, this suppression of Lin28b and pyroptosis may be partially mediated through TNFR1. Additionally, 20E upregulated let-7 miRNA, particularly let-7d, a critical downstream target of Lin28b. To elucidate the role of Lin28b in 20E-mediated pyroptosis attenuation, we performed Lin28b overexpression and silencing experiments. Overexpressing Lin28b negated 20E's inhibition of LDH release and PI-related fluorescence, exacerbating GSDMD and GSDME cleavage. Conversely, Lin28b knockdown augmented the suppressive effect of 20E on pyroptosis, which was reversed by a let-7d inhibitor. Co-transfection with let-7d mimic and Lin28b plasmid demonstrated let-7d's role in mitigating pyroptosis aggravated by Lin28b overexpression. Overall, this study demonstrates that 20E may mitigate GSDMD and GSDME-mediated pyroptosis in HUVECs through the Lin28b/let-7d-dependent signaling pathway. These results highlight the potential of 20E as a promising inhibitor of pyroptosis, offering new insights into its anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Danli Chen
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P.R. China
| | - Jianjun Yang
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P.R. China
| | - Lingxuan Ren
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P.R. China
| | - Zihan Zheng
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P.R. China
| | - Zhen Jin
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P.R. China
| | - Jiazheng Wen
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P.R. China
| | - Jianyu He
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P.R. China
| | - Rongcheng Ding
- Xinjiang Rongcheng Hake Pharmaceutical Co.LTD, Altay region, 836500, Xinjiang, P.R. China
| | - Jianjiang Wang
- Xinjiang Rongcheng Hake Pharmaceutical Co.LTD, Altay region, 836500, Xinjiang, P.R. China
| | - Rong Lin
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P.R. China.
| | - Qiang Song
- Department of Structural Heart Disease, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China.
| |
Collapse
|
2
|
Nassr N, Rharbaoui F, Weitz D, Gassenhuber J, Rehberg M, Kohlmann M, Schumacher F, Lahmar A, Kovar A, Perrin L, Wagner FD, Wiekowski M, Nguyen MA. First-in-Human Single and Multiple Ascending Dose Studies of Balinatunfib, a Small Molecule Inhibitor of TNFR1 Signaling in Healthy Participants. Clin Pharmacol Ther 2025. [PMID: 40159671 DOI: 10.1002/cpt.3655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025]
Abstract
Oral small molecule inhibitors of tumor necrosis factor alpha (TNFα) are emerging as attractive therapeutic agents for the treatment of various autoimmune diseases. Balinatunfib (SAR441566), a novel oral inhibitor of tumor necrosis factor receptor 1 (TNFR1) signaling, changes the configuration of the soluble TNFα (sTNFα) trimer and prevents its heterotrimerization with TNFR1 but not TNFR2, thereby blocking TNFR1 signaling. Herein, we report the results from a first-in-human (FIH) study that evaluated the safety, pharmacokinetics (PK), and pharmacodynamics (PD) following single ascending doses (SAD) and multiple ascending doses (MAD) of balinatunfib in healthy male participants. Single (5-600 mg) and multiple (100-600 mg total daily dose for up to 14 days) oral doses of balinatunfib were well-tolerated in all participants. Consistent PK data were obtained across the studies, with a median tmax of 2.5-5 hours, a mean terminal half-life of 22-30 hours, and a time to steady state of 5-6 days. A supra-proportional exposure increase was observed in both SAD and MAD studies, which was less pronounced at doses ≥ 180 mg. Food had no relevant effects on the PK characteristics of balinatunfib. As the main PD read-out, complete TNFα occupancy was shown at all tested time points after the treatment started. Balinatunfib, as the first clinically tested oral TNFR1 signal inhibitor, demonstrated a good safety profile along with favorable PK/PD characteristics that allowed both once and twice daily dosing, confirming a successful preclinical-to-clinical translation and guiding dose selection for further clinical efficacy studies.
Collapse
|
3
|
Ma W, Tang S, Yao P, Zhou T, Niu Q, Liu P, Tang S, Chen Y, Gan L, Cao Y. Advances in acute respiratory distress syndrome: focusing on heterogeneity, pathophysiology, and therapeutic strategies. Signal Transduct Target Ther 2025; 10:75. [PMID: 40050633 PMCID: PMC11885678 DOI: 10.1038/s41392-025-02127-9] [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: 01/17/2024] [Revised: 12/27/2024] [Accepted: 12/27/2024] [Indexed: 03/09/2025] Open
Abstract
In recent years, the incidence of acute respiratory distress syndrome (ARDS) has been gradually increasing. Despite advances in supportive care, ARDS remains a significant cause of morbidity and mortality in critically ill patients. ARDS is characterized by acute hypoxaemic respiratory failure with diffuse pulmonary inflammation and bilateral edema due to excessive alveolocapillary permeability in patients with non-cardiogenic pulmonary diseases. Over the past seven decades, our understanding of the pathology and clinical characteristics of ARDS has evolved significantly, yet it remains an area of active research and discovery. ARDS is highly heterogeneous, including diverse pathological causes, clinical presentations, and treatment responses, presenting a significant challenge for clinicians and researchers. In this review, we comprehensively discuss the latest advancements in ARDS research, focusing on its heterogeneity, pathophysiological mechanisms, and emerging therapeutic approaches, such as cellular therapy, immunotherapy, and targeted therapy. Moreover, we also examine the pathological characteristics of COVID-19-related ARDS and discuss the corresponding therapeutic approaches. In the face of challenges posed by ARDS heterogeneity, recent advancements offer hope for improved patient outcomes. Further research is essential to translate these findings into effective clinical interventions and personalized treatment approaches for ARDS, ultimately leading to better outcomes for patients suffering from ARDS.
Collapse
Affiliation(s)
- Wen Ma
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
- Institute for Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, China
| | - Songling Tang
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Yao
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Tingyuan Zhou
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
- Institute for Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, China
| | - Qingsheng Niu
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Liu
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Shiyuan Tang
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Chen
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Gan
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China.
| | - Yu Cao
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China.
- Institute for Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, China.
| |
Collapse
|
4
|
Li Y, Ye R, Dai H, Lin J, Cheng Y, Zhou Y, Lu Y. Exploring TNFR1: from discovery to targeted therapy development. J Transl Med 2025; 23:71. [PMID: 39815286 PMCID: PMC11734553 DOI: 10.1186/s12967-025-06122-0] [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: 11/13/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
This review seeks to elucidate the therapeutic potential of tumor necrosis factor receptor 1 (TNFR1) and enhance our comprehension of its role in disease mechanisms. As a critical cell-surface receptor, TNFR1 regulates key signaling pathways, such as nuclear factor kappa-B (NF-κB) and mitogen-activated protein kinase (MAPK), which are associated with pro-inflammatory responses and cell death. The intricate regulatory mechanisms of TNFR1 signaling and its involvement in various diseases, including inflammatory disorders, infectious diseases, cancer, and metabolic syndromes, have attracted increasing scholarly attention. Given the potential risks associated with targeting tumor necrosis factor-alpha (TNF-α), selective inhibition of the TNFR1 signaling pathway has been proposed as a promising strategy to reduce side effects and enhance therapeutic efficacy. This review emphasizes the emerging field of targeted therapies aimed at selectively modulating TNFR1 activity, identifying promising therapeutic strategies that exploit TNFR1 as a drug target through an evaluation of current clinical trials and preclinical studies. In conclusion, this study contributes novel insights into the biological functions of TNFR1 and presents potential therapeutic strategies for clinical application, thereby having substantial scientific and clinical significance.
Collapse
Affiliation(s)
- Yingying Li
- School of Medicine, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, 201908, China
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Ruiwei Ye
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Haorui Dai
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Jiayi Lin
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yue Cheng
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yonghong Zhou
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China.
| | - Yiming Lu
- School of Medicine, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, 201908, China.
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
5
|
Liu G, Wang H, Zhang C, Li X, Mi Y, Chen Y, Xu L, Miao L, Long H, Liu Y. Tumor Necrosis Factor Receptor 1 Is Required for Human Umbilical Cord-Derived Mesenchymal Stem Cell-Mediated Rheumatoid Arthritis Therapy. Cell Transplant 2025; 34:9636897241301703. [PMID: 39831589 PMCID: PMC11748158 DOI: 10.1177/09636897241301703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 01/22/2025] Open
Abstract
Rheumatoid arthritis (RA) is a systemic, chronic inflammatory disease characterized by altered levels of inflammatory cytokines. One of the key cytokines involved in the pathogenesis of RA is tumor necrosis factor α (TNF-α), which plays a crucial role in the differentiation of T cells and B cells and serves as a primary trigger of inflammation and joint damage in RA. Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) have shown potential in alleviating the symptoms of RA. Previous in vitro studies indicate that TNF-α secreted by T cells can activate NF-κB in human MSCs, thereby triggering the immunoregulatory capacity of MSCs in a manner dependent on tumor necrosis factor receptor 1 (TNFR1). Inspired by these findings, we aimed to evaluate whether TNFR1 determine the therapeutic effects of hUC-MSCs on RA. First, we investigated whether TNFR1 is necessary for hUC-MSCs to inhibit TNF-α production of PBMCs, a source of elevated TNF-α in patients. Through coculture experiment, we confirmed that this inhibition was dependent on TNFR1. Subsequently, we administered hUC-MSCs or siTNFR1-MSCs to DBA/1J male mice with collagen-induced arthritis. The results indicated that hUC-MSCs significantly alleviated the pathological features of RA and suppressed the inflammatory cytokines IFN-γ, TNF-α, and IL-6 in peripheral blood, also in a manner dependent on TNFR1 either. Given the dramatic pathologic differences between hUC-MSCs and siTNFR1-MSCs treatments, we questioned whether production of growth factors and chemokines was significantly influenced by TNFR1. Consequently, we stimulated hUC-MSCs or siTNFR1-MSCs through IFN-γ, TNF-α, and IL-6, and profiled growth factors and chemokines in serum, which revealed significant changes of hepatocyte growth factor (HGF) and keratinocyte growth factor (KGF), as well as chemokines CXCL9, CXCL10, IL-8, and RANTES. In summary, our findings suggest that TNFR1 may determine whether hUC-MSCs will gain abilities of anti-inflammation and tissue regeneration.
Collapse
Affiliation(s)
- Guangyang Liu
- Stem Cell Biology and Regenerative Medicine Institution, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Herui Wang
- Stem Cell Biology and Regenerative Medicine Institution, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Chenliang Zhang
- Stem Cell Biology and Regenerative Medicine Institution, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Xin Li
- Stem Cell Biology and Regenerative Medicine Institution, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Yi Mi
- Stem Cell Biology and Regenerative Medicine Institution, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Yaoyao Chen
- Stem Cell Biology and Regenerative Medicine Institution, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Liqiang Xu
- Stem Cell Biology and Regenerative Medicine Institution, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Li Miao
- Stem Cell Biology and Regenerative Medicine Institution, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Haomiao Long
- Stem Cell Biology and Regenerative Medicine Institution, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Yongjun Liu
- Stem Cell Biology and Regenerative Medicine Institution, Yi-Chuang Institute of Bio-Industry, Beijing, China
| |
Collapse
|
6
|
Javaid N, Ahmad B, Patra MC, Choi S. Decoy peptides that inhibit TNF signaling by disrupting the TNF homotrimeric oligomer. FEBS J 2024; 291:4372-4391. [PMID: 39003565 DOI: 10.1111/febs.17220] [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: 03/26/2023] [Revised: 04/18/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
Tumor necrosis factor (TNF) is a pro-inflammatory cytokine and its functional homotrimeric form interacts with the TNF receptor (TNFR) to activate downstream apoptotic, necroptotic, and inflammatory signaling pathways. Excessive activation of these pathways leads to various inflammatory diseases, which makes TNF a promising therapeutic target. Here, 12-mer peptides were selected from the interface of TNF-TNFR based upon their relative binding energies and were named 'TNF-inhibiting decoys' (TIDs). These decoy peptides inhibited TNF-mediated secretion of cytokines and cell death, as well as activation of downstream signaling effectors. Effective TIDs inhibited TNF signaling by disrupting the formation of TNF's functional homotrimeric form. Among derivatives of TIDs, TID3c showed slightly better efficacy in cell-based assays by disrupting TNF trimer formation. Moreover, TID3c oligomerized TNF to a high molecular weight configuration. In silico modeling and simulations revealed that TID3c and its parent peptide, TID3, form a stable complex with TNF through hydrogen bonds and electrostatic interactions, which makes them the promising lead to develop peptide-based anti-TNF therapeutics.
Collapse
Affiliation(s)
- Nasir Javaid
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
- S&K Therapeutics, Suwon, Korea
| | - Bilal Ahmad
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
- S&K Therapeutics, Suwon, Korea
| | | | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
- S&K Therapeutics, Suwon, Korea
| |
Collapse
|
7
|
Pudjihartono N, Ho D, O’Sullivan JM. Integrative analysis reveals novel insights into juvenile idiopathic arthritis pathogenesis and shared molecular pathways with associated traits. Front Genet 2024; 15:1448363. [PMID: 39175752 PMCID: PMC11338781 DOI: 10.3389/fgene.2024.1448363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
Background Juvenile idiopathic arthritis (JIA) is an autoimmune joint disease that frequently co-occurs with other complex phenotypes, including cancers and other autoimmune diseases. Despite the identification of numerous risk variants through genome-wide association studies (GWAS), the affected genes, their connection to JIA pathogenesis, and their role in the development of associated traits remain unclear. This study aims to address these gaps by elucidating the gene-regulatory mechanisms underlying JIA pathogenesis and exploring its potential role in the emergence of associated traits. Methods A two-sample Mendelian Randomization (MR) analysis was conducted to identify blood-expressed genes causally linked to JIA. A curated protein interaction network was subsequently used to identify sets of single-nucleotide polymorphisms (i.e., spatial eQTL SNPs) that regulate the expression of JIA causal genes and their protein interaction partners. These SNPs were cross-referenced against the GWAS catalog to identify statistically enriched traits associated with JIA. Results The two-sample MR analysis identified 52 genes whose expression changes in the blood are putatively causal for JIA. These genes (e.g., HLA, LTA, LTB, IL6ST) participate in a range of immune-related pathways (e.g., antigen presentation, cytokine signalling) and demonstrate cell type-specific regulatory patterns across different immune cell types (e.g., PPP1R11 in CD4+ T cells). The spatial eQTLs that regulate JIA causal genes and their interaction partners were statistically enriched for GWAS SNPs linked with 95 other traits, including both known and novel JIA-associated traits. This integrative analysis identified genes whose dysregulation may explain the links between JIA and associated traits, such as autoimmune/inflammatory diseases (genes at 6p22.1 locus), Hodgkin lymphoma (genes at 6p21.3 [FKBPL, PBX2, AGER]), and chronic lymphocytic leukemia (BAK1). Conclusion Our approach provides a significant advance in understanding the genetic architecture of JIA and associated traits. The results suggest that the burden of associated traits may differ among JIA patients, influenced by their combined genetic risk across different clusters of traits. Future experimental validation of the identified connections could pave the way for refined patient stratification, the discovery of new biomarkers, and shared therapeutic targets.
Collapse
Affiliation(s)
- N. Pudjihartono
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - D. Ho
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - J. M. O’Sullivan
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, United Kingdom
- Australian Parkinsons Mission, Garvan Institute of Medical Research, Sydney, NSW, Australia
- A*STAR Singapore Institute for Clinical Sciences, Singapore, Singapore
| |
Collapse
|
8
|
Yilmaz O, Pinto JP, Torres T. New and emerging oral therapies for psoriasis. Drugs Context 2024; 13:2024-5-6. [PMID: 39131603 PMCID: PMC11313207 DOI: 10.7573/dic.2024-5-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/04/2024] [Indexed: 08/13/2024] Open
Abstract
Psoriasis is a chronic inflammatory skin disease affecting 2-3% of the global population. Traditional systemic treatments, such as methotrexate, cyclosporine, acitretin and fumaric acid esters, have limited efficacy and are associated with significant adverse effects, necessitating regular monitoring and posing risks of long-term toxicity. Recent advancements have introduced biologic drugs that offer improved efficacy and safety profiles. However, their high cost and the inconvenience of parenteral administration limit their accessibility. Consequently, there is a growing interest in developing new, targeted oral therapies. Small molecules, such as phosphodiesterase 4 inhibitors (e.g. apremilast) and TYK2 inhibitor (e.g. deucravacitinib), have shown promising results with favourable safety profiles. Additionally, other novel oral agents targeting specific pathways, including IL-17, IL-23, TNF, S1PR1 and A3AR, are under investigation. These treatments aim to combine the efficacy of biologics with the convenience and accessibility of oral administration, addressing the limitations of current therapies. This narrative review synthesizes the emerging oral therapeutic agents for psoriasis, focusing on their mechanisms of action, stages of development and clinical trial results.
Collapse
Affiliation(s)
- Orhan Yilmaz
- College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan,
Canada
| | - João Pedro Pinto
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto,
Portugal
| | - Tiago Torres
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto,
Portugal
- Department of Dermatology, Centro Hospitalar Universitário do Porto, Porto,
Portugal
| |
Collapse
|
9
|
Wu Z, Shi R, Yan S, Zhang S, Lu B, Huang Z, Ji L. Integrating network pharmacology, experimental validation and molecular docking to reveal the alleviation of Yinhuang granule on idiopathic pulmonary fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155368. [PMID: 38498951 DOI: 10.1016/j.phymed.2024.155368] [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: 08/14/2023] [Revised: 12/21/2023] [Accepted: 01/15/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic disease characterized by the abnormal proliferation of fibroblast and excessive deposition of extracellular matrix (ECM), accompanied by inflammation and ultimately respiratory failure. Yinhuang granule (YHG), with clinical properties of clearing heat, detoxifying and anti-inflammation, is commonly used to heal upper respiratory diseases in China for decades. PURPOSE To explore the improvement of YHG on bleomycin (BLM)-induced IPF in mice and its possible engaged mechanism. METHODS The mortality rate was recorded, lung function was determined and hematoxylin-eosin (H&E) staining was carried out to explore the alleviation of YHG on BLM-caused IPF in mice. Hydroxyproline, collagen I and collagen III contents were detected, and Sirius red and Masson staining were conducted to evaluate YHG's alleviation on lung fibrosis. The underlying mechanism was predicted by network pharmacology, and confirmed by Real-time polymerase chain reaction (RT-PCR), Western-blot (WB) and enzyme linked immunosorbent assay (ELISA). The binding affinity between related key proteins and active compounds in YHG was calculated by using molecular docking, and further validated by cellular thermal shift assay (CESTA). RESULTS YHG (400, 800 mg/kg) weakened lung damage and pulmonary fibrosis in mice induced by BLM. Network pharmacology and experimental validation displayed that inflammation and angiogenesis participated in the YHG-provided improvement on IPF, and key involved molecules included tumor necrosis factor-α (TNFα), vascular endothelial growth factor-A (VEGFA), interleukine-6 (IL-6), etc. The data of molecular docking presented that some main active compounds from YHG had a high binding affinity with TNFR1 or VEGFR2, and some of them were further validated by CESTA. CONCLUSION YHG effectively improved the BLM-induced IPF in mice via reducing inflammation and angiogenesis.
Collapse
Affiliation(s)
- Zeqi Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ruijia Shi
- School of Basic Medical Science of Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shihao Yan
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai,200123, China
| | - Shaobo Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bin Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhenlin Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
10
|
Bi R, Zhang D, Quan R, Lin X, Zhang W, Li C, Yuan M, Fang B, Wang D, Li Y. Edible bird's nest alleviates pneumonia caused by tobacco smoke inhalation through the TNFR1/NF-κB/NLRP3 pathway. Food Sci Nutr 2024; 12:4196-4210. [PMID: 38873472 PMCID: PMC11167147 DOI: 10.1002/fsn3.4080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/19/2024] [Accepted: 02/28/2024] [Indexed: 06/15/2024] Open
Abstract
Exposure to cigarette smoke directly damages the lungs and causes lung inflammation. The anti-inflammatory properties of edible bird's nest (EBN) have been reported. We aimed to determine the effect of EBN on pneumonia in a mouse model exposed to cigarette smoke. Fifty BALB/c mice were randomly divided into control, model, positive drug, low-dose EBN, and high-dose EBN groups (n = 10 each). Except for the control group, the mice in each group were exposed to four cigarettes once a day for 8 days. In addition, we validated the effects of EBN on A549 cells and investigated the mechanism by which EBN alleviates lung inflammation. Edible bird's nest (EBN) could alleviate the structural damage of lung tissue and the smoke-induced inflammatory response in mice. The best effect was observed at the high dose of EBN (0.019 g). The mice treated with EBN had a stronger ability than those in the model group to resist cigarette smoke stimulation, as indicated by a decrease in serum and lung inflammatory markers (interleukin 6 [IL-6], tumor necrosis factor-α [TNF-α], and interleukin 8 [IL-8]), an increase in serum interleukin 10 (IL-10) levels, and a decrease in the expression of inflammasome NOD-like receptor pyrin 3 (NLRP3). In addition, our cell experiments showed that EBN attenuated cigarette smoke-induced pulmonary inflammation mainly by inhibiting the tumor necrosis factor receptor 1 (TNFR1)/nuclear factor-kappa B (NF-κB)/NLRP3 pathway. These findings provide theoretical evidence for the positive nutritional qualities of EBN for the lung by demonstrating that it inhibits the TNFR1/NF-κB/NLRP3 signaling pathway, which prevents the development of cigarette smoke-induced pulmonary inflammation.
Collapse
Affiliation(s)
- Ran Bi
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and HealthChina Agricultural UniversityBeijingChina
| | - Dan Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and HealthChina Agricultural UniversityBeijingChina
| | - Rui Quan
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and HealthChina Agricultural UniversityBeijingChina
| | - Xiaoxian Lin
- Hebei Edible Bird's Nest Fresh Stew Technology Innovation CenterLangfangChina
| | - Wen Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and HealthChina Agricultural UniversityBeijingChina
| | - Chuangang Li
- Key Laboratory of Function Dairy, Co‐Constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional EngineeringChina Agricultural UniversityBeijingChina
| | - Man Yuan
- Hebei Edible Bird's Nest Fresh Stew Technology Innovation CenterLangfangChina
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and HealthChina Agricultural UniversityBeijingChina
| | - Dongliang Wang
- Hebei Edible Bird's Nest Fresh Stew Technology Innovation CenterLangfangChina
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and HealthChina Agricultural UniversityBeijingChina
| |
Collapse
|
11
|
Gao M, Song Y, Liu Y, Miao Y, Guo Y, Chai H. TNF-α/TNFR1 activated astrocytes exacerbate depression-like behavior in CUMS mice. Cell Death Discov 2024; 10:220. [PMID: 38710713 DOI: 10.1038/s41420-024-01987-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/08/2024] Open
Abstract
Neuroinflammation is considered to be a significant mechanism contributing to depression. Several studies have reported that A1 astrocytes were highly prevalent in human neuroinflammatory and neurodegenerative diseases. However, the precise mechanism by which A1 astrocytes contribute to depression remains unclear. Clinical studies have suggested a correlation between TNF-α, an activator of A1 astrocytes, and the severity of depression. Based on these findings, we hypothesized that TNF-α might worsen depression by activating A1 astrocytes. Our previous studies indicated that Rhodomyrtone (Rho) has the potential to improve depression-like behavior in mice. However, the exact mechanism for this effect has not been fully elucidated. Importantly, it was reported that Rho alleviated skin inflammation in a mouse model of psoriasis by inhibiting the expression of TNF-α. Based on this finding, we hypothesized that rhodomyrtone may exert antidepressant effects by modulating the TNF-α pathway. However, further research is required to investigate and validate these hypotheses, shedding light on the relationships between neuroinflammation, A1 astrocytes, TNF-α, and depression. By obtaining a deeper understanding of the underlying mechanisms, these findings could lead to the development of novel antidepressant strategies that target the TNF-α pathway in the context of neuroinflammation. In vivo, based on the established chronic unpredictable mild stress (CUMS) mouse depression model, we characterized the mechanism of TNF-α and Rho during depression by using several behavioral assays, adeno-associated virus(AAV) transfection, western blotting, immunofluorescence, and other experimental methods. In vitro, we characterized the effect of Rho on inflammation in TNF-α-treated primary astrocytes. TNFR1 expression was significantly increased in the hippocampus of depression-like mice, with increased astrocytes activation and neuronal apoptosis. These processes were further enhanced with increasing levels of TNF-α in the cerebrospinal fluid of mice. However, this process was attenuated by knockdown of TNFR1 and infliximab (Inf; a TNF-α antagonist). Injection of rhodomyrtone decreased the expressions of TNFR1 and TNF-α, resulting in significant improvements in mouse depression-like behaviors and reduction of astrocyte activation. TNF-α could be involved in the pathophysiological process of depression, through mediating astrocytes activation by binding to TNFR1. By blocking this pathway, Rho may be a novel antidepressant.
Collapse
Affiliation(s)
- Mengjiao Gao
- Neurosurgery Center, Department of Functional Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Yu Song
- Neurosurgery Center, Department of Functional Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Yaqi Liu
- Department of Cerebrovascular Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Yuqing Miao
- Neurosurgery Center, Department of Functional Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Yanwu Guo
- Neurosurgery Center, Department of Functional Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Huihui Chai
- Department of Cerebrovascular Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, National Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Shanghai Key Laboratory of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai, 200040, China.
| |
Collapse
|
12
|
Zeng J, Loi GWZ, Saipuljumri EN, Romero Durán MA, Silva-García O, Perez-Aguilar JM, Baizabal-Aguirre VM, Lo CH. Peptide-based allosteric inhibitor targets TNFR1 conformationally active region and disables receptor-ligand signaling complex. Proc Natl Acad Sci U S A 2024; 121:e2308132121. [PMID: 38551841 PMCID: PMC10998571 DOI: 10.1073/pnas.2308132121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 01/23/2024] [Indexed: 04/02/2024] Open
Abstract
Tumor necrosis factor (TNF) receptor 1 (TNFR1) plays a pivotal role in mediating TNF induced downstream signaling and regulating inflammatory response. Recent studies have suggested that TNFR1 activation involves conformational rearrangements of preligand assembled receptor dimers and targeting receptor conformational dynamics is a viable strategy to modulate TNFR1 signaling. Here, we used a combination of biophysical, biochemical, and cellular assays, as well as molecular dynamics simulation to show that an anti-inflammatory peptide (FKCRRWQWRMKK), which we termed FKC, inhibits TNFR1 activation allosterically by altering the conformational states of the receptor dimer without blocking receptor-ligand interaction or disrupting receptor dimerization. We also demonstrated the efficacy of FKC by showing that the peptide inhibits TNFR1 signaling in HEK293 cells and attenuates inflammation in mice with intraperitoneal TNF injection. Mechanistically, we found that FKC binds to TNFR1 cysteine-rich domains (CRD2/3) and perturbs the conformational dynamics required for receptor activation. Importantly, FKC increases the frequency in the opening of both CRD2/3 and CRD4 in the receptor dimer, as well as induces a conformational opening in the cytosolic regions of the receptor. This results in an inhibitory conformational state that impedes the recruitment of downstream signaling molecules. Together, these data provide evidence on the feasibility of targeting TNFR1 conformationally active region and open new avenues for receptor-specific inhibition of TNFR1 signaling.
Collapse
Affiliation(s)
- Jialiu Zeng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Gavin Wen Zhao Loi
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Eka Norfaishanty Saipuljumri
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- School of Applied Science, Republic Polytechnic, Singapore 738964, Singapore
| | - Marco Antonio Romero Durán
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58893, México
| | - Octavio Silva-García
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58893, México
| | - Jose Manuel Perez-Aguilar
- School of Chemical Sciences, Meritorious Autonomous University of Puebla, University City, Puebla 72570, México
| | - Víctor M Baizabal-Aguirre
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58893, México
| | - Chih Hung Lo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| |
Collapse
|
13
|
Drakos A, Torres T, Vender R. Emerging Oral Therapies for the Treatment of Psoriasis: A Review of Pipeline Agents. Pharmaceutics 2024; 16:111. [PMID: 38258121 PMCID: PMC10819460 DOI: 10.3390/pharmaceutics16010111] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/19/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The introduction of biologic agents for the treatment of psoriasis has revolutionized the current treatment landscape, targeting cytokines in the interleukin (IL)-23/IL-17 pathway and demonstrating strong efficacy and safety profiles in clinical trials. These agents however are costly, are associated with a risk of immunogenicity, and require administration by intravenous or subcutaneous injection, limiting their use among patients. Oral therapies, specifically small molecule and microbiome therapeutics, have the potential to be more convenient and cost-effective agents for patients and have been a focus of development in recent years, with few targeted oral medications available for the disease. In this manuscript, we review pipeline oral therapies for psoriasis identified through a search of ClinicalTrials.gov (30 June 2022-1 October 2023). Available preclinical and clinical trial data on each therapeutic agent are discussed. Small molecules under development include tumor necrosis factor inhibitors, IL-23 inhibitors, IL-17 inhibitors, phosphodiesterase-4 inhibitors, Janus kinase inhibitors, A3 adenosine receptor agonists, and sphingosine-1-phosphate receptor 1 agonists, several of which are entering phase III trials. Oral microbials have also demonstrated success in early phase studies. As new oral therapies emerge for the treatment of psoriasis, real-world data and comparative trials are needed to better inform their use among patients.
Collapse
Affiliation(s)
- Anastasia Drakos
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| | - Tiago Torres
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal;
- Department of Dermatology, Centro Hospitalar de Santo António, 4099-001 Porto, Portugal
| | - Ronald Vender
- Dermatrials Research Inc. & Venderm Consulting, Hamilton, ON L8N 1Y2, Canada
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
| |
Collapse
|
14
|
Sun L, Liu H, Ye Y, Lei Y, Islam R, Tan S, Tong R, Miao YB, Cai L. Smart nanoparticles for cancer therapy. Signal Transduct Target Ther 2023; 8:418. [PMID: 37919282 PMCID: PMC10622502 DOI: 10.1038/s41392-023-01642-x] [Citation(s) in RCA: 215] [Impact Index Per Article: 107.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/24/2023] [Accepted: 09/05/2023] [Indexed: 11/04/2023] Open
Abstract
Smart nanoparticles, which can respond to biological cues or be guided by them, are emerging as a promising drug delivery platform for precise cancer treatment. The field of oncology, nanotechnology, and biomedicine has witnessed rapid progress, leading to innovative developments in smart nanoparticles for safer and more effective cancer therapy. In this review, we will highlight recent advancements in smart nanoparticles, including polymeric nanoparticles, dendrimers, micelles, liposomes, protein nanoparticles, cell membrane nanoparticles, mesoporous silica nanoparticles, gold nanoparticles, iron oxide nanoparticles, quantum dots, carbon nanotubes, black phosphorus, MOF nanoparticles, and others. We will focus on their classification, structures, synthesis, and intelligent features. These smart nanoparticles possess the ability to respond to various external and internal stimuli, such as enzymes, pH, temperature, optics, and magnetism, making them intelligent systems. Additionally, this review will explore the latest studies on tumor targeting by functionalizing the surfaces of smart nanoparticles with tumor-specific ligands like antibodies, peptides, transferrin, and folic acid. We will also summarize different types of drug delivery options, including small molecules, peptides, proteins, nucleic acids, and even living cells, for their potential use in cancer therapy. While the potential of smart nanoparticles is promising, we will also acknowledge the challenges and clinical prospects associated with their use. Finally, we will propose a blueprint that involves the use of artificial intelligence-powered nanoparticles in cancer treatment applications. By harnessing the potential of smart nanoparticles, this review aims to usher in a new era of precise and personalized cancer therapy, providing patients with individualized treatment options.
Collapse
Affiliation(s)
- Leming Sun
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Hongmei Liu
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yanqi Ye
- Sorrento Therapeutics Inc., 4955 Directors Place, San Diego, CA, 92121, USA
| | - Yang Lei
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Rehmat Islam
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Sumin Tan
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yang-Bao Miao
- Department of Haematology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Lulu Cai
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
15
|
Almeida-Junior LA, de Carvalho MS, Almeida LKY, Silva-Sousa AC, Sousa-Neto MD, Silva RAB, Silva LAB, Paula-Silva FWG. TNF-α-TNFR1 Signaling Mediates Inflammation and Bone Resorption in Apical Periodontitis. J Endod 2023; 49:1319-1328.e2. [PMID: 37499863 DOI: 10.1016/j.joen.2023.07.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023]
Abstract
INTRODUCTION The aim of this study was to investigate the role of the proinflammatory axis TNF-α-TNFR1 in experimentally induced periapical inflammation and bone resorption in mice. METHODS After receiving Ethics Committee Approval (2019.1.139.58.0), experimental apical periodontitis was induced by means of inoculating oral microorganisms into the root canals of molars of mice. Genetically deficient tumor necrosis factor-α receptor-1 mice (TNFR1-/-; n = 50) response was compared with that of C57Bl6 wild-type mice (wild-type; n = 50) after 7, 14, 28, and 42 days. The analyses performed were micro-computed tomographic, histopathologic, histomicrobiological, and histometric evaluation, tartrate-resistant acid phosphatase staining, immunohistochemistry, and quantitative reverse transcriptase polymerase chain reaction. Data were analyzed by using one-way analysis of variance, followed by Tukey or Bonferroni tests (α = 5%). RESULTS TNFR1-/- mice exhibited lower recruitment of neutrophils at 14, 28, and 42 days (P < .05), which resulted in reduced area and volume of apical periodontitis at 42 days (P < .05). The number of osteoclasts was also lower in TNFR1-/- animals at 14 and 42 days (P < .01), along with reduced synthesis of CTSK, MMP-9, and COX-2. Expression of RANKL, but not OPG, was reduced at 14 and 42 days (P < .001). The highest RANKL expression over OPG (ratio > 1) was found in wild-type animals at 7 (P < .0001) and 42 days (P < .001). CONCLUSIONS Periapical inflammation and bone resorption were exacerbated in wild-type animals compared with TNFR1-/- mice, demonstrating that the TNF-α-TNFR1 signaling pathway mediated catabolic events in bone after root canal contamination.
Collapse
Affiliation(s)
| | - Marcio Santos de Carvalho
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Lana Kei Yamamoto Almeida
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Alice Corrêa Silva-Sousa
- Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Manoel Damião Sousa-Neto
- Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Raquel Assed Bezerra Silva
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Léa Assed Bezerra Silva
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
16
|
Wang Y, Ye R, Fan L, Zhao X, Li L, Zheng H, Qiu Y, He X, Lu Y. A TNF-α blocking peptide that reduces NF-κB and MAPK activity for attenuating inflammation. Bioorg Med Chem 2023; 92:117420. [PMID: 37573821 DOI: 10.1016/j.bmc.2023.117420] [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: 04/28/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023]
Abstract
Overexpression of tumor necrosis factor-α (TNF-α) is implicated in many inflammatory diseases, including septic shock, hepatitis, asthma, insulin resistance and autoimmune diseases, such as rheumatoid arthritis and Crohn's disease. The TNF-α signaling pathway is a valuable target, and anti-TNF-α drugs are successfully used to treat autoimmune and inflammatory diseases. Here, we study anti-inflammatory activity of an anti-TNF-α peptide (SN1-13, DEFHLELHLYQSW). In the cellular level assessment, SN1-13 inhibited TNF-α-induced cytotoxicity and blocks TNF-α-triggered signaling activities (IC50 = 15.40 μM). Moreover, the potential binding model between SN1-13 and TNF-α/TNFRs conducted through molecular docking revealed that SN1-13 could stunt TNF-α mediated signaling thought blocking TNF-α and its receptor TNFR1 and TNFR2. These results suggest that SN1-13 would be a potential lead peptide to treat TNF-α-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Yue Wang
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun 130013, China
| | - Ruiwei Ye
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Liming Fan
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai 201299, China
| | - Xin Zhao
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072,China
| | - Linxue Li
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Hao Zheng
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Yan Qiu
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai 201299, China.
| | - Xiuxia He
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun 130013, China.
| | - Yiming Lu
- School of Medicine, Shanghai University, Shanghai 200444, China; Department of Critical Care Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072,China.
| |
Collapse
|
17
|
Ci T, Xiong Y, Zhang J, Zang J, Feng N. Immunosuppressive dead cell as lung-targeting vehicle and cytokine absorption material for cytokine storm attenuation of pneumonia. Mater Today Bio 2023; 20:100684. [PMID: 37304577 PMCID: PMC10250915 DOI: 10.1016/j.mtbio.2023.100684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/13/2023] Open
Abstract
Effectively controlling cytokine storm is important to reduce the mortality of severe pneumonia. In this work a bio-functional dead cell was engineered by one-time quick shock of live immune cells in liquid nitrogen, and the obtained immunosuppressive dead cell could server as both lung-targeting vehicle and cytokine absorption material. After loading the anti-inflammatory drugs of dexamethasone (DEX) and baicalin (BAI), the drug-loaded dead cell (DEX&BAI/Dead cell) could first passively target to the lung after intravenous administration and quickly release the drugs under high shearing stress of pulmonary capillaries, realizing drug enrichment in the lung. Then, the immunosuppressive dead cell acted as the camouflage of normal immune cells with various cytokine receptors exposing on their surface, to "capture" the cytokines and further reduce the state of inflammation. With above formulation design, a synergic anti-inflammatory effect between drugs and carrier could be achieved. In a lipopolysaccharide-induced pneumonia mice model, this system could calm down the cytokine storm with high efficacy and elongate the survival of mice.
Collapse
Affiliation(s)
| | | | - Jinniu Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing Zang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Nianping Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
18
|
Wang H, Zhang L, Sun H, Xu S, Li K, Su X. Screening and application of inhibitory aptamers for DNA repair protein apurinic/apyrimidinic endonuclease 1. Int J Biol Macromol 2023:124918. [PMID: 37244341 DOI: 10.1016/j.ijbiomac.2023.124918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/12/2023] [Accepted: 05/13/2023] [Indexed: 05/29/2023]
Abstract
The base excision repair (BER) pathway is crucial for DNA repair, and apurinic/apyrimidinic endonuclease 1 (APE1) is a critical enzyme in this pathway. Overexpression of APE1 has been linked to multidrug resistance in various cancers, including lung cancer, colorectal cancer, and other malignant tumors. Therefore, reducing APE1 activity is desirable to improve cancer treatment. Inhibitory aptamers, which are versatile oligonucleotides for protein recognition and function restriction, are a promising tool for this purpose. In this study, we developed an inhibitory aptamer for APE1 using systematic evolution of ligands by exponential (SELEX) technology. We used carboxyl magnetic beads as the carrier and APE1 with a His-Tag as the positive screening target, while the His-Tag itself served as the negative screening target. The aptamer APT-D1 was selected based on its high binding affinity for APE1, with a dissociation constant (Kd) of 1.306 ± 0.1418 nM. Gel electrophoresis analysis showed that APT-D1 at a concentration of 1.6 μM could entirely inhibit APE1 with 21 nM. Our results suggest that these aptamers can be utilized for early cancer diagnosis and the treatment, and as an essential tool for studying the function of APE1.
Collapse
Affiliation(s)
- Huanhuan Wang
- College of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, China; Hebei Provincial Key Laboratory of NanoBiotechnology, Yanshan University, Qinhuangdao 066004, China; State Key Laboratory of Metastable Material Preparation Technology and Science, Yanshan University, Qinhuangdao 066004, China
| | - Linghao Zhang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Huaqing Sun
- College of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, China; Hebei Provincial Key Laboratory of NanoBiotechnology, Yanshan University, Qinhuangdao 066004, China; State Key Laboratory of Metastable Material Preparation Technology and Science, Yanshan University, Qinhuangdao 066004, China
| | - Shufeng Xu
- First Hospital of Qinhuangdao, Hebei Province 066000, China
| | - Kun Li
- College of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, China; Hebei Provincial Key Laboratory of NanoBiotechnology, Yanshan University, Qinhuangdao 066004, China; State Key Laboratory of Metastable Material Preparation Technology and Science, Yanshan University, Qinhuangdao 066004, China.
| | - Xin Su
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
19
|
Chu X, Du X, Yang L, Wang Z, Zhang Y, Wang X, Dai L, Zhang J, Liu J, Zhang N, Zhao Y, Gu H. Targeting Tumor Necrosis Factor Receptor 1 with Selected Aptamers for Anti-Inflammatory Activity. ACS APPLIED MATERIALS & INTERFACES 2023; 15:11599-11608. [PMID: 36812453 DOI: 10.1021/acsami.3c00131] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Tumor necrosis factor-α (TNFα) inhibitors are widely used in treating autoimmune diseases like rheumatoid arthritis (RA). These inhibitors can presumably alleviate RA symptoms by blocking TNFα-TNF receptor 1 (TNFR1)-mediated pro-inflammatory signaling pathways. However, the strategy also interrupts the survival and reproduction functions conducted by TNFα-TNFR2 interaction and causes side effects. Thus, it is urgently needed to develop inhibitors that can selectively block TNFα-TNFR1 but not TNFα-TNFR2. Here, nucleic acid-based aptamers against TNFR1 are explored as potential anti-RA candidates. Through the systematic evolution of ligands by exponential enrichment (SELEX), two types of TNFR1-targeting aptamers were obtained, and their KD values are approximately 100-300 nM. In silico analysis shows that the binding interface of aptamer-TNFR1 highly overlapped with natural TNFα-TNFR1 binding. On the cellular level, the aptamers can exert TNFα inhibitory activity by binding to TNFR1. The anti-inflammatory efficiencies of aptamers were assessed and further enhanced using divalent aptamer constructs. These findings provide a new strategy to block TNFR1 for potential anti-RA treatment precisely.
Collapse
Affiliation(s)
- Xiao Chu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, China
| | - Xinyu Du
- Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, China
| | - Longhua Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ziyi Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yi Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaonan Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Lijun Dai
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jiangnan Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jie Liu
- Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, China
| | - Nan Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yongxing Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Hongzhou Gu
- Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, China
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
20
|
Zeng S, Liang Y, Lai S, Bi S, Huang L, Li Y, Deng W, Xu P, Liu M, Xiong Z, Chen J, Tu Z, Chen D, Du L. TNFα/TNFR1 signal induces excessive senescence of decidua stromal cells in recurrent pregnancy loss. J Reprod Immunol 2023; 155:103776. [PMID: 36495656 DOI: 10.1016/j.jri.2022.103776] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 10/26/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022]
Abstract
Defects in decidual response are associated with adverse pregnancy outcomes which includes recurrent pregnancy loss (RPL). It is reported that cellular senescence happens during decidualization and pro-senescent decidual response in the luteal phase endometrium is related to RPL. However, the underlying mechanisms of how excessive decidual senescence takes place in RPL decidua cells remain largely unexplored. The senescent phenotype of RPL decidua and tumor necrosis factor receptor 1(TNFR1) expression were analyzed by using our previously published single-cell sequencing dataset of decidua cells from 6 RPL and 5 matched normal decidua, which were further verified by PCR and WB in decidual tissues. Effects of TNFα on the decidual stromal cells (DSCs) senescence and underlying molecular pathways were analyzed using the in vitro decidualization model of human endometrial stromal cells (HESCs). We showed that decidual stroma cells from RPL patients exhibited transcriptomic features of cellular senescence by analysis of single-cell datasets. The TNFα level and TNFR1 expression were increased in RPL decidua tissues. Furthermore, in vitro cell model demonstrated that increased TNFα induced excessive senescence during decidualization and TNFR1/p53/p16 pathway mediates TNFα-induced stromal senescence. In addition, we also found that the expression of IGFBP1 was regulated by TNFα-TNFR1 interaction during decidualization. Taken together, the present findings suggest that the increased secretion of TNFα induced stromal cell excessive senescence in RPL decidua, which is mediated via TNFR1, and thus provide a possible therapeutic target for the treatment of RPL.
Collapse
Affiliation(s)
- Shanshan Zeng
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Yingyu Liang
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Siying Lai
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Shilei Bi
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Lijun Huang
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Yulian Li
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Weinan Deng
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Pei Xu
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Mingxing Liu
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Zhongtang Xiong
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Jingsi Chen
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China; Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangzhou 510150, China; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China
| | - Zhaowei Tu
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China; Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangzhou 510150, China; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China
| | - Dunjin Chen
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China; Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangzhou 510150, China; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China.
| | - Lili Du
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China; Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangzhou 510150, China; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China.
| |
Collapse
|
21
|
Zhao X, Si S. Five genes as diagnostic biomarkers of dermatomyositis and their correlation with immune cell infiltration. Front Immunol 2023; 14:1053099. [PMID: 36742332 PMCID: PMC9889851 DOI: 10.3389/fimmu.2023.1053099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
Background Dermatomyositis (DM) is a rare autoimmune disease characterized by severe muscle dysfunction, and the immune response of the muscles plays an important role in the development of DM. Currently, the diagnosis of DM relies on symptoms, physical examination, and biopsy techniques. Therefore, we used machine learning algorithm to screen key genes, and constructed and verified a diagnostic model composed of 5 key genes. In terms of immunity, The relationship between 5 genes and immune cell infiltration in muscle samples was analyzed. These diagnostic and immune-cell-related genes may contribute to the diagnosis and treatment of DM. Methods GSE5370 and GSE128470 datasets were utilised from the Gene Expression Omnibus database as DM test sets. And we also used R software to merge two datasets and to analyze the results of differentially expressed genes (DEGs) and functional correlation analysis. Then, we could detect diagnostic genes adopting least absolute shrinkage and selection operator (LASSO) logistic regression and support vector machine recursive feature elimination (SVM-RFE) analyses. The validity of putative biomarkers was assessed using the GSE1551 dataset, and we confirmed the area under the receiver operating characteristic curve (AUC) values. Finally, CIBERSORT was used to evaluate immune cell infiltration in DM muscles and the correlations between disease-related biomarkers and immune cells. Results In this study, a total of 414 DEGs were screened. ISG15, TNFRSF1A, GUSBP11, SERPINB1 and PTMA were identified as potential DM diagnostic biomarkers(AUC > 0.85),and the expressions of 5 genes in DM group were higher than that in healthy group (p < 0.05). Immune cell infiltration analyses indicated that identified DM diagnostic biomarkers may be associated with M1 macrophages, activated NK cells, Tfh cells, resting NK cells and Treg cells. Conclusion The study identified that ISG15, TNFRSF1A, GUSBP11, SERPINB1 and PTMA as potential diagnostic biomarkers of DM and these genes were closely correlated with immune cell infiltration.This will contribute to future studies in diagnosis and treatment of DM.
Collapse
|
22
|
Barta BP, Onhausz B, AL Doghmi A, Szalai Z, Balázs J, Bagyánszki M, Bódi N. Gut region-specific TNFR expression: TNFR2 is more affected than TNFR1 in duodenal myenteric ganglia of diabetic rats. World J Diabetes 2023; 14:48-61. [PMID: 36684383 PMCID: PMC9850801 DOI: 10.4239/wjd.v14.i1.48] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/16/2022] [Accepted: 10/28/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Cytokines are essential in autoimmune inflammatory processes that accompany type 1 diabetes. Tumor necrosis factor alpha plays a key role among others in modulating enteric neuroinflammation, however, it has a dual role in cell degeneration or survival depending on different TNFRs. In general, TNFR1 is believed to trigger apoptosis, while TNFR2 promotes cell regeneration. The importance of the neuronal microenvironment has been recently highlighted in gut region-specific diabetic enteric neuropathy, however, the expression and alterations of different TNFRs in the gastrointestinal tract has not been reported.
AIM To investigate the TNFR1 and TNFR2 expression in myenteric ganglia and their environment in different intestinal segments of diabetic rats.
METHODS Ten weeks after the onset of hyperglycemia, gut segments were taken from the duodenum, ileum and colon of streptozotocin-induced (60 mg/body weight kg i.p.) diabetic (n = 17), insulin-treated diabetic (n = 15) and sex- and age-matched control (n = 15) rats. Myenteric plexus whole-mount preparations were prepared from different gut regions for TNFR1/HuCD or TNFR2/HuCD double-labeling fluorescent immunohistochemistry. TNFR1 and TNFR2 expression was evaluated by post-embedding immunogold electron microscopy on ultrathin sections of myenteric ganglia. TNFRs levels were measured by enzyme-linked immun-osorbent assay in muscle/myenteric plexus-containing (MUSCLE-MP) tissue homogenates from different gut segments and experimental conditions.
RESULTS A distinct region-dependent TNFRs expression was detected in controls. The density of TNFR1-labeling gold particles was lowest, while TNFR2 density was highest in duodenal ganglia and a decreased TNFRs expression from proximal to distal segments was observed in MUSCLE-MP homogenates. In diabetics, the TNFR2 density was only significantly altered in the duodenum with decrease in the ganglia (0.32 ± 0.02 vs 0.45 ± 0.04, P < 0.05), while no significant changes in TNFR1 density was observed. In diabetic MUSCLE-MP homogenates, both TNFRs levels significantly decreased in the duodenum (TNFR1: 4.06 ± 0.65 vs 20.32 ± 3.1, P < 0.001; TNFR2: 11.72 ± 0.39 vs 15.91 ± 1.04, P < 0.01), which markedly influenced the TNFR2/TNFR1 proportion in both the ganglia and their muscular environment. Insulin treatment had controversial effects on TNFR expression.
CONCLUSION Our findings show diabetes-related region-dependent changes in TNFR expression and suggest that TNFR2 is more affected than TNFR1 in myenteric ganglia in the duodenum of type 1 diabetic rats.
Collapse
Affiliation(s)
- Bence Pál Barta
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged 6726, Hungary
| | - Benita Onhausz
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged 6726, Hungary
| | - Afnan AL Doghmi
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged 6726, Hungary
| | - Zita Szalai
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged 6726, Hungary
| | - János Balázs
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged 6726, Hungary
| | - Mária Bagyánszki
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged 6726, Hungary
| | - Nikolett Bódi
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged 6726, Hungary
| |
Collapse
|
23
|
Wang X, Guo F, Zhang Y, Wang Z, Wang J, Luo R, Chu X, Zhao Y, Sun P. Dual-targeting inhibition of TNFR1 for alleviating rheumatoid arthritis by a novel composite nucleic acid nanodrug. Int J Pharm X 2023. [DOI: 10.1016/j.ijpx.2023.100162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
24
|
Liu X, Jiang Q, Lv J, Yang S, Huang Z, Duan R, Tao T, Li Z, Ju R, Zheng Y, Su W. Insights gained from single-cell analysis of immune cells in tofacitinib treatment of Vogt-Koyanagi-Harada disease. JCI Insight 2022; 7:162335. [PMID: 36301664 PMCID: PMC9746911 DOI: 10.1172/jci.insight.162335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/26/2022] [Indexed: 01/12/2023] Open
Abstract
Vogt-Koyanagi-Harada disease (VKH) is an important refractory uveitis mediated by pathological T cells (TCs). Tofacitinib (TOFA) is a JAK- targeted therapy for several autoimmune diseases. However, the specific pathogenesis and targeted therapeutics for VKH remain largely unknown. Based on single-cell RNA sequencing and mass cytometry, we present what we believe is the first multimodal, high-dimensional analysis to generate a comprehensive human immune atlas regarding subset composition, gene signatures, enriched pathways, and intercellular interactions of VKH patients undergoing TOFA therapy. Patients with VKH are characterized by TCs' polarization from naive to effector and memory subsets, together with accrued monocytes and upregulated cytokines and JAK/STAT signaling pathways. In vitro, TOFA reversed Th17/Treg imbalance and inhibited IL-2-induced STAT1/3 phosphorylation. TOFA alleviated VKH symptoms by restoring pathological TCs' polarization and functional marker expression and downregulating cytokine signaling and lymphocyte function. Remarkably, inflammation-related responses and intercellular interactions decreased after TOFA treatment, particularly in monocytes. Notably, we identified 2 inflammation- and JAK-associated monocyte subpopulations that were strongly implicated in VKH pathogenesis and mechanisms involved in TOFA treatment. Here, we provide a potentially novel JAK-targeted therapy for VKH and elaborate on the possible therapeutic mechanisms of TOFA, expanding our knowledge of VKH pathological patterns.
Collapse
|
25
|
Song Z, Ji L, Wu S, Fan Y, Zhang Q, Yang K, Fang S. Molecular mechanism of QH-BJ drug pair in the treatment of systemic lupus erythematosus based on network pharmacology and molecular docking. Medicine (Baltimore) 2022; 101:e32062. [PMID: 36482627 PMCID: PMC9726393 DOI: 10.1097/md.0000000000032062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To analyze the molecular mechanism of Qinghao-Biejia (QH-BJ) drug pair in the treatment of systemic lupus erythematosus (SLE) based on the method of network pharmacology and molecular docking technology. The components and related targets of QH-BJ drug pair, as well as SLE-related targets, were obtained. Intersection targets of QH-BJ drug pair and SLE were screened to construct the protein-protein interaction network, conduct gene ontology analysis and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, and establish the component-target-pathway network. The core active components and core targets of QH-BJ drug pair for the treatment of SLE were selected, and molecular docking was carried out between the ligand components and the receptor target proteins. The core active components of QH-BJ drug pair for the treatment of SLE are luteolin, quercetin, and kaempferol; the core targets are PTGS2, HSP90AA1, RELA, MAPK1, MAPK14, AKT1, JUN, TNF, TP53. The ligand components can spontaneously bind to the receptor target proteins. Besides, QH-BJ drug pair is likely to act on PI3K/Akt signal pathway, interleukin-17 signal pathway, and TNF signal pathway in the treatment of SLE. The study indicates that QH-BJ drug pair might play a role in the treatment of SLE through multi-components, multi-targets, and multi-pathways.
Collapse
Affiliation(s)
- Ziyu Song
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lina Ji
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shan Wu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yongsheng Fan
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qin Zhang
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Kepeng Yang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Sijia Fang
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- * Correspondence: Sijia Fang, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310007, China (e-mail: )
| |
Collapse
|
26
|
Zhang Y, Gao Z, Chao S, Lu W, Zhang P. Transdermal delivery of inflammatory factors regulated drugs for rheumatoid arthritis. Drug Deliv 2022; 29:1934-1950. [PMID: 35757855 PMCID: PMC9246099 DOI: 10.1080/10717544.2022.2089295] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Rheumatoid arthritis is a chronic autoimmune disease, with the features of recurrent chronic inflammation of synovial tissue, destruction of cartilage, and bone erosion, which further affects joints tissue, organs, and systems, and eventually leads to irreversible joint deformities and body dysfunction. Therapeutic drugs for rheumatoid arthritis mainly reduce inflammation through regulating inflammatory factors. Transdermal administration is gradually being applied to the treatment of rheumatoid arthritis, which can allow the drug to overcome the skin stratum corneum barrier, reduce gastrointestinal side effects, and avoid the first-pass effect, thus improving bioavailability and relieving inflammation. This paper reviewed the latest research progress of transdermal drug delivery in the treatment of rheumatoid arthritis, and discussed in detail the dosage forms such as gel (microemulsion gel, nanoemulsion gel, nanomicelle gel, sanaplastic nano-vesiclegel, ethosomal gel, transfersomal gel, nanoparticles gel), patch, drug microneedles, nanostructured lipid carrier, transfersomes, lyotropic liquid crystal, and drug loaded electrospinning nanofibers, which provide inspiration for the rich dosage forms of transdermal drug delivery systems for rheumatoid arthritis.
Collapse
Affiliation(s)
- Yanyan Zhang
- School of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Zhaoju Gao
- School of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Shushu Chao
- School of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Wenjuan Lu
- School of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Pingping Zhang
- School of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| |
Collapse
|
27
|
Evangelatos G, Bamias G, Kitas GD, Kollias G, Sfikakis PP. The second decade of anti-TNF-a therapy in clinical practice: new lessons and future directions in the COVID-19 era. Rheumatol Int 2022; 42:1493-1511. [PMID: 35503130 PMCID: PMC9063259 DOI: 10.1007/s00296-022-05136-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/12/2022] [Indexed: 11/22/2022]
Abstract
Since the late 1990s, tumor necrosis factor alpha (TNF-α) inhibitors (anti-TNFs) have revolutionized the therapy of immune-mediated inflammatory diseases (IMIDs) affecting the gut, joints, skin and eyes. Although the therapeutic armamentarium in IMIDs is being constantly expanded, anti-TNFs remain the cornerstone of their treatment. During the second decade of their application in clinical practice, a large body of additional knowledge has accumulated regarding various aspects of anti-TNF-α therapy, whereas new indications have been added. Recent experimental studies have shown that anti-TNFs exert their beneficial effects not only by restoring aberrant TNF-mediated immune mechanisms, but also by de-activating pathogenic fibroblast-like mesenchymal cells. Real-world data on millions of patients further confirmed the remarkable efficacy of anti-TNFs. It is now clear that anti-TNFs alter the physical course of inflammatory arthritis and inflammatory bowel disease, leading to inhibition of local and systemic bone loss and to a decline in the number of surgeries for disease-related complications, while anti-TNFs improve morbidity and mortality, acting beneficially also on cardiovascular comorbidities. On the other hand, no new safety signals emerged, whereas anti-TNF-α safety in pregnancy and amid the COVID-19 pandemic was confirmed. The use of biosimilars was associated with cost reductions making anti-TNFs more widely available. Moreover, the current implementation of the "treat-to-target" approach and treatment de-escalation strategies of IMIDs were based on anti-TNFs. An intensive search to discover biomarkers to optimize response to anti-TNF-α treatment is currently ongoing. Finally, selective targeting of TNF-α receptors, new forms of anti-TNFs and combinations with other agents, are being tested in clinical trials and will probably expand the spectrum of TNF-α inhibition as a therapeutic strategy for IMIDs.
Collapse
Affiliation(s)
- Gerasimos Evangelatos
- Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - Giorgos Bamias
- Gastrointestinal Unit, Third Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - George D Kitas
- Department of Rheumatology, Russells Hall Hospital, Dudley Group NHS Foundation Trust, Dudley, UK
- Arthritis Research UK Centre for Epidemiology, University of Manchester, Manchester, UK
| | - George Kollias
- Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Petros P Sfikakis
- Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
28
|
Luo Z, Chen S, Zhou J, Wang C, Li K, Liu J, Tang Y, Wang L. Application of aptamers in regenerative medicine. Front Bioeng Biotechnol 2022; 10:976960. [PMID: 36105606 PMCID: PMC9465253 DOI: 10.3389/fbioe.2022.976960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/08/2022] [Indexed: 12/03/2022] Open
Abstract
Regenerative medicine is a discipline that studies how to use biological and engineering principles and operation methods to repair and regenerate damaged tissues and organs. Until now, regenerative medicine has focused mainly on the in-depth study of the pathological mechanism of diseases, the further development and application of new drugs, and tissue engineering technology strategies. The emergence of aptamers has supplemented the development methods and types of new drugs and enriched the application elements of tissue engineering technology, injecting new vitality into regenerative medicine. The role and application status of aptamers screened in recent years in various tissue regeneration and repair are reviewed, and the prospects and challenges of aptamer technology are discussed, providing a basis for the design and application of aptamers in long-term transformation.
Collapse
Affiliation(s)
- Zhaohui Luo
- Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Department of Orthopedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Shimin Chen
- Guangxi Botanical Garden of Medicinal Plants, Nanning, China
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Jing Zhou
- Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Chong Wang
- School of Mechanical Engineering, Dongguan University of Technology, Dongguan, Guangdong, China
| | - Kai Li
- Academy of Orthopedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- *Correspondence: Kai Li, ; Jia Liu, ; Yujin Tang,
| | - Jia Liu
- Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Department of Orthopedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- *Correspondence: Kai Li, ; Jia Liu, ; Yujin Tang,
| | - Yujin Tang
- Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Department of Orthopedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- *Correspondence: Kai Li, ; Jia Liu, ; Yujin Tang,
| | - Liqiang Wang
- State Key Laboratory of Metal Matrix Composites, School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
29
|
From vaccines to nanovaccines: A promising strategy to revolutionize rheumatoid arthritis treatment. J Control Release 2022; 350:107-121. [PMID: 35977582 DOI: 10.1016/j.jconrel.2022.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
Rheumatoid arthritis (RA) is a joint-related autoimmune disease that is difficult to cure. Most therapeutics act to alleviate the symptoms but not correct the causes of RA. Novel strategies that specifically target the causes are highly needed for RA management. Currently, early interruption of RA is increasingly suggested but the corresponding therapeutics are not available. Vaccines that have shown great success to combat infection, cancer, degenerative diseases, autoimmune diseases, etc. are ideal candidates for a new generation of anti-RA therapeutics to correct the causes and prevent RA or interrupt RA in early phases. Anti-RA vaccines can be divided into two major categories. One is to induce neutralizing antibodies and the other is to induce antigen-specific immune tolerance. The vaccines are inherently linked to nanotechnology because they usually need a biomacromolecule or carrier to provoke sufficient immune responses. In the past decade, designed nanocarriers such as nanoparticles, liposomes, nanoemulsion, etc., have been applied to optimize the vaccines for autoimmune disease treatment. Nanotechnology endows vaccines with a higher biostability, tunable in vivo behavior, better targeting, co-delivery with stimulatory agents, regulatory effects on immune responses, etc. In this review, unmet medical needs for RA treatment and anti-RA vaccinology are first introduced. The development of anti-RA therapies from vaccines to nanovaccines are then reviewed and perspectives on how nanotechnology promotes vaccine development and advancement are finally provided. In addition, challenges for anti-RA vaccine development are summarized and advantages of nanovaccines are analyzed. In conclusion, nanovaccines will be a promising strategy to revolutionize the treatment of RA by correcting the causes in an early phase of RA.
Collapse
|
30
|
de Carvalho MS, de Almeida-Junior LA, Silva-Sousa AC, Damião Sousa-Neto M, Lucisano MP, Arnez MFM, da Silva LAB, Paula-Silva FWG. Absence of tumor necrosis factor receptor 1 inhibits osteoclast activity in apical dental resorption caused by endodontic infection in mice. J Endod 2022; 48:1400-1406. [PMID: 35964707 DOI: 10.1016/j.joen.2022.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 07/25/2022] [Accepted: 08/06/2022] [Indexed: 02/06/2023]
Abstract
INTRODUCTION To evaluate osteoclastogenesis and dental resorption resulting from endodontic infection in wild-type (WT) and tumor necrosis factor receptor 1 genetically deficient (TNFR1 KO) mice. METHODS After approval by the Ethics Committee on the use of Animals, 40 mice were distributed into two experimental groups based on periods: 14 days (n=10 WT mice; n=10 TNFR1 KO mice) and 42 days (n=10 WT mice; n=10 TNFR1 KO mice). After these periods, morphometrics analysis was done using bright field and fluorescence microscopy and tartrate-resistant acid phosphatase histoenzymology to identify osteoclasts. One-way analysis of variance followed by Tukey's post-hoc test was used for the statistical analysis (a=0.05). RESULTS WT mice in the 42-day period had a greater resorption in the apical region distal root of the first molar than TNFR1 KO mice (p<0.05). On the other hand, TNFR1 KO mice showed a smaller number of osteoclasts on the dental surface than WT mice (p<0.05). CONCLUSION WT mice had more extensive bone and apical dental resorptions and a larger number of osteoclasts on the tooth surface than TNFR1 KO mice.
Collapse
Affiliation(s)
- Marcio Santos de Carvalho
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Alice Corrêa Silva-Sousa
- Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Manoel Damião Sousa-Neto
- Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marília Pacífico Lucisano
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Maya Fernanda Manfrin Arnez
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Léa Assed Bezerra da Silva
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | |
Collapse
|
31
|
An M, Shi M, Su J, Wei Y, Luo R, Sun P, Zhao Y. Dual-Drug Loaded Separable Microneedles for Efficient Rheumatoid Arthritis Therapy. Pharmaceutics 2022; 14:pharmaceutics14071518. [PMID: 35890412 PMCID: PMC9324764 DOI: 10.3390/pharmaceutics14071518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/16/2022] [Accepted: 07/19/2022] [Indexed: 01/27/2023] Open
Abstract
Although the inhibitors of the interleukin-6 receptor (IL-6R) and tumor necrosis factor-α (TNF-α) have achieved a certain success in the clinical treatment of rheumatoid arthritis (RA), great effort should be made to overcome side effects and to improve patient compliance. The present research aimed to address these problems by the co-delivery of tocilizumab (TCZ)—an inhibitor of IL-6R—and an aptamer Apt1-67, which specifically inhibits TNF receptor 1 via separable microneedles (MN). MN were featured with a sustained release of TCZ from needle tips and a rapid release of Apt1-67 from needle bodies by using methacrylate groups grafted hyaluronic acid as the fillings of needle tips and polyvinyl alcohol/polyvinyl pyrrolidone as the fillings of needle bodies. Our results demonstrated that TCZ and Apt1-67 were distributed in MN as expected, and they could be released to the surroundings in the skin. In vivo studies revealed that combined medication via MN (TCZ/Apt1-67@MN) was superior to MN loaded with a single drug. Compared with subcutaneous injection, TCZ/Apt1-67@MN was of great advantage in inhibiting bone erosion and alleviating symptoms of CIA mice. This study not only provides a novel approach for combined medication with different release properties but also supplies a strategy for improving drug efficacy.
Collapse
Affiliation(s)
- Mengchen An
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China; (M.A.); (M.S.); (J.S.); (Y.W.); (R.L.)
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Mengxiao Shi
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China; (M.A.); (M.S.); (J.S.); (Y.W.); (R.L.)
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Jingjing Su
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China; (M.A.); (M.S.); (J.S.); (Y.W.); (R.L.)
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Yueru Wei
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China; (M.A.); (M.S.); (J.S.); (Y.W.); (R.L.)
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Rongrong Luo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China; (M.A.); (M.S.); (J.S.); (Y.W.); (R.L.)
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Pengchao Sun
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China; (M.A.); (M.S.); (J.S.); (Y.W.); (R.L.)
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
- Correspondence: (P.S.); (Y.Z.)
| | - Yongxing Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China; (M.A.); (M.S.); (J.S.); (Y.W.); (R.L.)
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, No. 100 Science Ave, Zhengzhou 450001, China
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
- Correspondence: (P.S.); (Y.Z.)
| |
Collapse
|
32
|
Immunosuppressant Therapies in COVID-19: Is the TNF Axis an Alternative? Pharmaceuticals (Basel) 2022; 15:ph15050616. [PMID: 35631442 PMCID: PMC9147078 DOI: 10.3390/ph15050616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 02/05/2023] Open
Abstract
The study of cytokine storm in COVID-19 has been having different edges in accordance with the knowledge of the disease. Various cytokines have been the focus, especially to define specific treatments; however, there are no conclusive results that fully support any of the options proposed for emergency treatment. One of the cytokines that requires a more exhaustive review is the tumor necrosis factor (TNF) and its receptors (TNFRs) as increased values of soluble formats for both TNFR1 and TNFR2 have been identified. TNF is a versatile cytokine with different impacts at the cellular level depending on the action form (transmembrane or soluble) and the receptor to which it is associated. In that sense, the triggered mechanisms can be diversified. Furthermore, there is the possibility of the joint action provided by synergism between one or more cytokines with TNF, where the detonation of combined cellular processes has been suggested. This review aims to discuss some roles of TNF and its receptors in the pro-inflammatory stage of COVID-19, understand its ways of action, and let to reposition this cytokine or some of its receptors as therapeutic targets.
Collapse
|
33
|
Shang H, Younas A, Zhang N. Recent advances on transdermal delivery systems for the treatment of arthritic injuries: From classical treatment to nanomedicines. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1778. [PMID: 35112483 DOI: 10.1002/wnan.1778] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 12/17/2022]
Abstract
Arthritic injuries happen frequently during a lifetime due to accidents, sports, aging, diseases, etc. Such injuries can be cartilage/bone injuries, tendon injuries, ligament injuries, inflammation, pain, and/or synovitis. Oral and injective administration of therapeutics are typically used but cause many side effects. Transdermal administration is an alternative route for safe and efficient delivery. Transdermal formulations of non-steroidal anti-inflammatory drugs have been available on market for years and show promising efficacy in pain relieving, inflammation alleviation, infection control, and so on. Innovative transdermal patches, gels/films, and microneedles have also been widely explored as formulations to deliver therapeutics to combat arthritic injuries. However, transdermal formulations that halt disease progression and promote damage repair are translated slowly from lab bench to clinical applications. One major reason is that the skin barrier and synovial capsule barrier limit the efficacy of transdermal delivery. Recently, many nanocarriers, such as nanoparticles, nanolipids, nanoemulsions, nanocrystals, exosomes, etc., have been incorporated into transdermal formulations to advance drug delivery. The combined transdermal formulations show promising safety and efficacy. Therefore, this review will focus on stating the current development of nanomedicine-based transdermal formulations for the treatment of arthritic injuries. The advances, limitations, and future perspectives in this field will also be provided to inspire future studies and accelerate clinical translational studies. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement Biology-Inspired Nanomaterials > Lipid-Based Structures.
Collapse
Affiliation(s)
- Hongtao Shang
- School of Sports Sciences (Main Campus), Zhengzhou University, Zhengzhou, Henan, China
| | - Ayesha Younas
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Nan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
34
|
Niessen NM, Fricker M, McDonald VM, Gibson PG. T2-low: what do we know?: Past, present, and future of biologic therapies in noneosinophilic asthma. Ann Allergy Asthma Immunol 2022; 129:150-159. [PMID: 35487388 DOI: 10.1016/j.anai.2022.04.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/30/2022] [Accepted: 04/19/2022] [Indexed: 02/07/2023]
Abstract
T2-low asthma is an often severe asthma subtype with limited treatment options and biologic therapeutics are lacking. Several monoclonal antibodies (mAbs) targeting non-T2 cytokines were previously reported to be ineffective in asthma. These trials often investigated heterogeneous asthma populations and negative outcomes could be related to unsuitable study cohorts. More tailored approaches in selecting participants based on specific biomarkers have been beneficial in treating severe T2-high asthma. Similarly, mAbs previously deemed ineffective bear the potential to be useful when administered to the correct target population. Here, we review individual clinical trials conducted between 2005 and 2021 and assess the suitability of the selected cohorts, whether study end points were met, and whether outcome measures were appropriate to investigate the effectiveness of the respective drug. We discuss potential target groups within the T2-low asthma population and suggest biomarkers that may predict a treatment response. Furthermore, we assess whether biomarker-guided approaches or subgroup analyses were associated with more positive study outcomes. The mAbs directed against alarmins intervene early in the inflammatory cascade and are the first mAbs found to have efficacy in T2-low asthma. Several randomized controlled trials performed predefined subgroup analyses that included T2-low asthma. Subgroup analyses were associated with positive outcomes and were able to reveal a stronger response in at least 1 subgroup. A better understanding of T2-low subgroups and specific biomarkers is necessary to identify the most responsive target population for a given mAb.
Collapse
Affiliation(s)
- Natalie M Niessen
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia; School of Medicine and Public Health, The University of Newcastle, Newcastle, NSW, Australia; Asthma and Breathing Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia.
| | - Michael Fricker
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia; School of Medicine and Public Health, The University of Newcastle, Newcastle, NSW, Australia; Asthma and Breathing Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Vanessa M McDonald
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia; School of Medicine and Public Health, The University of Newcastle, Newcastle, NSW, Australia; Asthma and Breathing Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia; School of Nursing and Midwifery, The University of Newcastle, Newcastle, NSW, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Peter G Gibson
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia; School of Medicine and Public Health, The University of Newcastle, Newcastle, NSW, Australia; Asthma and Breathing Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| |
Collapse
|
35
|
Ghorbaninezhad F, Leone P, Alemohammad H, Najafzadeh B, Nourbakhsh NS, Prete M, Malerba E, Saeedi H, Tabrizi NJ, Racanelli V, Baradaran B. Tumor necrosis factor‑α in systemic lupus erythematosus: Structure, function and therapeutic implications (Review). Int J Mol Med 2022; 49:43. [PMID: 35137914 DOI: 10.3892/ijmm.2022.5098] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/05/2022] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor‑α (TNF‑α) is a pleiotropic pro‑inflammatory cytokine that contributes to the pathophysiology of several autoimmune diseases, such as multiple sclerosis, inflammatory bowel disease, rheumatoid arthritis, psoriatic arthritis and systemic lupus erythematosus (SLE). The specific role of TNF‑α in autoimmunity is not yet fully understood however, partially, in a complex disease such as SLE. Through the engagement of the TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2), both the two variants, soluble and transmembrane TNF‑α, can exert multiple biological effects according to different settings. They can either function as immune regulators, impacting B‑, T‑ and dendritic cell activity, modulating the autoimmune response, or as pro‑inflammatory mediators, regulating the induction and maintenance of inflammatory processes in SLE. The present study reviews the dual role of TNF‑α, focusing on the different effects that TNF‑α may have on the pathogenesis of SLE. In addition, the efficacy and safety of anti‑TNF‑α therapies in preclinical and clinical trials SLE are discussed.
Collapse
Affiliation(s)
- Farid Ghorbaninezhad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan 5165665811, Iran
| | - Patrizia Leone
- Department of Biomedical Sciences and Human Oncology, 'Aldo Moro' University of Bari Medical School, I‑70124 Bari, Italy
| | - Hajar Alemohammad
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, East Azerbaijan 5166616471, Iran
| | - Basira Najafzadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, East Azerbaijan 5166616471, Iran
| | - Niloufar Sadat Nourbakhsh
- Department of Genetics, Faculty of Basic Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Fars 7319846451, Iran
| | - Marcella Prete
- Department of Biomedical Sciences and Human Oncology, 'Aldo Moro' University of Bari Medical School, I‑70124 Bari, Italy
| | - Eleonora Malerba
- Department of Biomedical Sciences and Human Oncology, 'Aldo Moro' University of Bari Medical School, I‑70124 Bari, Italy
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan 5165665811, Iran
| | - Neda Jalili Tabrizi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan 5165665811, Iran
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, 'Aldo Moro' University of Bari Medical School, I‑70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan 5165665811, Iran
| |
Collapse
|
36
|
Fighting fire with fire: the immune system might be key in our fight against Alzheimer's disease. Drug Discov Today 2022; 27:1261-1283. [PMID: 35032668 DOI: 10.1016/j.drudis.2022.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/25/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022]
Abstract
The ultimate cause of Alzheimer's disease (AD) is still unknown and no disease-modifying treatment exists. Emerging evidence supports the concept that the immune system has a key role in AD pathogenesis. This awareness leads to the idea that specific parts of the immune system must be engaged to ward off the disease. Immunotherapy has dramatically improved the management of several previously untreatable cancers and could hold similar promise as a novel therapy for treating AD. However, before potent immunotherapies can be rationally designed as treatment against AD, we need to fully understand the dynamic interplay between AD and the different parts of our immune system. Accordingly, here we review the most important aspects of both the innate and adaptive immune system in relation to AD pathology. Teaser: Emerging results support the concept that Alzheimer's disease is affected by the inability of the immune system to contain the pathology of the brain. Here, we discuss how we can engage our immune system to fight this devastating disease.
Collapse
|
37
|
Transmembrane TNF and Its Receptors TNFR1 and TNFR2 in Mycobacterial Infections. Int J Mol Sci 2021; 22:ijms22115461. [PMID: 34067256 PMCID: PMC8196896 DOI: 10.3390/ijms22115461] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 12/16/2022] Open
Abstract
Tumor necrosis factor (TNF) is one of the main cytokines regulating a pro-inflammatory environment. It has been related to several cell functions, for instance, phagocytosis, apoptosis, proliferation, mitochondrial dynamic. Moreover, during mycobacterial infections, TNF plays an essential role to maintain granuloma formation. Several effector mechanisms have been implicated according to the interactions of the two active forms, soluble TNF (solTNF) and transmembrane TNF (tmTNF), with their receptors TNFR1 and TNFR2. We review the impact of these interactions in the context of mycobacterial infections. TNF is tightly regulated by binding to receptors, however, during mycobacterial infections, upstream activation signalling pathways may be influenced by key regulatory factors either at the membrane or cytosol level. Detailing the structure and activation pathways used by TNF and its receptors, such as its interaction with solTNF/TNFRs versus tmTNF/TNFRs, may bring a better understanding of the molecular mechanisms involved in activation pathways which can be helpful for the development of new therapies aimed at being more efficient against mycobacterial infections.
Collapse
|
38
|
Cao J, Su J, An M, Yang Y, Zhang Y, Zuo J, Zhang N, Zhao Y. Novel DEK-Targeting Aptamer Delivered by a Hydrogel Microneedle Attenuates Collagen-Induced Arthritis. Mol Pharm 2020; 18:305-316. [PMID: 33253580 DOI: 10.1021/acs.molpharmaceut.0c00954] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
DEK protein is critical to the formation of neutrophil extracellular traps (NETs) in rheumatoid arthritis (RA). Blocking DEK using the aptamer DTA via articular injection has been shown to have robust anti-inflammatory efficacy in a previous study. However, DTA is prone to nuclease degradation and renal clearance in vivo. RA is a systemic disease that involves multiple joints, and local injection is impractical in clinical settings. In this study, DTA was modified with methoxy groups on all deoxyribose sugar units and inverted deoxythymidine on the 3' end (DTA4) to enhance its stability against nuclease. DTA4 is stable for 72 h in 90% mouse serum and maintains a high binding affinity to DEK. DTA4 effectively inhibits the formation of NETs and the migration of HUVECs in vitro. DTA4 was then modified with cholesterol on its 5' end to form DTA6. DTA6 dramatically reduces DEK expression in inflammatory RAW264.7 cells. A hydrogel microneedle (hMN) was then fabricated for the transdermal delivery of DTA6. The hMN maintains morphological integrity after absorbing the aptamer solution, effectively pierces the skin, and rapidly releases DTA6 into the dermis. The DTA6-loaded hMN significantly attenuates inflammation and protects joints from cartilage/bone erosion in collagen-induced arthritis (CIA) mice.
Collapse
Affiliation(s)
- Jian Cao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Jingjing Su
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Mengchen An
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Yang Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Yi Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Jing Zuo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Nan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, Zhengzhou 450001, Henan, P. R. China.,Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Henan Province, Zhengzhou 450001, Henan, P. R. China
| | - Yongxing Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, Zhengzhou 450001, Henan, P. R. China.,Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Henan Province, Zhengzhou 450001, Henan, P. R. China
| |
Collapse
|